Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
2.
bioRxiv ; 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37808640

ABSTRACT

Unchecked, chronic inflammation is a constitutive component of age-related diseases, including age-related macular degeneration (AMD). Here we identified interleukin-1 receptor-associated kinase (IRAK)-M as a key immunoregulator in retinal pigment epithelium (RPE) that declines with age. Rare genetic variants of IRAK-M increased the likelihood of AMD. IRAK-M expression in RPE declined with age or oxidative stress and was further reduced in AMD. IRAK-M-deficient mice exhibited increased incidence of outer retinal degeneration at earlier ages, which was further exacerbated by oxidative stressors. The absence of IRAK-M disrupted RPE cell homeostasis, including compromised mitochondrial function, cellular senescence, and aberrant cytokine production. IRAK-M overexpression protected RPE cells against oxidative or immune stressors. Subretinal delivery of AAV-expressing IRAK-M rescued light-induced outer retinal degeneration in wild-type mice and attenuated age-related spontaneous retinal degeneration in IRAK-M-deficient mice. Our data support that replenishment of IRAK-M expression may redress dysregulated pro-inflammatory processes in AMD, thereby treating degeneration.

4.
Nat Commun ; 13(1): 3243, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35688811

ABSTRACT

Cerebral organoids can be used to gain insights into cell type specific processes perturbed by genetic variants associated with neuropsychiatric disorders. However, robust and scalable phenotyping of organoids remains challenging. Here, we perform RNA sequencing on 71 samples comprising 1,420 cerebral organoids from 25 donors, and describe a framework (Orgo-Seq) to integrate bulk RNA and single-cell RNA sequence data. We apply Orgo-Seq to 16p11.2 deletions and 15q11-13 duplications, two loci associated with autism spectrum disorder, to identify immature neurons and intermediate progenitor cells as critical cell types for 16p11.2 deletions. We further applied Orgo-Seq to identify cell type-specific driver genes. Our work presents a quantitative phenotyping framework to integrate multi-transcriptomic datasets for the identification of cell types and cell type-specific co-expressed driver genes associated with neuropsychiatric disorders.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Intellectual Disability , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Chromosome Deletion , Chromosome Disorders , Chromosomes, Human, Pair 16 , Humans , Intellectual Disability/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics
6.
Transl Vis Sci Technol ; 10(4): 3, 2021 04 01.
Article in English | MEDLINE | ID: mdl-34003982

ABSTRACT

Translational Relevance: Subclinical or clinical inflammation often arises during ocular gene therapy with viral vectors. Understanding the biological bases and impacts on efficacy are important for clinical management and the improvement of future therapies.


Subject(s)
Genetic Therapy , Genetic Vectors , Blindness/genetics , Genetic Vectors/genetics , Humans , Inflammation/genetics , Vision, Ocular
7.
PLoS Pathog ; 17(3): e1009446, 2021 03.
Article in English | MEDLINE | ID: mdl-33770145

ABSTRACT

RIG-I and MDA5 are cytoplasmic RNA sensors that mediate cell intrinsic immunity against viral pathogens. While it has been well-established that RIG-I and MDA5 recognize RNA viruses, their interactive network with DNA viruses, including herpes simplex virus 1 (HSV-1), remains less clear. Using a combination of RNA-deep sequencing and genetic studies, we show that the γ134.5 gene product, a virus-encoded virulence factor, enables HSV growth by neutralization of RIG-I dependent restriction. When expressed in mammalian cells, HSV-1 γ134.5 targets RIG-I, which cripples cytosolic RNA sensing and subsequently suppresses antiviral gene expression. Rather than inhibition of RIG-I K63-linked ubiquitination, the γ134.5 protein precludes the assembly of RIG-I and cellular chaperone 14-3-3ε into an active complex for mitochondrial translocation. The γ134.5-mediated inhibition of RIG-I-14-3-3ε binding abrogates the access of RIG-I to mitochondrial antiviral-signaling protein (MAVS) and activation of interferon regulatory factor 3. As such, unlike wild type virus HSV-1, a recombinant HSV-1 in which γ134.5 is deleted elicits efficient cytokine induction and replicates poorly, while genetic ablation of RIG-I expression, but not of MDA5 expression, rescues viral growth. Collectively, these findings suggest that viral suppression of cytosolic RNA sensing is a key determinant in the evolutionary arms race of a large DNA virus and its host.


Subject(s)
DEAD Box Protein 58/metabolism , Herpes Simplex/metabolism , Herpesvirus 1, Human/pathogenicity , Receptors, Immunologic/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Animals , Chlorocebus aethiops , Fibroblasts/metabolism , HEK293 Cells , Herpesvirus 1, Human/metabolism , Humans , Mitochondria/metabolism , Protein Transport/physiology , Vero Cells
8.
Sci Transl Med ; 13(580)2021 02 10.
Article in English | MEDLINE | ID: mdl-33568518

ABSTRACT

Nucleic acids are used in many therapeutic modalities, including gene therapy, but their ability to trigger host immune responses in vivo can lead to decreased safety and efficacy. In the case of adeno-associated viral (AAV) vectors, studies have shown that the genome of the vector activates Toll-like receptor 9 (TLR9), a pattern recognition receptor that senses foreign DNA. Here, we engineered AAV vectors to be intrinsically less immunogenic by incorporating short DNA oligonucleotides that antagonize TLR9 activation directly into the vector genome. The engineered vectors elicited markedly reduced innate immune and T cell responses and enhanced gene expression in clinically relevant mouse and pig models across different tissues, including liver, muscle, and retina. Subretinal administration of higher-dose AAV in pigs resulted in photoreceptor pathology with microglia and T cell infiltration. These adverse findings were avoided in the contralateral eyes of the same animals that were injected with the engineered vectors. However, intravitreal injection of higher-dose AAV in macaques, a more immunogenic route of administration, showed that the engineered vector delayed but did not prevent clinical uveitis, suggesting that other immune factors in addition to TLR9 may contribute to intraocular inflammation in this model. Our results demonstrate that linking specific immunomodulatory noncoding sequences to much longer therapeutic nucleic acids can "cloak" the vector from inducing unwanted immune responses in multiple, but not all, models. This "coupled immunomodulation" strategy may widen the therapeutic window for AAV therapies as well as other DNA-based gene transfer methods.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Dependovirus/genetics , Gene Transfer Techniques , Genetic Therapy , Immunity, Innate , Mice , Swine
9.
Brain Sci ; 10(2)2020 Feb 22.
Article in English | MEDLINE | ID: mdl-32098339

ABSTRACT

Recently, adeno-associated virus (AAV)-mediated gene therapies have attracted clinical interest for treating neurodegenerative diseases including spinal muscular atrophy (SMA), Canavan disease (CD), Parkinson's disease (PD), and Friedreich's ataxia (FA). The influx of clinical findings led to the first approved gene therapy for neurodegenerative disorders in 2019 and highlighted new safety concerns for patients. Large doses of systemically administered AAV stimulate host immune responses, resulting in anti-capsid and anti-transgene immunity with implications for transgene expression, treatment longevity, and patient safety. Delivering lower doses directly to the central nervous system (CNS) is a promising alternative, resulting in higher transgene expression with decreased immune responses. However, neuroinflammatory responses after CNS-targeted delivery of AAV are a critical concern. Reported signs of AAV-associated neuroinflammation in preclinical studies include dorsal root ganglion (DRG) and spinal cord pathology with mononuclear cell infiltration. In this review, we discuss ways to manage neuroinflammation, including choice of AAV capsid serotypes, CNS-targeting routes of delivery, genetic modifications to the vector and/or transgene, and adding immunosuppressive strategies to clinical protocols. As additional gene therapies for neurodegenerative diseases enter clinics, tracking biomarkers of neuroinflammation will be important for understanding the impact immune reactions can have on treatment safety and efficacy.

10.
Cell Host Microbe ; 26(4): 493-503.e6, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31600501

ABSTRACT

14-3-3 protein family members facilitate the translocation of RIG-I-like receptors (RLRs) to organelles that mediate downstream RLR signaling, leading to interferon production. 14-3-3ϵ promotes the cytosolic-to-mitochondrial translocation of RIG-I, while 14-3-3η facilitates MDA5 translocation to mitochondria. We show that the NS3 protein of Zika virus (ZIKV) antagonizes antiviral gene induction by RIG-I and MDA5 by binding to and sequestering the scaffold proteins 14-3-3ϵ and 14-3-3η. 14-3-3-binding is mediated by a negatively charged RLDP motif in NS3 that is conserved in ZIKV strains of African and Asian lineages and is similar to the one found in dengue and West Nile viruses. ZIKV NS3 is sufficient to inhibit the RLR-14-3-3ϵ/η interaction and to suppress antiviral signaling. Mutational perturbation of 14-3-3ϵ/η binding in a recombinant ZIKV leads to enhanced innate immune responses and impaired growth kinetics. Our study provides molecular understanding of immune evasion functions of ZIKV, which may guide vaccine and anti-flaviviral therapy development.


Subject(s)
14-3-3 Proteins/metabolism , Immune Evasion/immunology , Peptide Hydrolases/metabolism , Viral Proteins/metabolism , Zika Virus Infection/immunology , Zika Virus/immunology , A549 Cells , Animals , Cell Line , Chlorocebus aethiops , DEAD Box Protein 58/antagonists & inhibitors , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/immunology , Interferon-Induced Helicase, IFIH1/antagonists & inhibitors , Interferon-beta/immunology , Mitochondria/metabolism , Peptide Hydrolases/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptors, Immunologic , Serine Endopeptidases , Vero Cells , Viral Proteins/genetics , Zika Virus/genetics
11.
JCI Insight ; 52019 05 21.
Article in English | MEDLINE | ID: mdl-31112525

ABSTRACT

Recombinant adeno-associated virus (rAAV)-mediated gene delivery can efficiently target muscle tissues to serve as "biofactories" for secreted proteins in prophylactic and therapeutic scenarios. Nevertheless, efficient rAAV-mediated gene delivery is often limited by host immune responses against the transgene product. The development of strategies to prevent anti-transgene immunity is therefore crucial. The employment of endogenous microRNA (miRNA)-mediated regulation to detarget transgene expression from antigen presenting cells (APCs) has shown promise for reducing immunogenicity. However, the mechanisms underlying miRNA-mediated modulation of anti-transgene immunity by APC detargeting are not fully understood. Using the highly immunogenic ovalbumin (OVA) protein as a proxy for foreign antigens, we show that rAAV vectors containing miR142 binding sites efficiently repress co-stimulatory signals in dendritic cells, significantly blunt the cytotoxic T cell response, allow for sustained transgene expression in skeletal myoblasts, and attenuate clearance of transduced muscle cells in mice. Furthermore, the blunting of humoral immunity against circulating OVA correlates with detargeting of OVA expression from APCs. This demonstrates that incorporating APC-specific miRNA binding sites into rAAV vectors provides an effective strategy for reducing transgene-specific immune response. This approach holds promise for clinical applications where the safe and efficient delivery of a prophylactic or therapeutic protein is desired.


Subject(s)
Dependovirus/genetics , Immunity, Cellular/immunology , Immunity, Humoral/immunology , MicroRNAs/genetics , MicroRNAs/metabolism , Ovalbumin/immunology , Animals , Antibody Formation , Antigen-Presenting Cells , CD8-Positive T-Lymphocytes , Cytokines/metabolism , Dendritic Cells/immunology , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Homeodomain Proteins , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscles/immunology , Muscles/pathology , T-Lymphocytes, Cytotoxic/metabolism
12.
Viruses ; 11(2)2019 01 25.
Article in English | MEDLINE | ID: mdl-30691064

ABSTRACT

Decades ago, Friedmann and Roblin postulated several barriers to gene therapy, including tissue targeting, delivery across the blood⁻brain barrier (BBB), and host immune responses. These issues remain pertinent till today. Since then, several advances have been made in elucidating structures of adeno-associated virus (AAV) serotypes, antibody epitopes, and ways to modify antibody-binding sites. AAVs capsid has also been engineered to re-direct tissue tropism, reduce ubiquitination, and promote passage across the BBB. Furthermore, the use of high(er) dose recombinant AAV (rAAV) has been accompanied by a better understanding of immune responses in both experimental animals and early clinical trials, and novel work is being performed to modulate the immune response. While the immune responses to rAAV remains a major challenge in translating experimental drugs to approved medicine, and will likely require more than a single solution, we now better understand the hurdles to formulate and test experimental solutions to surmount them.


Subject(s)
Dependovirus/immunology , Genetic Therapy , Genetic Vectors/immunology , Host Microbial Interactions/immunology , Immunity, Innate , Parvoviridae Infections/immunology , Adaptive Immunity , Animals , Capsid Proteins/genetics , Capsid Proteins/immunology , Clinical Trials as Topic , Humans , Mice
13.
Genome Med ; 10(1): 31, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29673390

ABSTRACT

We describe a method that enables the multiplex screening of a pool of many different donor cell lines. Our method accurately predicts each donor proportion from the pool without requiring the use of unique DNA barcodes as markers of donor identity. Instead, we take advantage of common single nucleotide polymorphisms, whole-genome sequencing, and an algorithm to calculate the proportions from the sequencing data. By testing using simulated and real data, we showed that our method robustly predicts the individual proportions from a mixed-pool of numerous donors, thus enabling the multiplexed testing of diverse donor cells en masse.More information is available at https://pgpresearch.med.harvard.edu/poolseq/.


Subject(s)
B-Lymphocytes/metabolism , High-Throughput Nucleotide Sequencing , Tissue Donors , Whole Genome Sequencing , Algorithms , Computer Simulation , Genome, Human , Humans , Polymorphism, Single Nucleotide/genetics , Sample Size
14.
Sci Rep ; 7: 46148, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387241

ABSTRACT

The Personal Genome Project (PGP) is an effort to enroll many participants to create an open-access repository of genome, health and trait data for research. However, PGP participants are not enrolled for studying any specific traits and participants choose the phenotypes to disclose. To measure the extent and willingness and to encourage and guide participants to contribute phenotypes, we developed an algorithm to score and rank the phenotypes and participants of the PGP. The scoring algorithm calculates the participation index (P-index) for every participant, where 0 indicates no reported phenotypes and 100 indicate complete phenotype reporting. We calculated the P-index for all 5,015 participants in the PGP and they ranged from 0 to 96.7. We found that participants mainly have either high scores (P-index > 90, 29.5%) or low scores (P-index < 10, 57.8%). While, there are significantly more males than female participants (1,793 versus 1,271), females tend to have on average higher P-indexes (P = 0.015). We also reported the P-indexes of participants based on demographics and states like Missouri and Massachusetts have better P-indexes than states like Utah and Minnesota. The P-index can therefore be used as an unbiased way to measure and rank participant's phenotypic contribution towards the PGP.


Subject(s)
Phenotype , Algorithms , Cohort Studies , Disease , Female , Genome, Human , Geography , Humans , Male , Quantitative Trait, Heritable , Surveys and Questionnaires , United States
15.
Nat Rev Microbiol ; 14(6): 360-73, 2016 06.
Article in English | MEDLINE | ID: mdl-27174148

ABSTRACT

The co-evolution of viruses with their hosts has led to the emergence of viral pathogens that are adept at evading or actively suppressing host immunity. Pattern recognition receptors (PRRs) are key components of antiviral immunity that detect conserved molecular features of viral pathogens and initiate signalling that results in the expression of antiviral genes. In this Review, we discuss the strategies that viruses use to escape immune surveillance by key intracellular sensors of viral RNA or DNA, with a focus on RIG-I-like receptors (RLRs), cyclic GMP-AMP synthase (cGAS) and interferon-γ (IFNγ)-inducible protein 16 (IFI16). Such viral strategies include the sequestration or modification of viral nucleic acids, interference with specific post-translational modifications of PRRs or their adaptor proteins, the degradation or cleavage of PRRs or their adaptors, and the sequestration or relocalization of PRRs. An understanding of viral immune-evasion mechanisms at the molecular level may guide the development of vaccines and antivirals.


Subject(s)
DNA, Viral/metabolism , Immune Evasion , RNA, Viral/metabolism , Receptors, Pattern Recognition/immunology , Viruses/immunology , Viruses/pathogenicity , Animals , Cytoplasm/genetics , DEAD Box Protein 58/metabolism , DNA, Viral/immunology , Humans , Immunity, Innate , Inflammasomes/metabolism , Interferon Inducers/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Processing, Post-Translational , RNA, Viral/immunology , Signal Transduction , Viruses/metabolism
16.
Nat Immunol ; 17(5): 523-30, 2016 May.
Article in English | MEDLINE | ID: mdl-26998762

ABSTRACT

14-3-3 proteins regulate biological processes by binding to phosphorylated serine or phosphorylated threonine motifs of cellular proteins. Among the 14-3-3 proteins, 14-3-3ɛ serves a crucial function in antiviral immunity by mediating the cytosol-to-mitochondrial membrane translocation of the pathogen sensor RIG-I. Here we found that the NS3 protein of dengue virus (DV) bound to 14-3-3ɛ and prevented translocation of RIG-I to the adaptor MAVS and thereby blocked antiviral signaling. Intriguingly, a highly conserved phosphomimetic RxEP motif in NS3 was essential for the binding of 14-3-3ɛ. A recombinant mutant DV deficient in binding to 14-3-3ɛ showed impairment in antagonism of RIG-I and elicited a markedly augmented innate immune response and enhanced T cell activation. Our work reveals a novel phosphomimetic-based mechanism for viral antagonism of 14-3-3-mediated immunity, which might guide the rational design of therapeutics.


Subject(s)
14-3-3 Proteins/immunology , DEAD-box RNA Helicases/immunology , Immunity, Innate/immunology , Serine Endopeptidases/immunology , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Cell Line , Cell Line, Tumor , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , HEK293 Cells , Humans , Immunoblotting , Microscopy, Confocal , Phosphorylation/immunology , RNA Interference/immunology , Receptors, Immunologic , Sequence Homology, Amino Acid , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/immunology
17.
Cell Host Microbe ; 17(3): 285-287, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25766287

ABSTRACT

The pathogen sensor RIG-I recognizes viral RNA and signals to induce an antiviral response. In this issue of Cell Host & Microbe, Weber et al. (2015), along with recent work by Sato et al. (2015), demonstrate that RIG-I directly inhibits viral replication independent of antiviral signaling.


Subject(s)
Gene Products, pol/antagonists & inhibitors , Hepatitis B virus/physiology , Hepatitis B, Chronic/immunology , Hepatocytes/physiology , Liver/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , RNA, Viral/immunology , Animals , Female , Humans
18.
Curr Opin Virol ; 12: 7-14, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25644461

ABSTRACT

Mammalian cells have the intrinsic capacity to detect viral pathogens and to initiate an antiviral response that is characterized by the induction of interferons (IFNs) and proinflammatory cytokines. A delicate regulation of the signaling pathways that lead to cytokine production is needed to ensure effective clearance of the virus, while preventing tissue damage caused by excessive cytokine release. Here, we focus on the mechanisms that modulate the signal transduction triggered by RIG-I-like receptors (RLRs) and their adaptor protein MAVS, key components of the host machinery for sensing foreign RNA. Specifically, we summarize recent advances in understanding how RLR signaling is regulated by posttranslational and posttranscriptional mechanisms, microRNAs (miRNAs) and autophagy. We further discuss how viruses target these regulatory mechanisms for immune evasion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Signal Transduction , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology , Viruses/pathogenicity , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy , Cytokines/immunology , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Gene Expression Regulation , Humans , Immune Evasion , Immunity, Innate , Interferons/immunology , MicroRNAs/genetics , Receptors, Immunologic
19.
Sci Signal ; 7(307): ra3, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24399297

ABSTRACT

Ubiquitylation is an important mechanism for regulating innate immune responses to viral infections. Attachment of lysine 63 (Lys(63))-linked ubiquitin chains to the RNA sensor retinoic acid-inducible gene-I (RIG-I) by the ubiquitin E3 ligase tripartite motif protein 25 (TRIM25) leads to the activation of RIG-I and stimulates production of the antiviral cytokines interferon-α (IFN-α) and IFN-ß. Conversely, Lys(48)-linked ubiquitylation of TRIM25 by the linear ubiquitin assembly complex (LUBAC) stimulates the proteasomal degradation of TRIM25, thereby inhibiting the RIG-I signaling pathway. Here, we report that ubiquitin-specific protease 15 (USP15) deubiquitylates TRIM25, preventing the LUBAC-dependent degradation of TRIM25. Through protein purification and mass spectrometry analysis, we identified USP15 as an interaction partner of TRIM25 in human cells. Knockdown of endogenous USP15 by specific small interfering RNA markedly enhanced the ubiquitylation of TRIM25. In contrast, expression of wild-type USP15, but not its catalytically inactive mutant, reduced the Lys(48)-linked ubiquitylation of TRIM25, leading to its stabilization. Furthermore, ectopic expression of USP15 enhanced the TRIM25- and RIG-I-dependent production of type I IFN and suppressed RNA virus replication. In contrast, depletion of USP15 resulted in decreased IFN production and markedly enhanced viral replication. Together, these data identify USP15 as a critical regulator of the TRIM25- and RIG-I-mediated antiviral immune response, thereby highlighting the intricate regulation of innate immune signaling.


Subject(s)
DEAD-box RNA Helicases/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Ubiquitin-Protein Ligases/immunology , Ubiquitin-Specific Proteases/immunology , Antiviral Agents/immunology , Antiviral Agents/metabolism , Blotting, Western , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/immunology , Interferon-alpha/immunology , Interferon-alpha/metabolism , Interferon-beta/immunology , Interferon-beta/metabolism , Lysine/metabolism , Microscopy, Confocal , Newcastle disease virus/genetics , Newcastle disease virus/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Binding/genetics , Protein Binding/immunology , Proteolysis , RNA Interference , Receptors, Immunologic , Sendai virus/immunology , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology , Virus Replication/immunology
20.
PLoS Pathog ; 9(3): e1003232, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555248

ABSTRACT

Human T-cell Immunoglobulin and Mucin-domain containing proteins (TIM1, 3, and 4) specifically bind phosphatidylserine (PS). TIM1 has been proposed to serve as a cellular receptor for hepatitis A virus and Ebola virus and as an entry factor for dengue virus. Here we show that TIM1 promotes infection of retroviruses and virus-like particles (VLPs) pseudotyped with a range of viral entry proteins, in particular those from the filovirus, flavivirus, New World arenavirus and alphavirus families. TIM1 also robustly enhanced the infection of replication-competent viruses from the same families, including dengue, Tacaribe, Sindbis and Ross River viruses. All interactions between TIM1 and pseudoviruses or VLPs were PS-mediated, as demonstrated with liposome blocking and TIM1 mutagenesis experiments. In addition, other PS-binding proteins, such as Axl and TIM4, promoted infection similarly to TIM1. Finally, the blocking of PS receptors on macrophages inhibited the entry of Ebola VLPs, suggesting that PS receptors can contribute to infection in physiologically relevant cells. Notably, infection mediated by the entry proteins of Lassa fever virus, influenza A virus and SARS coronavirus was largely unaffected by TIM1 expression. Taken together our data show that TIM1 and related PS-binding proteins promote infection of diverse families of enveloped viruses, and may therefore be useful targets for broad-spectrum antiviral therapies.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Host-Pathogen Interactions , Phosphatidylserines/metabolism , Proto-Oncogene Proteins/metabolism , Virus Diseases/metabolism , Viruses/pathogenicity , Animals , Antiviral Agents/pharmacology , Capsid , Cell Line , Dogs , Humans , Macrophages/metabolism , Macrophages/virology , Mice , Receptors, Virus/metabolism , Rho Guanine Nucleotide Exchange Factors , Viral Envelope Proteins , Virion/metabolism , Virus Diseases/virology , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...