Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Bioinformatics ; 34(10): 1741-1749, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29329369

ABSTRACT

Motivation: Individual genetic variants explain only a small fraction of heritability in some diseases. Some variants have weak marginal effects on disease risk, but their joint effects are significantly stronger when occurring together. Most studies on such epistatic interactions have focused on methods for identifying the interactions and interpreting individual cases, but few have explored their general functional basis. This was due to the lack of a comprehensive list of epistatic interactions and uncertainties in associating variants to genes. Results: We conducted a large-scale survey of published research articles to compile the first comprehensive list of epistatic interactions in human diseases with detailed annotations. We used various methods to associate these variants to genes to ensure robustness. We found that these genes are significantly more connected in protein interaction networks, are more co-expressed and participate more often in the same pathways. We demonstrate using the list to discover novel disease pathways. Contact: kevinyip@cse.cuhk.edu.hk. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Disease Susceptibility , Epistasis, Genetic , Proteins/genetics , Humans , Proteins/analysis , Software
2.
Genome Biol ; 18(1): 230, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29195502

ABSTRACT

We present a new method, OMSV, for accurately and comprehensively identifying structural variations (SVs) from optical maps. OMSV detects both homozygous and heterozygous SVs, SVs of various types and sizes, and SVs with or without creating or destroying restriction sites. We show that OMSV has high sensitivity and specificity, with clear performance gains over the latest method. Applying OMSV to a human cell line, we identified hundreds of SVs >2 kbp, with 68 % of them missed by sequencing-based callers. Independent experimental validation confirmed the high accuracy of these SVs. The OMSV software is available at http://yiplab.cse.cuhk.edu.hk/omsv/ .


Subject(s)
Genomic Structural Variation , Genomics/methods , Software , Computational Biology/methods , Computer Simulation , Genome, Human , Humans
3.
Elife ; 52016 06 28.
Article in English | MEDLINE | ID: mdl-27350258

ABSTRACT

The receptor tyrosine kinase (RTK) AXL is induced in response to type I interferons (IFNs) and limits their production through a negative feedback loop. Enhanced production of type I IFNs in Axl(-/-) dendritic cells (DCs) in vitro have led to speculation that inhibition of AXL would promote antiviral responses. Notwithstanding, type I IFNs also exert potent immunosuppressive functions. Here we demonstrate that ablation of AXL enhances the susceptibility to infection by influenza A virus and West Nile virus. The increased type I IFN response in Axl(-/-) mice was associated with diminished DC maturation, reduced production of IL-1ß, and defective antiviral T cell immunity. Blockade of type I IFN receptor or administration of IL-1ß to Axl(-/-) mice restored the antiviral adaptive response and control of infection. Our results demonstrate that AXL is essential for limiting the immunosuppressive effects of type I IFNs and enabling the induction of protective antiviral adaptive immunity.


Subject(s)
Influenza A virus/immunology , Lymphocyte Activation , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , T-Lymphocytes/immunology , West Nile virus/immunology , Animals , Cells, Cultured , Disease Models, Animal , Interferon Type I/metabolism , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , West Nile Fever/immunology , Axl Receptor Tyrosine Kinase
4.
Science ; 352(6284): 463-6, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27102485

ABSTRACT

Influenza A virus (IAV) causes up to half a million deaths worldwide annually, 90% of which occur in older adults. We show that IAV-infected monocytes from older humans have impaired antiviral interferon production but retain intact inflammasome responses. To understand the in vivo consequence, we used mice expressing a functional Mx gene encoding a major interferon-induced effector against IAV in humans. In Mx1-intact mice with weakened resistance due to deficiencies in Mavs and Tlr7, we found an elevated respiratory bacterial burden. Notably, mortality in the absence of Mavs and Tlr7 was independent of viral load or MyD88-dependent signaling but dependent on bacterial burden, caspase-1/11, and neutrophil-dependent tissue damage. Therefore, in the context of weakened antiviral resistance, vulnerability to IAV disease is a function of caspase-dependent pathology.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Myxovirus Resistance Proteins/physiology , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Animals , Bacterial Infections/etiology , Caspase 1/metabolism , Caspases/metabolism , Caspases, Initiator , Female , Humans , Immunity, Innate/genetics , Influenza, Human/complications , Interferon-beta/immunology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Monocytes/immunology , Myxovirus Resistance Proteins/genetics , Neutrophils/immunology , Respiratory Tract Infections/microbiology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Viral Load , Young Adult
5.
BMC Genomics ; 15: 1178, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25539629

ABSTRACT

BACKGROUND: In the competing endogenous RNA (ceRNA) hypothesis, different transcripts communicate through a competition for their common targeting microRNAs (miRNAs). Individual examples have clearly shown the functional importance of ceRNA in gene regulation and cancer biology. It remains unclear to what extent gene expression levels are regulated by ceRNA in general. One major hurdle to studying this problem is the intertwined connections in miRNA-target networks, which makes it difficult to isolate the effects of individual miRNAs. RESULTS: Here we propose computational methods for decomposing a complex miRNA-target network into largely autonomous modules called microRNA-target biclusters (MTBs). Each MTB contains a relatively small number of densely connected miRNAs and mRNAs with few connections to other miRNAs and mRNAs. Each MTB can thus be individually analyzed with minimal crosstalk with other MTBs. Our approach differs from previous methods for finding modules in miRNA-target networks by not making any pre-assumptions about expression patterns, thereby providing objective information for testing the ceRNA hypothesis. We show that the expression levels of miRNAs and mRNAs in an MTB are significantly more anti-correlated than random miRNA-mRNA pairs and other validated and predicted miRNA-target pairs, demonstrating the biological relevance of MTBs. We further show that there is widespread correlation of expression between mRNAs in same MTBs under a wide variety of parameter settings, and the correlation remains even when co-regulatory effects are controlled for, which suggests potential widespread expression buffering between these mRNAs, which is consistent with the ceRNA hypothesis. Lastly, we also propose a potential use of MTBs in functional annotation of miRNAs. CONCLUSIONS: MTBs can be used to help identify autonomous miRNA-target modules for testing the generality of the ceRNA hypothesis experimentally. The identified modules can also be used to test other properties of miRNA-target networks in general.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , MicroRNAs/genetics , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Proc Natl Acad Sci U S A ; 110(34): 13910-5, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918369

ABSTRACT

Induction of a proinflammatory response is the hallmark of host innate defense against invading pathogens. Host recognition of influenza A virus (IAV) infection relies on pattern-recognition receptors, including Toll-like receptor 7 (TLR7) and retinoic acid inducible gene-1 (RIG-I) for the activation of innate-immune responses. Here, we show that following a physiological low dose of IAV infection, viral sensing by either TLR7 or RIG-I induces a proinflammatory program that promotes viral replication. Transfer of bronchoalveolar lavage from infected wild-type mice into the airway of mice deficient in TLR7 and RIG-I pathways was sufficient to restore viral replication efficiency. Comparison of IAV-infected cells revealed that inflammatory mediators elicited by TLR7 and RIG-I signaling recruit viral target cells to the airway, thereby enhancing viral load within the respiratory tract. Our data suggest that IAV uses physiological levels of inflammatory responses for its replicative advantage and highlight the complex interplay between viruses and the host innate-immune responses.


Subject(s)
DEAD-box RNA Helicases/metabolism , Immunity, Innate/immunology , Influenza A virus/immunology , Membrane Glycoproteins/metabolism , Orthomyxoviridae Infections/immunology , Respiratory Tract Infections/virology , Signal Transduction/immunology , Toll-Like Receptor 7/metabolism , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/virology , Cytokines/analysis , DEAD Box Protein 58 , Flow Cytometry , Histological Techniques , Immunohistochemistry , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/immunology , Viral Load , Virus Replication/physiology
7.
Nat Immunol ; 14(3): 246-53, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23314004

ABSTRACT

Immune responses to vaccines require direct recognition of pathogen-associated molecular patterns (PAMPs) through pattern-recognition receptors (PRRs) on dendritic cells (DCs). Unlike vaccination, infection by a live pathogen often impairs DC function and inflicts additional damage on the host. Here we found that after infection with live influenza A virus, signaling through the interleukin 1 receptor (IL-1R) was required for productive priming of CD8(+) T cells, but signaling through the PRRs TLR7 and RIG-I was not. DCs activated by IL-1 in trans were both required and sufficient for the generation of virus-specific CD8(+) T cell immunity. Our data demonstrate a critical role for a bystander cytokine in the priming of CD8(+) T cells during infection with a live virus.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Influenza A virus/immunology , Receptors, Interleukin-1/metabolism , Receptors, Pattern Recognition/metabolism , Animals , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Cell Movement , Dendritic Cells/metabolism , Dendritic Cells/virology , Interleukin-1/immunology , Lymphocyte Activation , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Nerve Tissue Proteins/metabolism , Orthomyxoviridae Infections/immunology , Receptors, CCR7/biosynthesis , Receptors, Cell Surface , Receptors, Interleukin-1/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction , Toll-Like Receptor 7/metabolism
8.
Immunol Rev ; 245(1): 209-26, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22168422

ABSTRACT

Human skin and mucosal surfaces are in constant contact with resident and invasive microbes. Recognition of microbial products by receptors of the innate immune system triggers rapid innate defense and transduces signals necessary for initiating and maintaining the adaptive immune responses. Microbial sensing by innate pattern-recognition receptors is not restricted to pathogens. Rather, proper development, function, and maintenance of innate and adaptive immunity rely on continuous recognition of products derived from the microorganisms indigenous to the internal and external surfaces of mammalian host. Tonic immune activation by the resident microbiota governs host susceptibility to intestinal and extra-intestinal infections, including those caused by viruses. This review highlights recent developments in innate viral recognition leading to adaptive immunity, and discusses potential links between viruses, microbiota, and the host immune system. Furthermore, we discuss the possible roles of microbiome in chronic viral infection and pathogenesis of autoimmune disease and speculate on the benefit for probiotic therapies against such diseases.


Subject(s)
Antigens, Viral/immunology , Autoimmune Diseases/immunology , Metagenome/immunology , Receptors, Pattern Recognition/immunology , Virus Diseases/immunology , Adaptive Immunity , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/etiology , Autoimmune Diseases/microbiology , Humans , Immunity, Innate , Probiotics/therapeutic use , Virus Diseases/complications , Virus Diseases/drug therapy , Virus Diseases/microbiology
9.
Proc Natl Acad Sci U S A ; 108(13): 5354-9, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21402903

ABSTRACT

Although commensal bacteria are crucial in maintaining immune homeostasis of the intestine, the role of commensal bacteria in immune responses at other mucosal surfaces remains less clear. Here, we show that commensal microbiota composition critically regulates the generation of virus-specific CD4 and CD8 T cells and antibody responses following respiratory influenza virus infection. By using various antibiotic treatments, we found that neomycin-sensitive bacteria are associated with the induction of productive immune responses in the lung. Local or distal injection of Toll-like receptor (TLR) ligands could rescue the immune impairment in the antibiotic-treated mice. Intact microbiota provided signals leading to the expression of mRNA for pro-IL-1ß and pro-IL-18 at steady state. Following influenza virus infection, inflammasome activation led to migration of dendritic cells (DCs) from the lung to the draining lymph node and T-cell priming. Our results reveal the importance of commensal microbiota in regulating immunity in the respiratory mucosa through the proper activation of inflammasomes.


Subject(s)
Adaptive Immunity/immunology , Influenza A virus/immunology , Metagenome , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Bacteria/immunology , Bacteria/pathogenicity , Dendritic Cells/immunology , Homeostasis , Host-Pathogen Interactions , Humans , Inflammasomes , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/virology , Toll-Like Receptors/immunology
10.
Trends Immunol ; 32(1): 34-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21147034

ABSTRACT

Influenza viruses infect a wide range of avian and mammalian host species including humans. Influenza viruses are a major cause of human respiratory infections and mortality. The innate immune system recognizes influenza viruses through multiple mechanisms. These include endosomal recognition through the Toll-like receptor 7 (TLR7) and cytosolic recognition through the retinoic acid inducible gene I (RIG-I). Recent studies also identified the role of nucleotide binding oligomerization domain (NOD)-like receptors (NLRs) in innate detection of influenza viruses, leading to the activation of the inflammasomes. Here, we review the cellular and molecular mechanisms by which influenza virus infection leads to inflammasome activation, and discuss the consequences of such activation in innate and adaptive immune defense against influenza viruses.


Subject(s)
Inflammasomes/immunology , Influenza, Human/immunology , Orthomyxoviridae/physiology , Adaptive Immunity , Animals , Humans , Immunity, Innate
11.
Nat Immunol ; 11(5): 404-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20383149

ABSTRACT

Influenza virus, a negative-stranded RNA virus that causes severe illness in humans and animals, stimulates the inflammasome through the Nod-like receptor NLRP3. However, the mechanism by which influenza virus activates the NLRP3 inflammasome is unknown. Here we show that the influenza virus M2 protein, a proton-selective ion channel important in viral pathogenesis, stimulates the NLRP3 inflammasome pathway. M2 channel activity was required for the activation of inflammasomes by influenza and was sufficient to activate inflammasomes in primed macrophages and dendritic cells. M2-induced activation of inflammasomes required its localization to the Golgi apparatus and was dependent on the pH gradient. Our results show a mechanism by which influenza virus infection activates inflammasomes and identify the sensing of disturbances in intracellular ionic concentrations as a previously unknown pathogen-recognition pathway.


Subject(s)
Carrier Proteins/metabolism , Dendritic Cells/metabolism , Ion Channels/metabolism , Macrophages/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/physiology , Viral Matrix Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Genetic Engineering , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Golgi Apparatus/virology , Hydrogen-Ion Concentration/drug effects , Ion Channels/genetics , Ion Channels/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Macrophages/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monensin/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein , Oncogene Proteins, Viral/pharmacology , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/physiopathology , Potassium Chloride/pharmacology , Protein Transport/drug effects , Protons , Sequence Deletion/genetics , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...