Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Genet ; 54(5): 603-612, 2022 05.
Article in English | MEDLINE | ID: mdl-35513721

ABSTRACT

Genome-wide association studies (GWASs) have uncovered hundreds of autoimmune disease-associated loci; however, the causal genetic variants within each locus are mostly unknown. Here, we perform high-throughput allele-specific reporter assays to prioritize disease-associated variants for five autoimmune diseases. By examining variants that both promote allele-specific reporter expression and are located in accessible chromatin, we identify 60 putatively causal variants that enrich for statistically fine-mapped variants by up to 57.8-fold. We introduced the risk allele of a prioritized variant (rs72928038) into a human T cell line and deleted the orthologous sequence in mice, both resulting in reduced BACH2 expression. Naive CD8 T cells from mice containing the deletion had reduced expression of genes that suppress activation and maintain stemness and, upon acute viral infection, displayed greater propensity to become effector T cells. Our results represent an example of an effective approach for prioritizing variants and studying their physiologically relevant effects.


Subject(s)
Autoimmune Diseases , Genome-Wide Association Study , Alleles , Animals , Autoimmune Diseases/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Mice , Polymorphism, Single Nucleotide/genetics , Regulatory Sequences, Nucleic Acid , T-Lymphocytes
2.
Hum Mol Genet ; 31(12): 1946-1961, 2022 06 22.
Article in English | MEDLINE | ID: mdl-34970970

ABSTRACT

BACKGROUND: FCGR2A binds antibody-antigen complexes to regulate the abundance of circulating and deposited complexes along with downstream immune and autoimmune responses. Although the abundance of FCRG2A may be critical in immune-mediated diseases, little is known about whether its surface expression is regulated through cis genomic elements and non-coding variants. In the current study, we aimed to characterize the regulation of FCGR2A expression, the impact of genetic variation and its association with autoimmune disease. METHODS: We applied CRISPR-based interference and editing to scrutinize 1.7 Mb of open chromatin surrounding the FCGR2A gene to identify regulatory elements. Relevant transcription factors (TFs) binding to these regions were defined through public databases. Genetic variants affecting regulation were identified using luciferase reporter assays and were verified in a cohort of 1996 genotyped healthy individuals using flow cytometry. RESULTS: We identified a complex proximal region and five distal enhancers regulating FCGR2A. The proximal region split into subregions upstream and downstream of the transcription start site, was enriched in binding of inflammation-regulated TFs, and harbored a variant associated with FCGR2A expression in primary myeloid cells. One distal enhancer region was occupied by CCCTC-binding factor (CTCF) whose binding site was disrupted by a rare genetic variant, altering gene expression. CONCLUSIONS: The FCGR2A gene is regulated by multiple proximal and distal genomic regions, with links to autoimmune disease. These findings may open up novel therapeutic avenues where fine-tuning of FCGR2A levels may constitute a part of treatment strategies for immune-mediated diseases.


Subject(s)
Autoimmune Diseases , Enhancer Elements, Genetic , Receptors, IgG , Autoimmune Diseases/genetics , Binding Sites , Genomics , Genotype , Humans , Receptors, IgG/genetics
3.
Nature ; 593(7858): 238-243, 2021 05.
Article in English | MEDLINE | ID: mdl-33828297

ABSTRACT

Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer-gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions.


Subject(s)
Enhancer Elements, Genetic/genetics , Genetic Predisposition to Disease , Genetic Variation/genetics , Genome, Human/genetics , Genome-Wide Association Study , Inflammatory Bowel Diseases/genetics , Cell Line , Chromosomes, Human, Pair 10/genetics , Cyclophilins/genetics , Dendritic Cells , Female , Humans , Macrophages/metabolism , Male , Mitochondria/metabolism , Organ Specificity/genetics , Phenotype
4.
Nat Commun ; 12(1): 1611, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712590

ABSTRACT

Genome-wide association studies of Systemic Lupus Erythematosus (SLE) nominate 3073 genetic variants at 91 risk loci. To systematically screen these variants for allelic transcriptional enhancer activity, we construct a massively parallel reporter assay (MPRA) library comprising 12,396 DNA oligonucleotides containing the genomic context around every allele of each SLE variant. Transfection into the Epstein-Barr virus-transformed B cell line GM12878 reveals 482 variants with enhancer activity, with 51 variants showing genotype-dependent (allelic) enhancer activity at 27 risk loci. Comparison of MPRA results in GM12878 and Jurkat T cell lines highlights shared and unique allelic transcriptional regulatory mechanisms at SLE risk loci. In-depth analysis of allelic transcription factor (TF) binding at and around allelic variants identifies one class of TFs whose DNA-binding motif tends to be directly altered by the risk variant and a second class of TFs that bind allelically without direct alteration of their motif by the variant. Collectively, our approach provides a blueprint for the discovery of allelic gene regulation at risk loci for any disease and offers insight into the transcriptional regulatory mechanisms underlying SLE.


Subject(s)
Alleles , Genetic Predisposition to Disease/genetics , Lupus Erythematosus, Systemic/genetics , B-Lymphocytes , Cell Line , Chromatin , Gene Expression Regulation , Genome-Wide Association Study , Genotype , Herpesvirus 4, Human , Humans , Quantitative Trait Loci , Synaptogyrins/genetics , T-Lymphocytes
5.
Genome Biol ; 21(1): 134, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493396

ABSTRACT

Improved methods are needed to model CRISPR screen data for interrogation of genetic elements that alter reporter gene expression readout. We create MAUDE (Mean Alterations Using Discrete Expression) for quantifying the impact of guide RNAs on a target gene's expression in a pooled, sorting-based expression screen. MAUDE quantifies guide-level effects by modeling the distribution of cells across sorting expression bins. It then combines guides to estimate the statistical significance and effect size of targeted genetic elements. We demonstrate that MAUDE outperforms previous approaches and provide experimental design guidelines to best leverage MAUDE, which is available on https://github.com/Carldeboer/MAUDE.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression , Genetic Techniques , RNA, Guide, Kinetoplastida , Software , Algorithms , CRISPR-Cas Systems , Models, Genetic
6.
Nat Commun ; 11(1): 1237, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144282

ABSTRACT

Genome-wide association studies have associated thousands of genetic variants with complex traits and diseases, but pinpointing the causal variant(s) among those in tight linkage disequilibrium with each associated variant remains a major challenge. Here, we use seven experimental assays to characterize all common variants at the multiple disease-associated TNFAIP3 locus in five disease-relevant immune cell lines, based on a set of features related to regulatory potential. Trait/disease-associated variants are enriched among SNPs prioritized based on either: (1) residing within CRISPRi-sensitive regulatory regions, or (2) localizing in a chromatin accessible region while displaying allele-specific reporter activity. Of the 15 trait/disease-associated haplotypes at TNFAIP3, 9 have at least one variant meeting one or both of these criteria, 5 of which are further supported by genetic fine-mapping. Our work provides a comprehensive strategy to characterize genetic variation at important disease-associated loci, and aids in the effort to identify trait causal genetic variants.


Subject(s)
Autoimmune Diseases/genetics , Genetic Loci/genetics , Genome-Wide Association Study/methods , Multifactorial Inheritance/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Cell Line, Tumor , Genetic Predisposition to Disease , Genetic Variation/immunology , Haplotypes/genetics , Haplotypes/immunology , Humans , Linkage Disequilibrium , Multifactorial Inheritance/immunology , Proof of Concept Study
8.
Cell ; 175(4): 998-1013.e20, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388456

ABSTRACT

Treatment of cancer has been revolutionized by immune checkpoint blockade therapies. Despite the high rate of response in advanced melanoma, the majority of patients succumb to disease. To identify factors associated with success or failure of checkpoint therapy, we profiled transcriptomes of 16,291 individual immune cells from 48 tumor samples of melanoma patients treated with checkpoint inhibitors. Two distinct states of CD8+ T cells were defined by clustering and associated with patient tumor regression or progression. A single transcription factor, TCF7, was visualized within CD8+ T cells in fixed tumor samples and predicted positive clinical outcome in an independent cohort of checkpoint-treated patients. We delineated the epigenetic landscape and clonality of these T cell states and demonstrated enhanced antitumor immunity by targeting novel combinations of factors in exhausted cells. Our study of immune cell transcriptomes from tumors demonstrates a strategy for identifying predictors, mechanisms, and targets for enhancing checkpoint immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Melanoma/immunology , Transcriptome , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/immunology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Apyrase/antagonists & inhibitors , Apyrase/immunology , Cell Line, Tumor , Humans , Leukocyte Common Antigens/antagonists & inhibitors , Leukocyte Common Antigens/immunology , Melanoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T Cell Transcription Factor 1/metabolism
9.
Proc Natl Acad Sci U S A ; 115(30): E7222-E7230, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29987030

ABSTRACT

Gene expression is controlled by sequence-specific transcription factors (TFs), which bind to regulatory sequences in DNA. TF binding occurs in nucleosome-depleted regions of DNA (NDRs), which generally encompass regions with lengths similar to those protected by nucleosomes. However, less is known about where within these regions specific TFs tend to be found. Here, we characterize the positional bias of inferred binding sites for 103 TFs within ∼500,000 NDRs across 47 cell types. We find that distinct classes of TFs display different binding preferences: Some tend to have binding sites toward the edges, some toward the center, and some at other positions within the NDR. These patterns are highly consistent across cell types, suggesting that they may reflect TF-specific intrinsic structural or functional characteristics. In particular, TF classes with binding sites at NDR edges are enriched for those known to interact with histones and chromatin remodelers, whereas TFs with central enrichment interact with other TFs and cofactors such as p300. Our results suggest distinct regiospecific binding patterns and functions of TF classes within enhancers.


Subject(s)
Gene Expression Regulation/physiology , Response Elements/physiology , Transcription Factors/metabolism , Humans , Jurkat Cells , Transcription Factors/genetics , U937 Cells
10.
Nat Commun ; 8(1): 1136, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29070816

ABSTRACT

Treatment with immune checkpoint blockade (CPB) therapies often leads to prolonged responses in patients with metastatic melanoma, but the common mechanisms of primary and acquired resistance to these agents remain incompletely characterized and have yet to be validated in large cohorts. By analyzing longitudinal tumor biopsies from 17 metastatic melanoma patients treated with CPB therapies, we observed point mutations, deletions or loss of heterozygosity (LOH) in beta-2-microglobulin (B2M), an essential component of MHC class I antigen presentation, in 29.4% of patients with progressing disease. In two independent cohorts of melanoma patients treated with anti-CTLA4 and anti-PD1, respectively, we find that B2M LOH is enriched threefold in non-responders (~30%) compared to responders (~10%) and associated with poorer overall survival. Loss of both copies of B2M is found only in non-responders. B2M loss is likely a common mechanism of resistance to therapies targeting CTLA4 or PD1.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigen Presentation/drug effects , Drug Resistance, Neoplasm/drug effects , Melanoma/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antigen Presentation/genetics , CTLA-4 Antigen/immunology , Drug Resistance, Neoplasm/genetics , Female , Humans , Loss of Heterozygosity , Melanoma/genetics , Melanoma/pathology , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Point Mutation , Programmed Cell Death 1 Receptor/immunology , beta 2-Microglobulin/genetics
11.
Immunity ; 43(4): 690-702, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26410627

ABSTRACT

The differentiation of CD4(+) helper T cell subsets with diverse effector functions is accompanied by changes in metabolism required to meet their bioenergetic demands. We find that follicular B helper T (Tfh) cells exhibited less proliferation, glycolysis, and mitochondrial respiration, accompanied by reduced mTOR kinase activity compared to T helper 1 (Th1) cells in response to acute viral infection. IL-2-mediated activation of the Akt kinase and mTORc1 signaling was both necessary and sufficient to shift differentiation away from Tfh cells, instead promoting that of Th1 cells. These findings were not the result of generalized signaling attenuation in Tfh cells, because they retained the ability to flux calcium and activate NFAT-transcription-factor-dependent cytokine production. These data identify the interleukin-2 (IL-2)-mTORc1 axis as a critical orchestrator of the reciprocal balance between Tfh and Th1 cell fates and their respective metabolic activities after acute viral infection.


Subject(s)
Interleukin-2/physiology , Multiprotein Complexes/physiology , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis , Calcium Signaling , Cell Cycle , Cell Division , Enzyme Activation , Glucose/metabolism , Glycolysis , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocytic choriomeningitis virus/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , NFATC Transcription Factors/physiology , Oxygen Consumption , Positive Regulatory Domain I-Binding Factor 1 , Specific Pathogen-Free Organisms , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
12.
Genome Med ; 7(1): 57, 2015.
Article in English | MEDLINE | ID: mdl-26089985

ABSTRACT

Genetic analyses of autoimmune diseases have revealed hundreds of disease-associated DNA variants, but the identity and function of the causal variants are understudied and warrant deeper mechanistic studies. Here, we highlight methods for deciphering how alleles that are associated with autoimmune disease alter the human immune system, and suggest strategies for future autoimmune genetic research.

13.
Elife ; 4: e04851, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25569154

ABSTRACT

T follicular helper cells (Tfh) are crucial for the initiation and maintenance of germinal center (GC) reactions and high affinity, isotype-switched antibody responses. In this study, we demonstrate that direct TGF-ß signaling to CD4 T cells is important for the formation of influenza-specific Tfh cells, GC reactions, and development of isotype-switched, flu-specific antibody responses. Early during infection, TGF-ß signaling suppressed the expression of the high affinity IL-2 receptor α chain (CD25) on virus-specific CD4 T cells, which tempered IL-2 signaling and STAT5 and mammalian target of rapamycin (mTOR) activation in Tfh precursor CD4 T cells. Inhibition of mTOR allowed for the differentiation of Tfh cells in the absence of TGF-ßR signaling, suggesting that TGF-ß insulates Tfh progenitor cells from IL-2-delivered mTOR signals, thereby promoting Tfh differentiation during acute viral infection. These findings identify a new pathway critical for the generation of Tfh cells and humoral responses during respiratory viral infections.


Subject(s)
Antibody Formation/immunology , Immunoglobulin Class Switching/immunology , Lung/immunology , Mucous Membrane/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/metabolism , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Gene Expression Profiling , Germinal Center/immunology , Lung/pathology , Lung/virology , Mice , Mucous Membrane/pathology , Mucous Membrane/virology , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Species Specificity , TOR Serine-Threonine Kinases/metabolism
15.
Immunity ; 40(3): 367-77, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24631156

ABSTRACT

Follicular helper T (Tfh) cells are required for the establishment of T-dependent B cell memory and high affinity antibody-secreting cells. We have revealed herein opposing roles for signal transducer and activator of transcription 3 (STAT3) and type I interferon (IFN) signaling in the differentiation of Tfh cells following viral infection. STAT3-deficient CD4(+) T cells had a profound defect in Tfh cell differentiation, accompanied by decreased germinal center (GC) B cells and antigen-specific antibody production during acute infection with lymphocytic choriomeningitis virus. STAT3-deficient Tfh cells had strikingly increased expression of a number of IFN-inducible genes, in addition to enhanced T-bet synthesis, thus adopting a T helper 1 (Th1) cell-like effector phenotype. Conversely, IFN-αß receptor blockade restored Tfh and GC B cell phenotypes in mice containing STAT3-deficient CD4(+) T cells. These data suggest mutually repressive roles for STAT3 and type I IFN signaling pathways in the differentiation of Tfh cells following viral infection.


Subject(s)
Cell Differentiation , Interferon Type I/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antibodies, Viral/immunology , Antibody Specificity/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Gene Expression Profiling , Gene Expression Regulation , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulin Class Switching/genetics , Interferon Type I/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Knockout , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , Transcriptome
16.
PLoS One ; 8(7): e67828, 2013.
Article in English | MEDLINE | ID: mdl-23874453

ABSTRACT

While tuberculosis susceptibility has historically been ascribed to failed inflammation, it is now known that an excess of leukotriene A4 hydrolase (LTA4H), which catalyzes the final step in leukotriene B4 (LTB4) synthesis, produces a hyperinflammatory state and tuberculosis susceptibility. Here we show that the LTB4-inactivating enzyme leukotriene B4 dehydrogenase/prostaglandin reductase 1 (LTB4DH/PTGR1) restricts inflammation and independently confers resistance to tuberculous infection. LTB4DH overexpression counters the susceptibility resulting from LTA4H excess while ltb4dh-deficient animals can be rescued pharmacologically by LTB4 receptor antagonists. These data place LTB4DH as a key modulator of TB susceptibility and suggest new tuberculosis therapeutic strategies.


Subject(s)
Alcohol Oxidoreductases/metabolism , Leukotriene B4/metabolism , Mycobacterium Infections/enzymology , Mycobacterium Infections/metabolism , Tuberculosis/enzymology , Tuberculosis/metabolism , Amino Acid Sequence , Animals , Humans , Inflammation/enzymology , Inflammation/metabolism , Molecular Sequence Data , Receptors, Leukotriene B4/metabolism , Sequence Alignment , Zebrafish
17.
Cell ; 148(3): 434-46, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304914

ABSTRACT

Susceptibility to tuberculosis is historically ascribed to an inadequate immune response that fails to control infecting mycobacteria. In zebrafish, we find that susceptibility to Mycobacterium marinum can result from either inadequate or excessive acute inflammation. Modulation of the leukotriene A(4) hydrolase (LTA4H) locus, which controls the balance of pro- and anti-inflammatory eicosanoids, reveals two distinct molecular routes to mycobacterial susceptibility converging on dysregulated TNF levels: inadequate inflammation caused by excess lipoxins and hyperinflammation driven by excess leukotriene B(4). We identify therapies that specifically target each of these extremes. In humans, we identify a single nucleotide polymorphism in the LTA4H promoter that regulates its transcriptional activity. In tuberculous meningitis, the polymorphism is associated with inflammatory cell recruitment, patient survival and response to adjunctive anti-inflammatory therapy. Together, our findings suggest that host-directed therapies tailored to patient LTA4H genotypes may counter detrimental effects of either extreme of inflammation.


Subject(s)
Mycobacterium Infections/drug therapy , Mycobacterium Infections/immunology , Tuberculosis, Meningeal/drug therapy , Tuberculosis, Meningeal/immunology , Animals , Disease Models, Animal , Humans , Inflammation/immunology , Leukotriene A4/genetics , Leukotriene A4/immunology , Leukotriene B4/genetics , Leukotriene B4/immunology , Lipoxins/immunology , Mitochondria/metabolism , Mycobacterium Infections/genetics , Mycobacterium marinum , Polymorphism, Genetic , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Signal Transduction , Transcription, Genetic , Tuberculosis, Meningeal/genetics , Tumor Necrosis Factor-alpha/metabolism , Zebrafish/embryology , Zebrafish/immunology
18.
Cell ; 140(5): 717-30, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20211140

ABSTRACT

Exposure to Mycobacterium tuberculosis produces varied early outcomes, ranging from resistance to infection to progressive disease. Here we report results from a forward genetic screen in zebrafish larvae that identify multiple mutant classes with distinct patterns of innate susceptibility to Mycobacterium marinum. A hypersusceptible mutant maps to the lta4h locus encoding leukotriene A(4) hydrolase, which catalyzes the final step in the synthesis of leukotriene B(4) (LTB(4)), a potent chemoattractant and proinflammatory eicosanoid. lta4h mutations confer hypersusceptibility independent of LTB(4) reduction, by redirecting eicosanoid substrates to anti-inflammatory lipoxins. The resultant anti-inflammatory state permits increased mycobacterial proliferation by limiting production of tumor necrosis factor. In humans, we find that protection from both tuberculosis and multibacillary leprosy is associated with heterozygosity for LTA4H polymorphisms that have previously been correlated with differential LTB(4) production. Our results suggest conserved roles for balanced eicosanoid production in vertebrate resistance to mycobacterial infection.


Subject(s)
Epoxide Hydrolases/genetics , Fish Diseases/genetics , Leprosy/genetics , Tuberculosis/genetics , Animals , Disease Models, Animal , Fish Diseases/immunology , Genetic Predisposition to Disease , Humans , Leprosy/immunology , Tuberculosis/immunology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL