Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioinformatics ; 38(13): 3490-3492, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35608303

ABSTRACT

MOTIVATION: With continually improved instrumentation, Fourier transform infrared (FTIR) microspectroscopy can now be used to capture thousands of high-resolution spectra for chemical characterization of a sample. The spatially resolved nature of this method lends itself well to histological profiling of complex biological specimens. However, current software can make joint analysis of multiple samples challenging and, for large datasets, computationally infeasible. RESULTS: To overcome these limitations, we have developed Photizo-an open-source Python library enabling high-throughput spectral data pre-processing, visualization and downstream analysis, including principal component analysis, clustering, macromolecular quantification and mapping. Photizo can be used for analysis of data without a spatial component, as well as spatially resolved data, obtained e.g. by scanning mode IR microspectroscopy and IR imaging by focal plane array detector. AVAILABILITY AND IMPLEMENTATION: The code underlying this article is available at https://github.com/DendrouLab/Photizo with access to example data available at https://zenodo.org/record/6417982#.Yk2O9TfMI6A.


Subject(s)
Libraries , Software , Spectroscopy, Fourier Transform Infrared/methods , Gene Library , Principal Component Analysis
2.
Annu Rev Genomics Hum Genet ; 20: 73-97, 2019 08 31.
Article in English | MEDLINE | ID: mdl-30848957

ABSTRACT

Pregnancy presents a singular physiological scenario during which the maternal immune system must accommodate the semiallogeneic fetus. Fluctuations between pro- and anti-inflammatory states are required throughout gestation to facilitate uterine tissue remodeling, fetal growth and development, and finally birth. Tolerance for the fetus must be established and maintained without fundamentally compromising the maternal immune system function, so that both the mother and fetus are protected from foreign insults. Here, we review our current understanding of how genetic variation at both maternal and fetal loci affects implantation and placenta formation, thereby determining the likelihood of a successful pregnancy outcome or the development of pregnancy-related complications. We also consider the impact of pregnancy on both the maternal and fetal systemic immune systems and the related implications for modulating ongoing autoimmune diseases and triggering their development.


Subject(s)
Autoimmune Diseases/genetics , Fetus/immunology , Genome, Human/immunology , Immune System/metabolism , Placenta/immunology , Pregnancy Complications/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Female , Fetus/metabolism , Gene Expression Regulation, Developmental , Genetic Loci , Genetic Variation , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Immune System/growth & development , Immune Tolerance , Immunogenetics/methods , Placenta/metabolism , Pregnancy , Pregnancy Complications/immunology , Pregnancy Complications/pathology , Receptors, KIR/genetics , Receptors, KIR/immunology
3.
Nature ; 488(7412): 508-511, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22801493

ABSTRACT

Although there has been much success in identifying genetic variants associated with common diseases using genome-wide association studies (GWAS), it has been difficult to demonstrate which variants are causal and what role they have in disease. Moreover, the modest contribution that these variants make to disease risk has raised questions regarding their medical relevance. Here we have investigated a single nucleotide polymorphism (SNP) in the TNFRSF1A gene, that encodes tumour necrosis factor receptor 1 (TNFR1), which was discovered through GWAS to be associated with multiple sclerosis (MS), but not with other autoimmune conditions such as rheumatoid arthritis, psoriasis and Crohn's disease. By analysing MS GWAS data in conjunction with the 1000 Genomes Project data we provide genetic evidence that strongly implicates this SNP, rs1800693, as the causal variant in the TNFRSF1A region. We further substantiate this through functional studies showing that the MS risk allele directs expression of a novel, soluble form of TNFR1 that can block TNF. Importantly, TNF-blocking drugs can promote onset or exacerbation of MS, but they have proven highly efficacious in the treatment of autoimmune diseases for which there is no association with rs1800693. This indicates that the clinical experience with these drugs parallels the disease association of rs1800693, and that the MS-associated TNFR1 variant mimics the effect of TNF-blocking drugs. Hence, our study demonstrates that clinical practice can be informed by comparing GWAS across common autoimmune diseases and by investigating the functional consequences of the disease-associated genetic variation.


Subject(s)
Genetic Predisposition to Disease/genetics , Multiple Sclerosis/chemically induced , Multiple Sclerosis/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Alleles , Exons/genetics , Genome, Human/genetics , Genome-Wide Association Study , Genomics , Genotype , Humans , Multiple Sclerosis/drug therapy , RNA Splicing/genetics , Receptors, Tumor Necrosis Factor, Type I/analysis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Solubility , Tumor Necrosis Factor-alpha/metabolism , United Kingdom
4.
Immunol Rev ; 248(1): 10-22, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22725951

ABSTRACT

Multiple sclerosis (MS) is a disabling autoimmune disease of the central nervous system, which affects approximately 0.1% of the population with variable degrees of severity. Disease susceptibility is jointly determined by genetic predisposition and environmental contribution. However, as only a handful of genetic risk factors have been investigated beyond initial genome-wide association studies and environmental factors are largely unidentified, the exact mechanism of how these associations interact remains speculative. Our current understanding of this complex and heterogeneous disease has been advanced by experimental data obtained from animal modeling, with particular focus on the mouse MS model, experimental autoimmune encephalomyelitis. Manipulation of the mouse genome to study genetic risk factors has largely proved informative, but it also has limitations. Integration effects of transgene insertion, gene copy number, and expression variation, as well as differences in regulatory elements between mouse and human, are some of the hurdles faced when using such models to understand human gene variants in mice. Furthermore, as the list of MS disease-associated genetic variants continues to increase, so does the demand to find new approaches to study them. A new generation of humanized mice aims to tighten the gap between mouse and human, such that MS-associated genetic variants can be modeled more physiologically and systematically.


Subject(s)
Disease Models, Animal , Mice , Multiple Sclerosis/genetics , Animals , Environment , Genetic Association Studies , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Mice/genetics , Multiple Sclerosis/immunology
5.
J Immunol ; 190(6): 2554-66, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418630

ABSTRACT

As the thymus involutes with age, the maintenance of peripheral naive T cells in humans becomes strongly dependent on peripheral cell division. However, mechanisms that orchestrate homeostatic division remain unclear. In this study we present evidence that the frequency of naive CD4 T cells that express CD25 (IL-2 receptor α-chain) increases with age on subsets of both CD31(+) and CD31(-) naive CD4 T cells. Analyses of TCR excision circles from sorted subsets indicate that CD25(+) naive CD4 T cells have undergone more rounds of homeostatic proliferation than their CD25(-) counterparts in both the CD31(+) and CD31(-) subsets, indicating that CD25 is a marker of naive CD4 T cells that have preferentially responded to survival signals from self-Ags or cytokines. CD25 expression on CD25(-) naive CD4 T cells can be induced by IL-7 in vitro in the absence of TCR activation. Although CD25(+) naive T cells respond to lower concentrations of IL-2 as compared with their CD25(-) counterparts, IL-2 responsiveness is further increased in CD31(-) naive T cells by their expression of the signaling IL-2 receptor ß-chain CD122, forming with common γ-chain functional high-affinity IL-2 receptors. CD25 plays a role during activation: CD25(+) naive T cells stimulated in an APC-dependent manner were shown to produce increased levels of IL-2 as compared with their CD25(-) counterparts. This study establishes CD25(+) naive CD4 T cells, which are further delineated by CD31 expression, as a major functionally distinct immune cell subset in humans that warrants further characterization in health and disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cellular Senescence/immunology , Receptors, Interleukin-2/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Adult , Age Factors , CD4-Positive T-Lymphocytes/cytology , Cell Death/genetics , Cell Death/immunology , Cell Differentiation/genetics , Cells, Cultured , Cellular Senescence/genetics , Child , Humans , Immunophenotyping , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/blood , Interleukin-2 Receptor alpha Subunit/genetics , Protein Binding/genetics , Protein Binding/immunology , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-2/physiology , Thymus Gland/cytology , Young Adult
6.
Genome Biol ; 25(1): 109, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38671451

ABSTRACT

Single-cell multiplexing techniques (cell hashing and genetic multiplexing) combine multiple samples, optimizing sample processing and reducing costs. Cell hashing conjugates antibody-tags or chemical-oligonucleotides to cell membranes, while genetic multiplexing allows to mix genetically diverse samples and relies on aggregation of RNA reads at known genomic coordinates. We develop hadge (hashing deconvolution combined with genotype information), a Nextflow pipeline that combines 12 methods to perform both hashing- and genotype-based deconvolution. We propose a joint deconvolution strategy combining best-performing methods and demonstrate how this approach leads to the recovery of previously discarded cells in a nuclei hashing of fresh-frozen brain tissue.


Subject(s)
Single-Cell Analysis , Single-Cell Analysis/methods , Humans , Brain/metabolism , Brain/cytology , Software , Genotype
7.
J Immunol ; 184(9): 5075-84, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20363978

ABSTRACT

We have used the public sequencing and annotation of the mouse genome to delimit the previously resolved type 1 diabetes (T1D) insulin-dependent diabetes (Idd)18 interval to a region on chromosome 3 that includes the immunologically relevant candidate gene, Vav3. To test the candidacy of Vav3, we developed a novel congenic strain that enabled the resolution of Idd18 to a 604-kb interval, designated Idd18.1, which contains only two annotated genes: the complete sequence of Vav3 and the last exon of the gene encoding NETRIN G1, Ntng1. Targeted sequencing of Idd18.1 in the NOD mouse strain revealed that allelic variation between NOD and C57BL/6J (B6) occurs in noncoding regions with 138 single nucleotide polymorphisms concentrated in the introns between exons 20 and 27 and immediately after the 3' untranslated region. We observed differential expression of VAV3 RNA transcripts in thymocytes when comparing congenic mouse strains with B6 or NOD alleles at Idd18.1. The T1D protection associated with B6 alleles of Idd18.1/Vav3 requires the presence of B6 protective alleles at Idd3, which are correlated with increased IL-2 production and regulatory T cell function. In the absence of B6 protective alleles at Idd3, we detected a second T1D protective B6 locus, Idd18.3, which is closely linked to, but distinct from, Idd18.1. Therefore, genetic mapping, sequencing, and gene expression evidence indicate that alteration of VAV3 expression is an etiological factor in the development of autoimmune beta-cell destruction in NOD mice. This study also demonstrates that a congenic strain mapping approach can isolate closely linked susceptibility genes.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Genetic Loci/immunology , Genetic Predisposition to Disease , Insulin/physiology , Mice, Congenic , Physical Chromosome Mapping/methods , Proto-Oncogene Proteins c-vav/genetics , Alleles , Animals , Crosses, Genetic , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Exons/genetics , Female , Gene Expression Regulation/immunology , Insulin/genetics , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nerve Tissue Proteins/genetics , Netrins , Proto-Oncogene Proteins c-vav/biosynthesis
8.
Nat Neurosci ; 25(7): 944-955, 2022 07.
Article in English | MEDLINE | ID: mdl-35726057

ABSTRACT

Progressive multiple sclerosis (MS) is characterized by unrelenting neurodegeneration, which causes cumulative disability and is refractory to current treatments. Drug development to prevent disease progression is an urgent clinical need yet is constrained by an incomplete understanding of its complex pathogenesis. Using spatial transcriptomics and proteomics on fresh-frozen human MS brain tissue, we identified multicellular mechanisms of progressive MS pathogenesis and traced their origin in relation to spatially distributed stages of neurodegeneration. By resolving ligand-receptor interactions in local microenvironments, we discovered defunct trophic and anti-inflammatory intercellular communications within areas of early neuronal decline. Proteins associated with neuronal damage in patient samples showed mechanistic concordance with published in vivo knockdown and central nervous system (CNS) disease models, supporting their causal role and value as potential therapeutic targets in progressive MS. Our findings provide a new framework for drug development strategies, rooted in an understanding of the complex cellular and signaling dynamics in human diseased tissue that facilitate this debilitating disease.


Subject(s)
Central Nervous System Diseases , Multiple Sclerosis , Central Nervous System Diseases/complications , Disease Progression , Humans , Multiple Sclerosis/pathology , Neurons/metabolism , Proteomics
9.
Ann Clin Transl Neurol ; 8(7): 1502-1507, 2021 07.
Article in English | MEDLINE | ID: mdl-33991459

ABSTRACT

Improvements in assays for detecting serum antibodies against myelin oligodendrocyte glycoprotein (MOG) have led to the appreciation of MOG-antibody-associated disease (MOGAD) as a novel disorder. However, much remains unknown about its etiology. We performed human leukocyte antigen (HLA) analysis in 82 MOGAD patients of European ancestry in the UK population. No HLA class II associations were observed, thus questioning the mechanism of anti-MOG antibody generation. A weak protective association of HLA-C*03:04 was observed (OR = 0.26, 95% CI = 0.10-0.71, pc  = 0.013), suggesting a need for continued efforts to better understand MOGAD genetics and pathophysiology.


Subject(s)
Autoantibodies/blood , Genetic Association Studies/methods , HLA Antigens/blood , Myelin-Oligodendrocyte Glycoprotein/blood , Neuromyelitis Optica/blood , Neuromyelitis Optica/epidemiology , Adult , Aged , Biomarkers/blood , Cohort Studies , Female , HLA Antigens/genetics , Humans , Male , Middle Aged , Myelin-Oligodendrocyte Glycoprotein/genetics , Neuromyelitis Optica/genetics , United Kingdom/epidemiology , Young Adult
10.
Nat Genet ; 52(1): 126-134, 2020 01.
Article in English | MEDLINE | ID: mdl-31873298

ABSTRACT

Genetic risk factors frequently affect multiple common human diseases, providing insight into shared pathophysiological pathways and opportunities for therapeutic development. However, systematic identification of genetic profiles of disease risk is limited by the availability of both comprehensive clinical data on population-scale cohorts and the lack of suitable statistical methodology that can handle the scale of and differential power inherent in multi-phenotype data. Here, we develop a disease-agnostic approach to cluster the genetic risk profiles for 3,025 genome-wide independent loci across 19,155 disease classification codes from 320,644 participants in the UK Biobank, representing a large and heterogeneous population. We identify 339 distinct disease association profiles and use multiple approaches to link clusters to the underlying biological pathways. We show how clusters can decompose the variance and covariance in risk for disease, thereby identifying underlying biological processes and their impact. We demonstrate the use of clusters in defining disease relationships and their potential in informing therapeutic strategies.


Subject(s)
Biological Specimen Banks , Genetic Diseases, Inborn/genetics , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Quantitative Trait, Heritable , Adult , Aged , Female , Gene-Environment Interaction , Humans , Male , Middle Aged , Phenotype , Prospective Studies , Risk Factors , United Kingdom
11.
Cell Death Dis ; 11(5): 303, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358523

ABSTRACT

Mixed lineage kinase domain-like (MLKL) is the main executor of necroptosis, an inflammatory form of programmed cell death. Necroptosis is implicated in combating infections, but also in contributing to numerous other clinical conditions, including cardiovascular diseases and neurodegenerative disorders. Inhibition of necroptosis is therefore of therapeutic interest. Here we report two siblings both of whom over the course of 35 years developed a similar progressive, neurodegenerative spectrum disorder characterized by paresis, ataxia and dysarthria. Magnetic resonance imaging of their central nervous system (CNS) revealed severe global cerebral volume loss and atrophy of the cerebellum and brainstem. These brothers are homozygous for a rare haplotype identified by whole genome sequencing carrying a frameshift variant in MLKL, as well as an in-frame deletion of one amino acid in the adjacent fatty acid 2-hydroxylase (FA2H) gene. Functional studies of patient-derived primary cells demonstrated that the variant in MLKL leads to a deficiency of MLKL protein resulting in impairment of necroptosis. Conversely, shotgun lipidomic analysis of the variant in FA2H shows no impact on either the abundance or the enzymatic activity of the encoded hydroxylase. To our knowledge, this is the first report of complete necroptosis deficiency in humans. The findings may suggest that impaired necroptosis is a novel mechanism of neurodegeneration, promoting a disorder that shares some clinical features with primary progressive multiple sclerosis (PPMS) and other neurodegenerative diseases. Importantly, the necroptotic deficiency does not cause symptoms outside the nervous system, nor does it confer susceptibility to infections. Given the current interest in pharmacological inhibition of necroptosis by targeting MLKL and its associated pathways, this strategy should be developed with caution, with careful consideration of the possible development of adverse neurological effects.


Subject(s)
Apoptosis/genetics , Necroptosis/genetics , Neurodegenerative Diseases/pathology , Protein Kinases/deficiency , Animals , Apoptosis/physiology , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Phosphorylation , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
12.
Nat Rev Immunol ; 18(5): 325-339, 2018 05.
Article in English | MEDLINE | ID: mdl-29292391

ABSTRACT

Fifty years since the first description of an association between HLA and human disease, HLA molecules have proven to be central to physiology, protective immunity and deleterious, disease-causing autoimmune reactivity. Technological advances have enabled pivotal progress in the determination of the molecular mechanisms that underpin the association between HLA genetics and functional outcome. Here, we review our current understanding of HLA molecules as the fundamental platform for immune surveillance and responsiveness in health and disease. We evaluate the scope for personalized antigen-specific disease prevention, whereby harnessing HLA-ligand interactions for clinical benefit is becoming a realistic prospect.


Subject(s)
Disease/genetics , HLA Antigens/genetics , Antigen Presentation , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Gene Expression Regulation , Genes, MHC Class I , Genes, MHC Class II , Genetic Association Studies , Genetic Variation , HLA Antigens/chemistry , HLA Antigens/metabolism , Humans , Immunogenetic Phenomena , Immunotherapy/methods , Models, Immunological , Molecular Mimicry/immunology , Protein Binding/immunology , Protein Stability , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology
13.
Curr Opin Immunol ; 49: 37-43, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28926740

ABSTRACT

Multiple sclerosis (MS) afflicts about 2.5 million people globally and poses a major personal and socioeconomic burden. The recognition of MS as an inflammatory disease, characterized by infiltration of immune cells into the central nervous system, has spurred research into the autoimmune etiology of the condition and has provided the rationale for its treatment through immunomodulation. Experience with immunotherapies in MS to date has suggested a disparity between the observed immune cell infiltration and the progressive loss of neurons. However, recent clinical efforts are providing new insights into progressive MS that once again place the immune system at center stage. This article reviews the main mechanisms of MS immunopathogenesis, and the benefits, risks and challenges of immunomodulatory treatments for the disease.


Subject(s)
B-Lymphocytes/immunology , Immunologic Factors/therapeutic use , Multiple Sclerosis/therapy , Animals , Humans , Immunomodulation , Lymphocyte Depletion , Patient Care
14.
Nat Genet ; 49(9): 1311-1318, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759005

ABSTRACT

Genetic discovery from the multitude of phenotypes extractable from routine healthcare data can transform understanding of the human phenome and accelerate progress toward precision medicine. However, a critical question when analyzing high-dimensional and heterogeneous data is how best to interrogate increasingly specific subphenotypes while retaining statistical power to detect genetic associations. Here we develop and employ a new Bayesian analysis framework that exploits the hierarchical structure of diagnosis classifications to analyze genetic variants against UK Biobank disease phenotypes derived from self-reporting and hospital episode statistics. Our method displays a more than 20% increase in power to detect genetic effects over other approaches and identifies new associations between classical human leukocyte antigen (HLA) alleles and common immune-mediated diseases (IMDs). By applying the approach to genetic risk scores (GRSs), we show the extent of genetic sharing among IMDs and expose differences in disease perception or diagnosis with potential clinical implications.


Subject(s)
Bayes Theorem , Delivery of Health Care/statistics & numerical data , Genetic Association Studies/statistics & numerical data , Health Information Systems/statistics & numerical data , Adult , Aged , Alleles , Cluster Analysis , Delivery of Health Care/classification , Female , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/statistics & numerical data , HLA Antigens/genetics , Humans , International Classification of Diseases/classification , International Classification of Diseases/statistics & numerical data , Logistic Models , Male , Middle Aged , Polymorphism, Single Nucleotide , United Kingdom
15.
Nat Commun ; 8: 15924, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28649982

ABSTRACT

Expression of HLA-C varies widely across individuals in an allele-specific manner. This variation in expression can influence efficacy of the immune response, as shown for infectious and autoimmune diseases. MicroRNA binding partially influences differential HLA-C expression, but the additional contributing factors have remained undetermined. Here we use functional and structural analyses to demonstrate that HLA-C expression is modulated not just at the RNA level, but also at the protein level. Specifically, we show that variation in exons 2 and 3, which encode the α1/α2 domains, drives differential expression of HLA-C allomorphs at the cell surface by influencing the structure of the peptide-binding cleft and the diversity of peptides bound by the HLA-C molecules. Together with a phylogenetic analysis, these results highlight the diversity and long-term balancing selection of regulatory factors that modulate HLA-C expression.


Subject(s)
HLA-C Antigens/chemistry , HLA-C Antigens/genetics , Alleles , Animals , Exons , Gene Expression Regulation , Genetic Variation , HLA-C Antigens/metabolism , Humans , Mammals/classification , Mammals/genetics , Pan troglodytes , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Phylogeny , Promoter Regions, Genetic , Protein Binding
16.
Nat Rev Neurol ; 12(12): 685-698, 2016 12.
Article in English | MEDLINE | ID: mdl-27857124

ABSTRACT

Neuroinflammation is emerging as a central process in many neurological conditions, either as a causative factor or as a secondary response to nervous system insult. Understanding the causes and consequences of neuroinflammation could, therefore, provide insight that is needed to improve therapeutic interventions across many diseases. However, the complexity of the pathways involved necessitates the use of high-throughput approaches to extensively interrogate the process, and appropriate strategies to translate the data generated into clinical benefit. Use of 'big data' aims to generate, integrate and analyse large, heterogeneous datasets to provide in-depth insights into complex processes, and has the potential to unravel the complexities of neuroinflammation. Limitations in data analysis approaches currently prevent the full potential of big data being reached, but some aspects of big data are already yielding results. The implementation of 'omics' analyses in particular is becoming routine practice in biomedical research, and neuroimaging is producing large sets of complex data. In this Review, we evaluate the impact of the drive to collect and analyse big data on our understanding of neuroinflammation in disease. We describe the breadth of big data that are leading to an evolution in our understanding of this field, exemplify how these data are beginning to be of use in a clinical setting, and consider possible future directions.


Subject(s)
Datasets as Topic , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Inflammation/diagnosis , Metabolomics/methods , Nervous System Diseases/diagnosis , Neuroimaging/methods , Pharmacogenetics/methods , Precision Medicine/methods , Proteomics/methods , Animals , Humans , Inflammation/complications , Inflammation/drug therapy , Inflammation/immunology , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Nervous System Diseases/immunology
17.
Sci Transl Med ; 8(363): 363ra149, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807284

ABSTRACT

Thousands of genetic variants have been identified, which contribute to the development of complex diseases, but determining how to elucidate their biological consequences for translation into clinical benefit is challenging. Conflicting evidence regarding the functional impact of genetic variants in the tyrosine kinase 2 (TYK2) gene, which is differentially associated with common autoimmune diseases, currently obscures the potential of TYK2 as a therapeutic target. We aimed to resolve this conflict by performing genetic meta-analysis across disorders; subsequent molecular, cellular, in vivo, and structural functional follow-up; and epidemiological studies. Our data revealed a protective homozygous effect that defined a signaling optimum between autoimmunity and immunodeficiency and identified TYK2 as a potential drug target for certain common autoimmune disorders.


Subject(s)
Autoimmune Diseases/genetics , Genetic Association Studies , TYK2 Kinase/genetics , Animals , Autoimmunity , CD4-Positive T-Lymphocytes/cytology , Cytokines/metabolism , Epigenesis, Genetic , Female , Genetic Variation , Genomics , Genotype , HEK293 Cells , Homozygote , Humans , Immune System , Janus Kinase 2/chemistry , Leukocytes, Mononuclear/cytology , Male , Mice , Mutation, Missense , Phenotype , Polymorphism, Single Nucleotide , Protein Conformation , Quantitative Trait Loci , Recombination, Genetic , Sequence Analysis, RNA , Signal Transduction , Transcriptome
18.
Nat Rev Immunol ; 15(9): 545-58, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26250739

ABSTRACT

Two decades of clinical experience with immunomodulatory treatments for multiple sclerosis point to distinct immunological pathways that drive disease relapses and progression. In light of this, we discuss our current understanding of multiple sclerosis immunopathology, evaluate long-standing hypotheses regarding the role of the immune system in the disease and delineate key questions that are still unanswered. Recent and anticipated advances in the field of immunology, and the increasing recognition of inflammation as an important component of neurodegeneration, are shaping our conceptualization of disease pathophysiology, and we explore the potential implications for improved healthcare provision to patients in the future.


Subject(s)
Autoimmunity/immunology , Inflammation/immunology , Multiple Sclerosis/immunology , Autoimmunity/genetics , Environmental Exposure , Genetic Predisposition to Disease , Humans , Inflammation/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/therapy
19.
Nat Genet ; 47(10): 1107-1113, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343388

ABSTRACT

Association studies have greatly refined the understanding of how variation within the human leukocyte antigen (HLA) genes influences risk of multiple sclerosis. However, the extent to which major effects are modulated by interactions is poorly characterized. We analyzed high-density SNP data on 17,465 cases and 30,385 controls from 11 cohorts of European ancestry, in combination with imputation of classical HLA alleles, to build a high-resolution map of HLA genetic risk and assess the evidence for interactions involving classical HLA alleles. Among new and previously identified class II risk alleles (HLA-DRB1*15:01, HLA-DRB1*13:03, HLA-DRB1*03:01, HLA-DRB1*08:01 and HLA-DQB1*03:02) and class I protective alleles (HLA-A*02:01, HLA-B*44:02, HLA-B*38:01 and HLA-B*55:01), we find evidence for two interactions involving pairs of class II alleles: HLA-DQA1*01:01-HLA-DRB1*15:01 and HLA-DQB1*03:01-HLA-DQB1*03:02. We find no evidence for interactions between classical HLA alleles and non-HLA risk-associated variants and estimate a minimal effect of polygenic epistasis in modulating major risk alleles.


Subject(s)
Genetic Predisposition to Disease , Histocompatibility Antigens Class II/immunology , Multiple Sclerosis/genetics , Alleles , Epistasis, Genetic , Histocompatibility Antigens Class II/genetics , Humans , Polymorphism, Single Nucleotide
20.
Neuron ; 84(6): 1105-7, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25521369

ABSTRACT

In this issue, Sorbara et al. (2014) demonstrate that axonal transport impairment is an early feature of neurodegeneration in multiple sclerosis models. This transport deficit is reversible by anti-inflammatory intervention but, if untreated, can contribute to late-stage axonal dystrophy.


Subject(s)
Axonal Transport/physiology , Axons/physiology , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Animals
SELECTION OF CITATIONS
SEARCH DETAIL