Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 181(1): 63-80, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243797

ABSTRACT

Autoimmune diseases are a result of the immune system being misdirected toward its host and have major and increasing unmet clinical needs. In general, present therapies are broadly acting and non-disease specific; consequently, they are associated with numerous side effects. Precise and early intervention strategies are urgently needed. We highlight the challenges, progress, and prospects in achieving these goals.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , Humans
2.
Immunity ; 57(2): 256-270.e10, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38354703

ABSTRACT

Antibodies can block immune receptor engagement or trigger the receptor machinery to initiate signaling. We hypothesized that antibody agonists trigger signaling by sterically excluding large receptor-type protein tyrosine phosphatases (RPTPs) such as CD45 from sites of receptor engagement. An agonist targeting the costimulatory receptor CD28 produced signals that depended on antibody immobilization and were sensitive to the sizes of the receptor, the RPTPs, and the antibody itself. Although both the agonist and a non-agonistic anti-CD28 antibody locally excluded CD45, the agonistic antibody was more effective. An anti-PD-1 antibody that bound membrane proximally excluded CD45, triggered Src homology 2 domain-containing phosphatase 2 recruitment, and suppressed systemic lupus erythematosus and delayed-type hypersensitivity in experimental models. Paradoxically, nivolumab and pembrolizumab, anti-PD-1-blocking antibodies used clinically, also excluded CD45 and were agonistic in certain settings. Reducing these agonistic effects using antibody engineering improved PD-1 blockade. These findings establish a framework for developing new and improved therapies for autoimmunity and cancer.


Subject(s)
Protein Tyrosine Phosphatases , Signal Transduction , Protein Tyrosine Phosphatases/metabolism , CD28 Antigens , Receptors, Immunologic
4.
Nature ; 625(7994): 312-320, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200293

ABSTRACT

The Holocene (beginning around 12,000 years ago) encompassed some of the most significant changes in human evolution, with far-reaching consequences for the dietary, physical and mental health of present-day populations. Using a dataset of more than 1,600 imputed ancient genomes1, we modelled the selection landscape during the transition from hunting and gathering, to farming and pastoralism across West Eurasia. We identify key selection signals related to metabolism, including that selection at the FADS cluster began earlier than previously reported and that selection near the LCT locus predates the emergence of the lactase persistence allele by thousands of years. We also find strong selection in the HLA region, possibly due to increased exposure to pathogens during the Bronze Age. Using ancient individuals to infer local ancestry tracts in over 400,000 samples from the UK Biobank, we identify widespread differences in the distribution of Mesolithic, Neolithic and Bronze Age ancestries across Eurasia. By calculating ancestry-specific polygenic risk scores, we show that height differences between Northern and Southern Europe are associated with differential Steppe ancestry, rather than selection, and that risk alleles for mood-related phenotypes are enriched for Neolithic farmer ancestry, whereas risk alleles for diabetes and Alzheimer's disease are enriched for Western hunter-gatherer ancestry. Our results indicate that ancient selection and migration were large contributors to the distribution of phenotypic diversity in present-day Europeans.


Subject(s)
Asian , European People , Genome, Human , Selection, Genetic , Humans , Affect , Agriculture/history , Alleles , Alzheimer Disease/genetics , Asia/ethnology , Asian/genetics , Diabetes Mellitus/genetics , Europe/ethnology , European People/genetics , Farmers/history , Genetic Loci/genetics , Genetic Predisposition to Disease , Genome, Human/genetics , History, Ancient , Human Migration , Hunting/history , Multigene Family/genetics , Phenotype , UK Biobank , Multifactorial Inheritance/genetics
5.
Nature ; 625(7994): 321-328, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200296

ABSTRACT

Multiple sclerosis (MS) is a neuro-inflammatory and neurodegenerative disease that is most prevalent in Northern Europe. Although it is known that inherited risk for MS is located within or in close proximity to immune-related genes, it is unknown when, where and how this genetic risk originated1. Here, by using a large ancient genome dataset from the Mesolithic period to the Bronze Age2, along with new Medieval and post-Medieval genomes, we show that the genetic risk for MS rose among pastoralists from the Pontic steppe and was brought into Europe by the Yamnaya-related migration approximately 5,000 years ago. We further show that these MS-associated immunogenetic variants underwent positive selection both within the steppe population and later in Europe, probably driven by pathogenic challenges coinciding with changes in diet, lifestyle and population density. This study highlights the critical importance of the Neolithic period and Bronze Age as determinants of modern immune responses and their subsequent effect on the risk of developing MS in a changing environment.


Subject(s)
Genetic Predisposition to Disease , Genome, Human , Grassland , Multiple Sclerosis , Humans , Datasets as Topic , Diet/ethnology , Diet/history , Europe/ethnology , Genetic Predisposition to Disease/history , Genetics, Medical , History, 15th Century , History, Ancient , History, Medieval , Human Migration/history , Life Style/ethnology , Life Style/history , Multiple Sclerosis/genetics , Multiple Sclerosis/history , Multiple Sclerosis/immunology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/history , Neurodegenerative Diseases/immunology , Population Density
6.
Nature ; 545(7653): 243-247, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28467828

ABSTRACT

Susceptibility and protection against human autoimmune diseases, including type I diabetes, multiple sclerosis, and Goodpasture disease, is associated with particular human leukocyte antigen (HLA) alleles. However, the mechanisms underpinning such HLA-mediated effects on self-tolerance remain unclear. Here we investigate the molecular mechanism of Goodpasture disease, an HLA-linked autoimmune renal disorder characterized by an immunodominant CD4+ T-cell self-epitope derived from the α3 chain of type IV collagen (α3135-145). While HLA-DR15 confers a markedly increased disease risk, the protective HLA-DR1 allele is dominantly protective in trans with HLA-DR15 (ref. 2). We show that autoreactive α3135-145-specific T cells expand in patients with Goodpasture disease and, in α3135-145-immunized HLA-DR15 transgenic mice, α3135-145-specific T cells infiltrate the kidney and mice develop Goodpasture disease. HLA-DR15 and HLA-DR1 exhibit distinct peptide repertoires and binding preferences and present the α3135-145 epitope in different binding registers. HLA-DR15-α3135-145 tetramer+ T cells in HLA-DR15 transgenic mice exhibit a conventional T-cell phenotype (Tconv) that secretes pro-inflammatory cytokines. In contrast, HLA-DR1-α3135-145 tetramer+ T cells in HLA-DR1 and HLA-DR15/DR1 transgenic mice are predominantly CD4+Foxp3+ regulatory T cells (Treg cells) expressing tolerogenic cytokines. HLA-DR1-induced Treg cells confer resistance to disease in HLA-DR15/DR1 transgenic mice. HLA-DR15+ and HLA-DR1+ healthy human donors display altered α3135-145-specific T-cell antigen receptor usage, HLA-DR15-α3135-145 tetramer+ Foxp3- Tconv and HLA-DR1-α3135-145 tetramer+ Foxp3+CD25hiCD127lo Treg dominant phenotypes. Moreover, patients with Goodpasture disease display a clonally expanded α3135-145-specific CD4+ T-cell repertoire. Accordingly, we provide a mechanistic basis for the dominantly protective effect of HLA in autoimmune disease, whereby HLA polymorphism shapes the relative abundance of self-epitope specific Treg cells that leads to protection or causation of autoimmunity.


Subject(s)
Anti-Glomerular Basement Membrane Disease/immunology , Autoimmunity/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Anti-Glomerular Basement Membrane Disease/pathology , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Collagen Type IV/chemistry , Collagen Type IV/immunology , Cytokines/immunology , Female , Forkhead Transcription Factors/metabolism , HLA-DR Serological Subtypes/immunology , HLA-DR1 Antigen/immunology , Humans , Immunodominant Epitopes , Male , Mice , Mice, Transgenic , Models, Molecular
7.
Proc Natl Acad Sci U S A ; 117(23): 12952-12960, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32457139

ABSTRACT

Effective biomarkers for multiple sclerosis diagnosis, assessment of prognosis, and treatment responses, in particular those measurable in blood, are largely lacking. We have investigated a broad set of protein biomarkers in cerebrospinal fluid (CSF) and plasma using a highly sensitive proteomic immunoassay. Cases from two independent cohorts were compared with healthy controls and patients with other neurological diseases. We identified and replicated 10 cerebrospinal fluid proteins including IL-12B, CD5, MIP-1a, and CXCL9 which had a combined diagnostic efficacy similar to immunoglobulin G (IgG) index and neurofilament light chain (area under the curve [AUC] = 0.95). Two plasma proteins, OSM and HGF, were also associated with multiple sclerosis in comparison to healthy controls. Sensitivity and specificity of combined CSF and plasma markers for multiple sclerosis were 85.7% and 73.5%, respectively. In the discovery cohort, eotaxin-1 (CCL11) was associated with disease duration particularly in patients who had secondary progressive disease (PCSF < 4 × 10-5, Pplasma < 4 × 10-5), and plasma CCL20 was associated with disease severity (P = 4 × 10-5), although both require further validation. Treatment with natalizumab and fingolimod showed different compartmental changes in protein levels of CSF and peripheral blood, respectively, including many disease-associated markers (e.g., IL12B, CD5) showing potential application for both diagnosing disease and monitoring treatment efficacy. We report a number of multiple sclerosis biomarkers in CSF and plasma for early disease detection and potential indicators for disease activity. Of particular importance is the set of markers discovered in blood, where validated biomarkers are lacking.


Subject(s)
Chemokine CCL11/analysis , Chemokine CCL20/blood , Inflammation/immunology , Multiple Sclerosis/diagnosis , Adult , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Case-Control Studies , Chemokine CCL11/immunology , Chemokine CCL20/immunology , Cohort Studies , Female , Humans , Inflammation/blood , Inflammation/cerebrospinal fluid , Male , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/immunology , Prognosis , Proteomics , Reproducibility of Results , Severity of Illness Index , Young Adult
8.
Eur J Immunol ; 51(4): 1002-1005, 2021 04.
Article in English | MEDLINE | ID: mdl-33368209

ABSTRACT

We created a TCR transgenic mouse with CD4+ T cells recognizing the immunodominant DQ2.5-glia-ω2 gluten epitope. We show that these cells respond to deamidated gluten feed in vivo and compare them to previously published α2- and γ1-specific mice. These mice may help enlighten key aspects of celiac disease pathogenesis.


Subject(s)
Glutens/genetics , HLA-DQ Antigens/genetics , Immunodominant Epitopes/genetics , Receptors, Antigen, T-Cell/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/genetics , Celiac Disease/immunology , Disease Models, Animal , Glutens/immunology , HLA-DQ Antigens/immunology , Humans , Immunodominant Epitopes/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology
9.
Nat Immunol ; 10(6): 636-46, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19412183

ABSTRACT

Although cytotoxic T lymphocytes (CTLs) in people infected with human immunodeficiency virus type 1 can potentially target multiple virus epitopes, the same few are recognized repeatedly. We show here that CTL immunodominance in regions of the human immunodeficiency virus type 1 group-associated antigen proteins p17 and p24 correlated with epitope abundance, which was strongly influenced by proteasomal digestion profiles, affinity for the transporter protein TAP, and trimming mediated by the endoplasmatic reticulum aminopeptidase ERAAP, and was moderately influenced by HLA affinity. Structural and functional analyses demonstrated that proteasomal cleavage 'preferences' modulated the number and length of epitope-containing peptides, thereby affecting the response avidity and clonality of T cells. Cleavage patterns were affected by both flanking and intraepitope CTL-escape mutations. Our analyses show that antigen processing shapes CTL response hierarchies and that viral evolution modifies cleavage patterns and suggest strategies for in vitro vaccine optimization.


Subject(s)
Antigen Presentation , HIV Antigens/immunology , HIV Core Protein p24/immunology , T-Lymphocytes, Cytotoxic/immunology , gag Gene Products, Human Immunodeficiency Virus/immunology , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Evolution, Molecular , HIV Antigens/metabolism , HIV Core Protein p24/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HLA-A Antigens/immunology , HLA-A Antigens/metabolism , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Leucyl Aminopeptidase/metabolism , Major Histocompatibility Complex , Models, Molecular , Molecular Sequence Data , Mutation , Proteasome Endopeptidase Complex/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Binding , T-Lymphocytes, Cytotoxic/virology , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Mult Scler ; 26(10): 1261-1264, 2020 09.
Article in English | MEDLINE | ID: mdl-32762494

ABSTRACT

Approximately 200,000 multiple sclerosis (MS) patients worldwide receive B-cell-depleting immunotherapy with rituximab (anti-CD20), which eliminates the ability to generate an antibody response to new infections. As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific antibodies might help viral clearance, these patients could be at risk of severe complications if infected. Here, we report on an MS patient who had received rituximab for ~3 years. The patient was examined 5 days before the onset of coronavirus disease 2019 (COVID-19) symptoms and was admitted to the hospital 2 days after. She recovered 14 days after symptom onset despite having a 0% B lymphocyte count and not developing SARS-CoV-2 immunoglobulin G (IgG) antibodies.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Multiple Sclerosis, Chronic Progressive/drug therapy , Pneumonia, Viral/immunology , Rituximab/therapeutic use , Betacoronavirus , CD4-CD8 Ratio , COVID-19 , Coronavirus Infections/complications , Coronavirus Infections/physiopathology , Disease Progression , Female , Humans , Lymphocyte Count , Middle Aged , Multiple Sclerosis, Chronic Progressive/complications , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/physiopathology , SARS-CoV-2
11.
Immunity ; 32(6): 766-77, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20541955

ABSTRACT

T cell antigen receptor (TCR) and coreceptor ligation is thought to initiate signal transduction by inducing activation of the kinase Lck. Here we showed that catalytically active Lck was present in unstimulated naive T cells and thymocytes and was readily detectable in these cells in lymphoid organs. In naive T cells up to approximately 40% of total Lck was constitutively activated, part of which was also phosphorylated on the C-terminal inhibitory site. Formation of activated Lck was independent of TCR and coreceptors but required Lck catalytic activity and its maintenance relied on monitoring by the HSP90-CDC37 chaperone complex to avoid degradation. The amount of activated Lck did not change after TCR and coreceptor engagement; however it determined the extent of TCR-zeta phosphorylation. Our findings suggest a dynamic regulation of Lck activity that can be promptly utilized to initiate T cell activation and have implications for signaling by other immune receptors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Enzyme Activation/immunology , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Microscopy, Confocal , Receptors, Antigen, T-Cell/metabolism
12.
Proc Natl Acad Sci U S A ; 113(20): 5682-7, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27114505

ABSTRACT

The αß T-cell coreceptor CD4 enhances immune responses more than 1 million-fold in some assays, and yet the affinity of CD4 for its ligand, peptide-major histocompatibility class II (pMHC II) on antigen-presenting cells, is so weak that it was previously unquantifiable. Here, we report that a soluble form of CD4 failed to bind detectably to pMHC II in surface plasmon resonance-based assays, establishing a new upper limit for the solution affinity at 2.5 mM. However, when presented multivalently on magnetic beads, soluble CD4 bound pMHC II-expressing B cells, confirming that it is active and allowing mapping of the native coreceptor binding site on pMHC II. Whereas binding was undetectable in solution, the affinity of the CD4/pMHC II interaction could be measured in 2D using CD4- and adhesion molecule-functionalized, supported lipid bilayers, yielding a 2D Kd of ∼5,000 molecules/µm(2) This value is two to three orders of magnitude higher than previously measured 2D Kd values for interacting leukocyte surface proteins. Calculations indicated, however, that CD4/pMHC II binding would increase rates of T-cell receptor (TCR) complex phosphorylation by threefold via the recruitment of Lck, with only a small, 2-20% increase in the effective affinity of the TCR for pMHC II. The affinity of CD4/pMHC II therefore seems to be set at a value that increases T-cell sensitivity by enhancing phosphorylation, without compromising ligand discrimination.


Subject(s)
CD4 Antigens/chemistry , HLA-A24 Antigen/chemistry , HLA-DRB1 Chains/chemistry , Binding Sites , CD4 Antigens/metabolism , HEK293 Cells , HLA-A24 Antigen/metabolism , HLA-DRB1 Chains/metabolism , Humans , Maltose-Binding Proteins/chemistry , Models, Molecular , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Stability , Surface Plasmon Resonance
13.
Immunity ; 30(3): 348-57, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19303388

ABSTRACT

Environmental factors account for 75% of the risk of developing multiple sclerosis (MS). Numerous infections have been suspected as environmental disease triggers, but none of them has consistently been incriminated, and it is unclear how so many different infections may play a role. We show that a microbial peptide, common to several major classes of bacteria, can induce MS-like disease in humanized mice by crossreacting with a T cell receptor (TCR) that also recognizes a peptide from myelin basic protein, a candidate MS autoantigen. Structural analysis demonstrates this crossreactivity is due to structural mimicry of a binding hotspot shared by self and microbial antigens, rather than to degenerate TCR recognition. Biophysical studies reveal that the autoreactive TCR binding affinity is markedly lower for the microbial (mimicry) peptide than for the autoantigenic peptide. Thus, these data suggest a possible explanation for the difficulty in incriminating individual infections in the development of MS.


Subject(s)
Autoimmune Diseases/immunology , Bacterial Proteins/immunology , Molecular Mimicry/immunology , Peptides/immunology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Cerebellum/pathology , Cross Reactions/immunology , Drosophila , Escherichia coli/immunology , HLA-D Antigens/metabolism , HLA-DR2 Antigen/metabolism , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Models, Molecular , Multiple Sclerosis/immunology , Peptides/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Spinal Cord/pathology , T-Lymphocytes/physiology
14.
J Immunol ; 197(9): 3575-3585, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671110

ABSTRACT

It has been difficult to translate promising results from DNA vaccination in mice to larger animals and humans. Previously, DNA vaccines encoding proteins that target Ag to MHC class II (MHC-II) molecules on APCs have been shown to induce rapid, enhanced, and long-lasting Ag-specific Ab titers in mice. In this study, we describe two novel DNA vaccines that as proteins target HLA class II (HLA-II) molecules. These vaccine proteins cross-react with MHC-II molecules in several species of larger mammals. When tested in ferrets and pigs, a single DNA delivery with low doses of the HLA-II-targeted vaccines resulted in rapid and increased Ab responses. Importantly, painless intradermal jet delivery of DNA was as effective as delivery by needle injection followed by electroporation. As an indication that the vaccines could also be useful for human application, HLA-II-targeted vaccine proteins were found to increase human CD4+ T cell responses by a factor of ×103 in vitro. Thus, targeting of Ag to MHC-II molecules may represent an attractive strategy for increasing efficacy of DNA vaccines in larger animals and humans.


Subject(s)
Antibody Formation , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Antigens/metabolism , Cell Proliferation , Cells, Cultured , Cross Reactions , Ferrets , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Mice , Swine , Vaccination
15.
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
16.
Nat Rev Immunol ; 6(4): 271-82, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16557259

ABSTRACT

MHC class II molecules on the surface of antigen-presenting cells display a range of peptides for recognition by the T-cell receptors of CD4+ T helper cells. Therefore, MHC class II molecules are central to effective adaptive immune responses, but conversely, genetic and epidemiological data have implicated these molecules in the pathogenesis of autoimmune diseases. Indeed, the strength of the associations between particular MHC class II alleles and disease render them the main genetic risk factors for autoimmune disorders such as type 1 diabetes. Here, we discuss the insights that the crystal structures of MHC class II molecules provide into the molecular mechanisms by which sequence polymorphisms might contribute to disease susceptibility.


Subject(s)
Autoimmune Diseases/genetics , HLA-D Antigens/genetics , Autoimmune Diseases/immunology , Binding Sites/genetics , Binding Sites/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/metabolism , HLA-D Antigens/chemistry , HLA-D Antigens/metabolism , Humans , Models, Molecular , Protein Binding/genetics , Protein Binding/immunology , Protein Conformation
17.
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
18.
Proc Natl Acad Sci U S A ; 109(39): E2615-24, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22955884

ABSTRACT

Microscopic polyangiitis is an autoimmune small-vessel vasculitis that often manifests as focal and necrotizing glomerulonephritis and renal failure. Antineutrophil cytoplasmic Abs (ANCAs) specific for myeloperoxidase (MPO) play a role in this disease, but the role of autoreactive MPO-specific CD4(+) T cells is uncertain. By screening overlapping peptides of 20 amino acids spanning the MPO molecule, we identified an immunodominant MPO CD4(+) T-cell epitope (MPO(409-428)). Immunizing C57BL/6 mice with MPO(409-428) induced focal necrotizing glomerulonephritis similar to that seen after whole MPO immunization, when MPO was deposited in glomeruli. Transfer of an MPO(409-428)-specific CD4(+) T-cell clone to Rag1(-/-) mice induced focal necrotizing glomerulonephritis when glomerular MPO deposition was induced either by passive transfer of MPO-ANCA and LPS or by planting MPO(409-428) conjugated to a murine antiglomerular basement membrane mAb. MPO(409-428) also induced biologically active anti-MPO Abs in mice. The MPO(409-428) epitope has a minimum immunogenic core region of 11 amino acids, MPO(415-426), with several critical residues. ANCA-activated neutrophils not only induce injury but lodged the autoantigen MPO in glomeruli, allowing autoreactive anti-MPO CD4(+) cells to induce delayed type hypersensitivity-like necrotizing glomerular lesions. These studies identify an immunodominant MPO T-cell epitope and redefine how effector responses can induce injury in MPO-ANCA-associated microscopic polyangiitis.


Subject(s)
Antibodies, Antineutrophil Cytoplasmic/immunology , Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Glomerulonephritis/immunology , Immunity, Cellular , Microscopic Polyangiitis/immunology , Peroxidase/immunology , Animals , Antibodies, Antineutrophil Cytoplasmic/genetics , Antibodies, Antineutrophil Cytoplasmic/metabolism , Autoantigens/genetics , Autoantigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Glomerulonephritis/enzymology , Glomerulonephritis/genetics , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Mice , Mice, Knockout , Microscopic Polyangiitis/enzymology , Microscopic Polyangiitis/genetics , Microscopic Polyangiitis/pathology , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/pathology , Peroxidase/genetics , Peroxidase/metabolism
19.
PLoS Genet ; 8(3): e1002550, 2012.
Article in English | MEDLINE | ID: mdl-22457633

ABSTRACT

The rapid evolution of Human Immunodeficiency Virus (HIV-1) allows studies of ongoing host-pathogen interactions. One key selective host factor is APOBEC3G (hA3G) that can cause extensive and inactivating Guanosine-to-Adenosine (G-to-A) mutation on HIV plus-strand DNA (termed hypermutation). HIV can inhibit this innate anti-viral defense through binding of the viral protein Vif to hA3G, but binding efficiency varies and hypermutation frequencies fluctuate in patients. A pivotal question is whether hA3G-induced G-to-A mutation is always lethal to the virus or if it may occur at sub-lethal frequencies that could increase viral diversification. We show in vitro that limiting-levels of hA3G-activity (i.e. when only a single hA3G-unit is likely to act on HIV) produce hypermutation frequencies similar to those in patients and demonstrate in silico that potentially non-lethal G-to-A mutation rates are ∼10-fold lower than the lowest observed hypermutation levels in vitro and in vivo. Our results suggest that even a single incorporated hA3G-unit is likely to cause extensive and inactivating levels of HIV hypermutation and that hypermutation therefore is typically a discrete "all or nothing" phenomenon. Thus, therapeutic measures that inhibit the interaction between Vif and hA3G will likely not increase virus diversification but expand the fraction of hypermutated proviruses within the infected host.


Subject(s)
Cytidine Deaminase/genetics , HIV-1/genetics , Host-Pathogen Interactions , vif Gene Products, Human Immunodeficiency Virus/metabolism , APOBEC-3G Deaminase , Cytidine Deaminase/metabolism , HIV Infections/genetics , HIV Infections/metabolism , HIV-1/metabolism , Humans , Mutation , Proviruses/genetics , Proviruses/growth & development , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/genetics
20.
Brain ; 136(Pt 9): 2657-76, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22734127

ABSTRACT

The immune system has crucial roles in the pathogenesis of multiple sclerosis. While the adaptive immune cell subsets, T and B cells, have been the main focus of immunological research in multiple sclerosis, it is now important to realize that the innate immune system also has a key involvement in regulating autoimmune responses in the central nervous system. Natural killer cells are innate lymphocytes that play vital roles in a diverse range of infections. There is evidence that they influence a number of autoimmune conditions. Recent studies in multiple sclerosis and its murine model, experimental autoimmune encephalomyelitis, are starting to provide some understanding of the role of natural killer cells in regulating inflammation in the central nervous system. Natural killer cells express a diverse range of polymorphic cell surface receptors, which interact with polymorphic ligands; this interaction controls the function and the activation status of the natural killer cell. In this review, we discuss evidence for the role of natural killer cells in multiple sclerosis and experimental autoimmune encephalomyelitis. We consider how a change in the balance of signals received by the natural killer cell influences its involvement in the ensuing immune response, in relation to multiple sclerosis.


Subject(s)
Killer Cells, Natural/pathology , Multiple Sclerosis , Receptors, KIR/metabolism , Animals , Antigens, CD/metabolism , Central Nervous System/immunology , Central Nervous System/pathology , Disease Models, Animal , Humans , Immune System/pathology , Killer Cells, Natural/classification , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Receptors, KIR/immunology
SELECTION OF CITATIONS
SEARCH DETAIL