Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.241
Filter
Add more filters

Publication year range
1.
Immunity ; 45(4): 719-736, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760337

ABSTRACT

Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/physiology , Adaptive Immunity/immunology , Animals , Autoimmunity/immunology , Autoimmunity/physiology , Humans , Immunity, Innate/immunology , Mice
2.
Immunity ; 43(5): 933-44, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588779

ABSTRACT

Mutations in ADAR, which encodes the ADAR1 RNA-editing enzyme, cause Aicardi-Goutières syndrome (AGS), a severe autoimmune disease associated with an aberrant type I interferon response. How ADAR1 prevents autoimmunity remains incompletely defined. Here, we demonstrate that ADAR1 is a specific and essential negative regulator of the MDA5-MAVS RNA sensing pathway. Moreover, we uncovered a MDA5-MAVS-independent function for ADAR1 in the development of multiple organs. We showed that the p150 isoform of ADAR1 uniquely regulated the MDA5 pathway, whereas both the p150 and p110 isoforms contributed to development. Abrupt deletion of ADAR1 in adult mice revealed that both of these functions were required throughout life. Our findings delineate genetically separable roles for both ADAR1 isoforms in vivo, with implications for the human diseases caused by ADAR mutations.


Subject(s)
Adenosine Deaminase/metabolism , Autoimmunity/physiology , DEAD-box RNA Helicases/metabolism , Protein Isoforms/metabolism , RNA Editing/physiology , RNA/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmune Diseases of the Nervous System/metabolism , HEK293 Cells , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1 , Mice , Nervous System Malformations/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology
3.
Diabetologia ; 66(11): 1971-1982, 2023 11.
Article in English | MEDLINE | ID: mdl-37488322

ABSTRACT

Type 1 diabetes results from the poorly understood process of islet autoimmunity, which ultimately leads to the loss of functional pancreatic beta cells. Mounting evidence supports the notion that the activation and evolution of islet autoimmunity in genetically susceptible people is contingent upon early life exposures affecting the islets, especially beta cells. Here, we review some of the recent advances and studies that highlight the roles of these changes as well as antigen presentation and stress response pathways in beta cells in the onset and propagation of the autoimmune process in type 1 diabetes. Future progress in this area holds promise for advancing islet- and beta cell-directed therapies that could be implemented in the early stages of the disease and could be combined with immunotherapies.


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Diabetes Mellitus, Type 1/metabolism , Insulin-Secreting Cells/metabolism , Autoimmunity/physiology , Islets of Langerhans/metabolism , Genetic Predisposition to Disease
4.
J Autoimmun ; 137: 102947, 2023 May.
Article in English | MEDLINE | ID: mdl-36357240

ABSTRACT

Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.


Subject(s)
Autoimmune Diseases , Autoimmunity , Humans , Aged , Autoimmunity/physiology , T-Lymphocytes , Aging , Cellular Senescence , Risk Factors
5.
Proc Natl Acad Sci U S A ; 117(3): 1799-1805, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31852823

ABSTRACT

Heterotrimeric G proteins are important transducers of receptor signaling, functioning in plants with CLAVATA receptors in controlling shoot meristem size and with pathogen-associated molecular pattern receptors in basal immunity. However, whether specific members of the heterotrimeric complex potentiate cross-talk between development and defense, and the extent to which these functions are conserved across species, have not yet been addressed. Here we used CRISPR/Cas9 to knock out the maize G protein ß subunit gene (Gß) and found that the mutants are lethal, differing from those in Arabidopsis, in which homologous mutants have normal growth and fertility. We show that lethality is caused not by a specific developmental arrest, but by autoimmunity. We used a genetic diversity screen to suppress the lethal Gß phenotype and also identified a maize Gß allele with weak autoimmune responses but strong development phenotypes. Using these tools, we show that Gß controls meristem size in maize, acting epistatically with G protein α subunit gene (Gα), suggesting that Gß and Gα function in a common signaling complex. Furthermore, we used an association study to show that natural variation in Gß influences maize kernel row number, an important agronomic trait. Our results demonstrate the dual role of Gß in immunity and development in a cereal crop and suggest that it functions in cross-talk between these competing signaling networks. Therefore, modification of Gß has the potential to optimize the trade-off between growth and defense signaling to improve agronomic production.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , Meristem/growth & development , Plant Immunity/physiology , Plant Shoots/growth & development , Zea mays/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Autoimmunity/physiology , CRISPR-Cas Systems , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/genetics , Gene Knockout Techniques , Meristem/cytology , Meristem/immunology , Phenotype , Plant Shoots/cytology , Plant Shoots/immunology , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Signal Transduction , Transcriptome
6.
Plant J ; 105(4): 994-1009, 2021 02.
Article in English | MEDLINE | ID: mdl-33210758

ABSTRACT

IMPORTIN-α3/MOS6 (MODIFIER OF SNC1, 6) is one of nine importin-α isoforms in Arabidopsis that recruit nuclear localization signal-containing cargo proteins to the nuclear import machinery. IMP-α3/MOS6 is required genetically for full autoimmunity of the nucleotide-binding leucine-rich repeat immune receptor mutant snc1 (suppressor of npr1-1, constitutive 1) and MOS6 also contributes to basal disease resistance. Here, we investigated the contribution of the other importin-α genes to both types of immune responses, and we analyzed potential interactions of all importin-α isoforms with SNC1. By using reverse-genetic analyses in Arabidopsis and protein-protein interaction assays in Nicotiana benthamiana, we provide evidence that among the nine α-importins in Arabidopsis, IMP-α3/MOS6 is the main nuclear transport receptor of SNC1, and that IMP-α3/MOS6 is required selectively for autoimmunity of snc1 and basal resistance to mildly virulent Pseudomonas syringae in Arabidopsis.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/immunology , Disease Resistance/physiology , Karyopherins/physiology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Autoimmunity/physiology , Karyopherins/metabolism , Phylogeny , Plant Diseases/immunology , Plant Diseases/microbiology , Pseudomonas syringae
7.
Brain ; 144(10): 3126-3141, 2021 11 29.
Article in English | MEDLINE | ID: mdl-34849598

ABSTRACT

Dimethyl fumarate, an approved treatment for relapsing-remitting multiple sclerosis, exerts pleiotropic effects on immune cells as well as CNS resident cells. Here, we show that dimethyl fumarate exerts a profound alteration of the metabolic profile of human CD4+ as well as CD8+ T cells and restricts their antioxidative capacities by decreasing intracellular levels of the reactive oxygen species scavenger glutathione. This causes an increase in mitochondrial reactive oxygen species levels accompanied by an enhanced mitochondrial stress response, ultimately leading to impaired mitochondrial function. Enhanced mitochondrial reactive oxygen species levels not only result in enhanced T-cell apoptosis in vitro as well as in dimethyl fumarate-treated patients, but are key for the well-known immunomodulatory effects of dimethyl fumarate both in vitro and in an animal model of multiple sclerosis, i.e. experimental autoimmune encephalomyelitis. Indeed, dimethyl fumarate immune-modulatory effects on T cells were completely abrogated by pharmacological interference of mitochondrial reactive oxygen species production. These data shed new light on dimethyl fumarate as bona fide immune-metabolic drug that targets the intracellular stress response in activated T cells, thereby restricting mitochondrial function and energetic capacity, providing novel insight into the role of oxidative stress in modulating cellular immune responses and T cell-mediated autoimmunity.


Subject(s)
Antioxidants/pharmacology , Autoimmunity/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dimethyl Fumarate/pharmacology , Immunosuppressive Agents/pharmacology , Adult , Animals , Antioxidants/therapeutic use , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cohort Studies , Dimethyl Fumarate/therapeutic use , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Young Adult
8.
Proc Natl Acad Sci U S A ; 116(51): 25860-25869, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31796589

ABSTRACT

Environmental triggers acting at the intestinal barrier are thought to contribute to the initiation of autoimmune disorders. The transforming growth factor beta inhibitor Smad7 determines the phenotype of CD4+ T cells. We hypothesized that Smad7 in intestinal CD4+ T cells controls initiation of opticospinal encephalomyelitis (OSE), a murine model of multiple sclerosis (MS), depending on the presence of gut microbiota. Smad7 was overexpressed or deleted in OSE CD4+ T cells to determine the effect on clinical progression, T cell differentiation, and T cell migration from the intestine to the central nervous system (CNS). Smad7 overexpression worsened the clinical course of OSE and increased CNS inflammation and demyelination. It favored expansion of intestinal CD4+ T cells toward an inflammatory phenotype and migration of intestinal CD4+ T cells to the CNS. Intestinal biopsies from MS patients revealed decreased transforming growth factor beta signaling with a shift toward inflammatory T cell subtypes. Smad7 in intestinal T cells might represent a valuable therapeutic target for MS to achieve immunologic tolerance in the intestine and suppress CNS inflammation.


Subject(s)
Autoimmunity/physiology , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/metabolism , Multiple Sclerosis/metabolism , Smad7 Protein/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Encephalomyelitis/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gastrointestinal Microbiome/physiology , Gene Expression Regulation , Humans , Immune Tolerance , Inflammation , Intestines/pathology , Mice , Mice, Transgenic , Multiple Sclerosis/pathology , Signal Transduction , Smad7 Protein/genetics , Spinal Cord/pathology , Transforming Growth Factor beta/metabolism
9.
Proc Natl Acad Sci U S A ; 116(51): 25839-25849, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776254

ABSTRACT

Naive CD4+ T lymphocytes differentiate into different effector types, including helper and regulatory cells (Th and Treg, respectively). Heritable gene expression programs that define these effector types are established during differentiation, but little is known about the epigenetic mechanisms that install and maintain these programs. Here, we use mice defective for different components of heterochromatin-dependent gene silencing to investigate the epigenetic control of CD4+ T cell plasticity. We show that, upon T cell receptor (TCR) engagement, naive and regulatory T cells defective for TRIM28 (an epigenetic adaptor for histone binding modules) or for heterochromatin protein 1 ß and γ isoforms (HP1ß/γ, 2 histone-binding factors involved in gene silencing) fail to effectively signal through the PI3K-AKT-mTOR axis and switch to glycolysis. While differentiation of naive TRIM28-/- T cells into cytokine-producing effector T cells is impaired, resulting in reduced induction of autoimmune colitis, TRIM28-/- regulatory T cells also fail to expand in vivo and to suppress autoimmunity effectively. Using a combination of transcriptome and chromatin immunoprecipitation-sequencing (ChIP-seq) analyses for H3K9me3, H3K9Ac, and RNA polymerase II, we show that reduced effector differentiation correlates with impaired transcriptional silencing at distal regulatory regions of a defined set of Treg-associated genes, including, for example, NRP1 or Snai3. We conclude that TRIM28 and HP1ß/γ control metabolic reprograming through epigenetic silencing of a defined set of Treg-characteristic genes, thus allowing effective T cell expansion and differentiation into helper and regulatory phenotypes.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic/physiology , T-Lymphocytes/metabolism , Tripartite Motif-Containing Protein 28/metabolism , Animals , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Plasticity/physiology , Cellular Reprogramming/genetics , Chromobox Protein Homolog 5 , Colon/pathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Gene Silencing , Histones/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcriptome , Tripartite Motif-Containing Protein 28/genetics
10.
Proc Natl Acad Sci U S A ; 116(23): 11380-11389, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31097594

ABSTRACT

LPS-responsive beige-like anchor (LRBA) protein deficiency in humans causes immune dysregulation resulting in autoimmunity, inflammatory bowel disease (IBD), hypogammaglobulinemia, regulatory T (Treg) cell defects, and B cell functional defects, but the cellular and molecular mechanisms responsible are incompletely understood. In an ongoing forward genetic screen for N-ethyl-N-nitrosourea (ENU)-induced mutations that increase susceptibility to dextran sodium sulfate (DSS)-induced colitis in mice, we identified two nonsense mutations in Lrba Although Treg cells have been a main focus in LRBA research to date, we found that dendritic cells (DCs) contribute significantly to DSS-induced intestinal inflammation in LRBA-deficient mice. Lrba-/- DCs exhibited excessive IRF3/7- and PI3K/mTORC1-dependent signaling and type I IFN production in response to the stimulation of the Toll-like receptors (TLRs) 3, TLR7, and TLR9. Substantial reductions in cytokine expression and sensitivity to DSS in LRBA-deficient mice were caused by knockout of Unc93b1, a chaperone necessary for trafficking of TLR3, TLR7, and TLR9 to endosomes. Our data support a function for LRBA in limiting endosomal TLR signaling and consequent intestinal inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colitis/metabolism , Endosomes/metabolism , Signal Transduction/physiology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmunity/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Colitis/chemically induced , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dextran Sulfate/pharmacology , Female , Inflammation/metabolism , Male , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects
11.
Diabetologia ; 64(7): 1604-1612, 2021 07.
Article in English | MEDLINE | ID: mdl-33783586

ABSTRACT

AIMS/HYPOTHESIS: We aimed to investigate the association between maternal consumption of gluten-containing foods and other selected foods during late pregnancy and offspring risk of islet autoimmunity (IA) and type 1 diabetes in The Environmental Determinants of Diabetes in the Young (TEDDY) study. METHODS: The TEDDY study recruited children at high genetic risk for type 1 diabetes at birth, and prospectively follows them for the development of IA and type 1 diabetes (n = 8556). A questionnaire on the mother's diet in late pregnancy was completed by 3-4 months postpartum. The maternal daily intake was estimated from a food frequency questionnaire for eight food groups: gluten-containing foods, non-gluten cereals, fresh milk, sour milk, cheese products, soy products, lean/medium-fat fish and fatty fish. For each food, we described the distribution of maternal intake among the four participating countries in the TEDDY study and tested the association of tertile of maternal food consumption with risk of IA and type 1 diabetes using forward selection time-to-event Cox regression. RESULTS: By 28 February 2019, 791 cases of IA and 328 cases of type 1 diabetes developed in TEDDY. There was no association between maternal late-pregnancy consumption of gluten-containing foods or any of the other selected foods and risk of IA, type 1 diabetes, insulin autoantibody-first IA or GAD autoantibody-first IA (all p ≥ 0.01). Maternal gluten-containing food consumption in late pregnancy was higher in Sweden (242 g/day), Germany (247 g/day) and Finland (221 g/day) than in the USA (199 g/day) (pairwise p < 0.05). CONCLUSIONS/INTERPRETATION: Maternal food consumption during late pregnancy was not associated with offspring risk for IA or type 1 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT00279318.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/etiology , Islets of Langerhans/immunology , Maternal Nutritional Physiological Phenomena/physiology , Adult , Autoantibodies/analysis , Autoantibodies/blood , Autoimmunity/physiology , Breast Feeding , Diet , Diet Surveys , Eating/physiology , Female , Glutens/administration & dosage , Glutens/adverse effects , Humans , Infant, Newborn , Male , Postpartum Period , Pregnancy , Pregnancy Trimester, Third/blood , Pregnancy Trimester, Third/physiology , Risk Factors
12.
Diabetologia ; 64(7): 1626-1641, 2021 07.
Article in English | MEDLINE | ID: mdl-33912981

ABSTRACT

AIMS/HYPOTHESIS: We and others previously reported the presence of tertiary lymphoid organs (TLOs) in the pancreas of NOD mice, where they play a role in the development of type 1 diabetes. Our aims here are to investigate whether TLOs are present in the pancreas of individuals with type 1 diabetes and to characterise their distinctive features, in comparison with TLOs present in NOD mouse pancreases, in order to interpret their functional significance. METHODS: Using immunofluorescence confocal microscopy, we examined the extracellular matrix (ECM) and cellular constituents of pancreatic TLOs from individuals with ongoing islet autoimmunity in three distinct clinical settings of type 1 diabetes: at risk of diabetes; at/after diagnosis; and in the transplanted pancreas with recurrent diabetes. Comparisons were made with TLOs from 14-week-old NOD mice, which contain islets exhibiting mild to heavy leucocyte infiltration. We determined the frequency of the TLOs in human type 1diabetes with insulitis and investigated the presence of TLOs in relation to age of onset, disease duration and disease severity. RESULTS: TLOs were identified in preclinical and clinical settings of human type 1 diabetes. The main characteristics of these TLOs, including the cellular and ECM composition of reticular fibres (RFs), the presence of high endothelial venules and immune cell subtypes detected, were similar to those observed for TLOs from NOD mouse pancreases. Among 21 donors with clinical type 1 diabetes who exhibited insulitis, 12 had TLOs and had developed disease at younger age compared with those lacking TLOs. Compartmentalised TLOs with distinct T cell and B cell zones were detected in donors with short disease duration. Overall, TLOs were mainly associated with insulin-containing islets and their frequency decreased with increasing severity of beta cell loss. Parallel studies in NOD mice further revealed some differences in so far as regulatory T cells were essentially absent from human pancreatic TLOs and CCL21 was not associated with RFs. CONCLUSIONS/INTERPRETATION: We demonstrate a novel feature of pancreas pathology in type 1 diabetes. TLOs represent a potential site of autoreactive effector T cell generation in islet autoimmunity and our data from mouse and human tissues suggest that they disappear once the destructive process has run its course. Thus, TLOs may be important for type 1 diabetes progression.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Tertiary Lymphoid Structures/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Autoantibodies/analysis , Autoantibodies/blood , Autoimmunity/physiology , Child , Child, Preschool , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Female , Humans , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred NOD , Microscopy, Fluorescence , Middle Aged , Pancreas/pathology , Tertiary Lymphoid Structures/blood , Tertiary Lymphoid Structures/immunology , Young Adult
13.
Circulation ; 141(15): 1238-1248, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32114801

ABSTRACT

BACKGROUND: Serum anti-heart autoantibodies (AHAs) and anti-intercalated disk autoantibodies (AIDAs) are autoimmune markers in myocarditis. Myocarditis has been reported in arrhythmogenic right ventricular cardiomyopathy (ARVC). To provide evidence for autoimmunity, we searched for AHAs and AIDAs in ARVC. METHODS: We studied: 42 ARVC probands, 23 male, aged 42, interquartile range 33-49, 20 from familial and 22 nonfamilial pedigrees; 37 clinically affected relatives (ARs), 24 male aged 35, interquartile range 18-46; and 96 healthy relatives, 49 male, aged 27, interquartile range 17-45. Serum AHAs and AIDAs were tested by indirect immunofluorescence on human myocardium and skeletal muscle in 171 of the 175 ARVC individuals and in controls with noninflammatory cardiac disease (n=160), ischemic heart failure (n=141), and healthy blood donors (n=270). Screening of 5 desmosomal genes was performed in probands; when a sequence variant was identified, cascade family screening followed, blind to immunologic results. RESULTS: AHA frequency was higher (36.8%) in probands, ARs (37.8%), and healthy relatives (25%) than in noninflammatory cardiac disease (1%), ischemic heart failure (1%), or healthy blood donors (2.5%; P=0.0001). AIDA frequency was higher in probands (8%, P=0.006), in ARs (21.6%, P=0.00001), and in healthy relatives (14.6%, P=0.00001) than in noninflammatory cardiac disease (3.75%), ischemic heart failure (2%), or healthy blood donors (0.3%). AHA-positive status was associated with higher frequency of palpitation (P=0.004), implantable cardioverter defibrillator implantation (P=0.021), lower left ventricular ejection fraction (P=0.004), AIDA-positive status with both lower right ventricular and left ventricular ejection fractions (P=0.027 and P=0.027, respectively). AHA- and/or AIDA-positive status in the proband and at least one of the respective relatives was more common in familial (17/20, 85%) than in sporadic (10/22, 45%) pedigrees (P=0.007). CONCLUSIONS: The presence of AHAs and AIDAs provides evidence of autoimmunity in the majority of familial and in almost half of sporadic ARVC. In probands and in ARs, these antibodies were associated with features of disease severity. Longitudinal studies are needed to clarify whether they may predict ARVC development in healthy relatives or if they be a result of manifest ARVC.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia/physiopathology , Autoantibodies/genetics , Autoimmunity/physiology , Cardiomyopathies/physiopathology , Genetic Testing/methods , Medical History Taking/methods , Adult , Female , Humans , Male , Middle Aged
14.
Mol Med ; 27(1): 160, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930107

ABSTRACT

COVID-19 clinical presentation differs considerably between individuals, ranging from asymptomatic, mild/moderate and severe disease which in some cases are fatal or result in long-term effects. Identifying immune mechanisms behind severe disease development informs screening strategies to predict who are at greater risk of developing life-threatening complications. However, to date clear prognostic indicators of individual risk of severe or long COVID remain elusive. Autoantibodies recognize a range of self-antigens and upon antigen recognition and binding, important processes involved in inflammation, pathogen defence and coagulation are modified. Recent studies report a significantly higher prevalence of autoantibodies that target immunomodulatory proteins including cytokines, chemokines, complement components, and cell surface proteins in COVID-19 patients experiencing severe disease compared to those who experience mild or asymptomatic infections. Here we discuss the diverse impacts of autoantibodies on immune processes and associations with severe COVID-19 disease.


Subject(s)
Autoantibodies/immunology , Autoantibodies/metabolism , COVID-19/complications , COVID-19/immunology , SARS-CoV-2/immunology , Animals , Autoimmunity/physiology , COVID-19/metabolism , Humans , SARS-CoV-2/metabolism , Post-Acute COVID-19 Syndrome
15.
Am J Gastroenterol ; 116(3): 593-599, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33560653

ABSTRACT

INTRODUCTION: Untreated symptomatic celiac disease (CD) adversely affects female reproduction; however, the effect of hidden CD autoimmunity is uncertain. METHODS: We identified women who were not previously diagnosed with CD and tested positive for tissue transglutaminase and endomysial antibodies between 2006 and 2011 in a community-based retrospective cohort study. We evaluated (i) the rate of adverse pregnancy outcomes and medical complications of pregnancy in successful singleton deliveries and (ii) reproductive characteristics in seropositive women without a clinical diagnosis of CD and age-matched seronegative women. RESULTS: Among 17,888 women whose serum samples were tested for CD autoimmunity, 215 seropositive and 415 seronegative women were included. We reviewed 231 and 509 live singleton deliveries of 117 seropositive and 250 seronegative mothers, respectively. Menarche and menopausal age, gravidity, parity, and age at first child were similar in seropositive and seronegative women. CD seropositivity was not associated with an increased risk of maternal pregnancy complications. Maternal seropositivity was associated with small for gestational age in boys (OR 3.77, 95% CI: 1.47-9.71; P = 0.006), but not in girls (OR 0.57, 95% CI: 0.15-2.17; P = 0.41). CD serum positivity was not associated with prematurity, small for gestational age (birth weight <10th percentile), or 5-minute Apgar score of less than 7. DISCUSSION: Although underpowered, the present study did not show any difference in reproductive characteristics or rates of adverse pregnancy outcomes in women with and without CD autoimmunity, except for birth weight in male offspring. Larger studies are needed to determine the effects of CD autoimmunity on female reproduction.


Subject(s)
Autoimmunity/physiology , Celiac Disease/immunology , Pregnancy Complications/immunology , Pregnancy Outcome , Adult , Autoantibodies/immunology , Birth Weight , Celiac Disease/diagnosis , Female , Humans , Infant, Newborn , Parity/immunology , Pregnancy , Pregnancy Complications/diagnosis , Retrospective Studies , Young Adult
16.
J Neuroinflammation ; 18(1): 292, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34920747

ABSTRACT

BACKGROUND: Recent evidence has shown dopamine as a major regulator of inflammation. Accordingly, dopaminergic regulation of immune cells plays an important role in the physiopathology of inflammatory disorders. Multiple sclerosis (MS) is an inflammatory disease involving a CD4+ T-cell-driven autoimmune response to central nervous system (CNS) derived antigens. Evidence from animal models has suggested that B cells play a fundamental role as antigen-presenting cells (APC) re-stimulating CD4+ T cells in the CNS as well as regulating T-cell response by mean of inflammatory or anti-inflammatory cytokines. Here, we addressed the role of the dopamine receptor D3 (DRD3), which displays the highest affinity for dopamine, in B cells in animal models of MS. METHODS: Mice harbouring Drd3-deficient or Drd3-sufficient B cells were generated by bone marrow transplantation into recipient mice devoid of B cells. In these mice, we compared the development of experimental autoimmune encephalomyelitis (EAE) induced by immunization with a myelin oligodendrocyte glycoprotein (MOG)-derived peptide (pMOG), a model that leads to CNS-autoimmunity irrespective of the APC-function of B cells, or by immunization with full-length human MOG protein (huMOG), a model in which antigen-specific activated B cells display a fundamental APC-function in the CNS. APC-function was assessed in vitro by pulsing B cells with huMOG-coated beads and then co-culturing with MOG-specific T cells. RESULTS: Our data show that the selective Drd3 deficiency in B cells abolishes the disease development in the huMOG-induced EAE model. Mechanistic analysis indicates that although DRD3-signalling did not affect the APC-function of B cells, DRD3 favours the CNS-tropism in a subset of pro-inflammatory B cells in the huMOG-induced EAE model, an effect that was associated with higher CXCR3 expression. Conversely, the results show that the selective Drd3 deficiency in B cells exacerbates the disease severity in the pMOG-induced EAE model. Further analysis shows that DRD3-stimulation increased the expression of the CNS-homing molecule CD49d in a B-cell subset with anti-inflammatory features, thus attenuating EAE manifestation in the pMOG-induced EAE model. CONCLUSIONS: Our findings demonstrate that DRD3 in B cells exerts a dual role in CNS-autoimmunity, favouring CNS-tropism of pro-inflammatory B cells with APC-function and promoting CNS-homing of B cells with anti-inflammatory features. Thus, these results show DRD3-signalling in B cells as a critical regulator of CNS-autoimmunity.


Subject(s)
Autoimmunity/physiology , B-Lymphocytes/metabolism , Dopamine/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Receptors, Dopamine D3/metabolism , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/metabolism , Dopamine/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Dopamine D3/genetics , Receptors, Dopamine D3/immunology
17.
FASEB J ; 34(3): 4329-4347, 2020 03.
Article in English | MEDLINE | ID: mdl-31971297

ABSTRACT

Plasmacytoid dendritic cells (pDCs) express Toll like receptors (TLRs) that modulate the immune response by production of type I interferons. Here, we report that sphingosine kinase 1 (SphK1) which produces the bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P), plays a critical role in the pDC functions and interferon production. Although dispensable for the pDC development, SphK1 is essential for the pDC activation and production of type I IFN and pro-inflammatory cytokines stimulated by TLR7/9 ligands. SphK1 interacts with TLRs and specific inhibition or deletion of SphK1 in pDCs mitigates uptake of CpG oligonucleotide ligands by TLR9 ligand. In the pristane-induced murine lupus model, pharmacological inhibition of SphK1 or its genetic deletion markedly decreased the IFN signature, pDC activation, and glomerulonephritis. Moreover, increases in the SphK1 expression and S1P levels were observed in human lupus patients. Taken together, our results indicate a pivotal regulatory role for the SphK1/S1P axis in maintaining the balance between immunosurveillance and immunopathology and suggest that specific SphK1 inhibitors might be a new therapeutic avenue for the treatment of type I IFN-linked autoimmune disorders.


Subject(s)
Autoimmunity/physiology , Interferon Type I/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmunity/genetics , Blotting, Western , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Mice , Mice, Inbred C57BL , Models, Biological , Toll-Like Receptor 7/genetics , Toll-Like Receptor 9/genetics
18.
Pediatr Diabetes ; 22(4): 577-585, 2021 06.
Article in English | MEDLINE | ID: mdl-33543815

ABSTRACT

BACKGROUND: Circulating fatty acids have been linked to development of type 1 diabetes. OBJECTIVES: To study the prospective associations of serum fatty acids with the risk of islet autoimmunity in high-risk children. METHODS: A nested case-control selection was carried out within the TRIGR cohort, which included infants with HLA (DQB1 or DQA1)-conferred disease susceptibility and a first-degree relative with type 1 diabetes, born between 2002 and 2007 in 15 countries and followed-up until 2017. The present study included 244 case children positive for at least two islet autoantibodies (ICA, IAA, GADA, and IA-2A) and two control children were matched for country and age. Proportions of 26 serum fatty acids at cord blood and at 6, 12, and 18 months of age were assessed using gas-chromatography. RESULTS: The average proportions of the following fatty acids were associated with an increased risk of islet autoimmunity, adjusted for sex, HLA risk, and maternal type 1 diabetes: pentadecanoic acid (15:0) (OR 3.41: 95% CI 1.70, 6.85), heptadecanoic acid (iso 17:0) (2.64: 1.62, 4.28) and (anteiso 17:0) (2.27: 1.39, 3.70), stearic acid (18:0) (23.8: 2.32, 244.6), and conjugated linoleic acid (18:2n-7) (2.60: 1.47, 4.59). Breastfeeding and not having maternal type 1 diabetes were positively associated with levels of the above-mentioned fatty acids. N-3 fatty acids were not consistently associated with islet autoimmunity. CONCLUSIONS: We found direct associations of pentadecanoic acid, heptadecanoic acid, stearic acid, and conjugated linoleic acid with the risk of islet autoimmunity. Further studies are needed to understand the complex role of fatty acids in the development of type 1 diabetes.


Subject(s)
Autoantibodies/blood , Autoimmunity/physiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Fatty Acids/blood , Islets of Langerhans/immunology , Age Factors , Birth Cohort , Case-Control Studies , Child , Child, Preschool , Female , Humans , Infant , Male
19.
Pediatr Diabetes ; 22(3): 425-433, 2021 05.
Article in English | MEDLINE | ID: mdl-33470492

ABSTRACT

AIM: We aimed to characterize associations between diet and the gut microbiome and short chain fatty acid (SCFA) products in youth with islet autoimmunity or type 1 diabetes (IA/T1D) in comparison with controls. RESEARCH DESIGN AND METHODS: Eighty participants (25 diagnosed with T1D, 17 with confirmed IA, 38 sibling or unrelated controls) from the Australian T1D Gut Study cohort were studied (median [IQR] age 11.7 [8.9, 14.0] years, 43% female). A Food Frequency Questionnaire characterized daily macronutrient intake over the preceding 6 months. Plasma and fecal SCFA were measured by gas chromatography; gut microbiome composition and diversity by 16S rRNA gene sequencing. RESULTS: A 10 g increase in daily carbohydrate intake associated with higher plasma acetate in IA/T1D (adjusted estimate +5.2 (95% CI 1.1, 9.2) µmol/L p = 0.01) and controls (adjusted estimate +4.1 [95% CI 1.7, 8.5] µmol/L p = 0.04). A 5 g increase in total fat intake associated with lower plasma acetate in IA/T1D and controls. A 5% increase in noncore (junk) food intake associated with reduced richness (adjusted estimate -4.09 [95%CI -7.83, -0.35] p = .03) and evenness (-1.25 [95% CI -2.00, -0.49] p < 0.01) of the gut microbiome in IA/T1D. Fiber intake associated with community structure of the microbiome in IA/T1D. CONCLUSIONS: Modest increments in carbohydrate and fat intake associated with plasma acetate in all youth. Increased junk food intake associated with reduced diversity of the gut microbiome in IA/T1D alone. These associations with the gut microbiome in IA/T1D support future efforts to promote SCFA by using dietary interventions.


Subject(s)
Autoimmunity/physiology , Diabetes Mellitus, Type 1/metabolism , Diet , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Islets of Langerhans/immunology , Adolescent , Case-Control Studies , Child , Cross-Sectional Studies , Diabetes Mellitus, Type 1/complications , Female , Humans , Male , Surveys and Questionnaires
20.
BMC Endocr Disord ; 21(1): 66, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33849519

ABSTRACT

BACKGROUND: Omega-3 polyunsaturated fatty acids (PUFAs) produce lipid mediators with both anti-inflammatory and pro-resolution properties, including resolvins. The purpose of this study was to detect serum resolvin E1 (RVE1) levels in Hashimoto's thyroiditis (HT) patients and healthy controls (HCs) and to evaluate the relationship of RVE1 with thyroid autoimmunity. METHODS: A total of 57 participants were recruited, including 30 untreated HT patients and 27 age- and sex-matched HCs. The levels of RVE1 in serum were measured via enzyme-linked immunosorbent assay (ELISA). An electrochemiluminescence immunoassay was used for the measurement of thyroid-stimulating hormone (TSH), total T4 (TT4), TT3, free T4 (FT4), FT3, anti-thyroid peroxidase antibody (TPOAb) and anti-thyroglobulin antibody (TgAb) levels. Hemogram tests and routine biochemical analyses were performed on each sample. RESULTS: The serum level of RVE1 of HT patients (24.09, 15.76-34.38 pg/mL) was significantly lower than that of healthy controls (28.51, 20.76-51.23 pg/mL) (P = 0.027). RVE1 levels showed a downward trend with increasing TgAb levels (P for trend = 0.001). Multivariable ordinal logistic regression analysis showed that RVE1 levels were negatively correlated with increasing TgAb levels in both the unadjusted (OR = 0.9446, 95 % CI = 0.9111-0.9782, P = 0.002) and adjusted models (OR = 0.9380, 95 % CI = 0.8967-0.9811, P = 0.005). CONCLUSIONS: Decreased RVE1 levels might be a sign that HT is associated with inflammatory resolution dysfunction. RVE1 may serve as a protective factor against increased TgAb levels.


Subject(s)
Autoimmunity/physiology , Eicosapentaenoic Acid/analogs & derivatives , Hashimoto Disease/blood , Hashimoto Disease/immunology , Thyroid Gland/immunology , Adult , Aged , Biomarkers/blood , Cross-Sectional Studies , Eicosapentaenoic Acid/blood , Female , Hashimoto Disease/diagnosis , Humans , Male , Middle Aged , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL