Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.894
Filtrer
1.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-39062856

RÉSUMÉ

The 3 Screen ICA ELISA is a novel assay capable of simultaneously measuring autoantibodies to glutamic acid decarboxylase (GADA), insulinoma-associated antigen-2 (IA-2A), and zinc transporter 8 (ZnT8A), making it a valuable tool for screening type 1 diabetes. Despite its advantages, it cannot specify which individual autoantibodies are positive or negative. This study aimed to estimate individual positive autoantibodies based on the 3 Screen ICA titer. Six hundred seventeen patients with type 1 diabetes, simultaneously measured for 3 Screen ICA and three individual autoantibodies, were divided into five groups based on their 3 Screen ICA titer. The sensitivities and contribution rates of the individual autoantibodies were then examined. The study had a cross-sectional design. Sixty-nine percent (424 of 617) of patients with type 1 diabetes had 3 Screen ICA titers exceeding the 99th percentile cut-off level (20 index). The prevalence of GADA ranged from 80% to 100% in patients with a 3 Screen ICA over 30 index and 97% of patients with a 3 Screen ICA ≥300 index. Furthermore, the prevalence of all individual autoantibodies being positive was 0% for ≤80 index and as high as 92% for ≥300 index. Significant associations were observed in specific titer groups: the 20-29.9 index group when all the individual autoantibodies were negative, the 30-79.9 index group when positive for GADA alone or IA-2A alone, the 30-299.9 index group when positive for ZnT8A alone, the 80-299.9 index group when positive for both IA-2A and ZnT8A, the 300-499.9 index group when positive for both GADA and ZnT8A, and the ≥300 index group when positive for all individual autoantibodies. These results suggest that the 3 Screen ICA titer may be helpful in estimating individual positive autoantibodies.


Sujet(s)
Autoanticorps , Diabète de type 1 , Glutamate decarboxylase , Transporteur de zinc ZnT-8 , Humains , Autoanticorps/sang , Autoanticorps/immunologie , Mâle , Femelle , Diabète de type 1/immunologie , Diabète de type 1/sang , Diabète de type 1/diagnostic , Adulte , Transporteur de zinc ZnT-8/immunologie , Glutamate decarboxylase/immunologie , Études transversales , Adolescent , Adulte d'âge moyen , Test ELISA/méthodes , Ilots pancréatiques/immunologie , Jeune adulte , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunologie , Enfant
2.
Front Immunol ; 15: 1415102, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007132

RÉSUMÉ

Human regulatory T cells (Treg) suppress other immune cells. Their dysfunction contributes to the pathophysiology of autoimmune diseases, including type 1 diabetes (T1D). Infusion of Tregs is being clinically evaluated as a novel way to prevent or treat T1D. Genetic modification of Tregs, most notably through the introduction of a chimeric antigen receptor (CAR) targeting Tregs to pancreatic islets, may improve their efficacy. We evaluated CAR targeting of human Tregs to monocytes, a human ß cell line and human islet ß cells in vitro. Targeting of HLA-A2-CAR (A2-CAR) bulk Tregs to HLA-A2+ cells resulted in dichotomous cytotoxic killing of human monocytes and islet ß cells. In exploring subsets and mechanisms that may explain this pattern, we found that CD39 expression segregated CAR Treg cytotoxicity. CAR Tregs from individuals with more CD39low/- Tregs and from individuals with genetic polymorphism associated with lower CD39 expression (rs10748643) had more cytotoxicity. Isolated CD39- CAR Tregs had elevated granzyme B expression and cytotoxicity compared to the CD39+ CAR Treg subset. Genetic overexpression of CD39 in CD39low CAR Tregs reduced their cytotoxicity. Importantly, ß cells upregulated protein surface expression of PD-L1 and PD-L2 in response to A2-CAR Tregs. Blockade of PD-L1/PD-L2 increased ß cell death in A2-CAR Treg co-cultures suggesting that the PD-1/PD-L1 pathway is important in protecting islet ß cells in the setting of CAR immunotherapy. In summary, introduction of CAR can enhance biological differences in subsets of Tregs. CD39+ Tregs represent a safer choice for CAR Treg therapies targeting tissues for tolerance induction.


Sujet(s)
Apyrase , Récepteurs chimériques pour l'antigène , Lymphocytes T régulateurs , Humains , Apyrase/immunologie , Apyrase/métabolisme , Lymphocytes T régulateurs/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/métabolisme , Cytotoxicité immunologique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Diabète de type 1/immunologie , Diabète de type 1/thérapie , Antigène HLA-A2/immunologie , Antigène HLA-A2/génétique , Antigène HLA-A2/métabolisme , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Antigènes CD
3.
BMJ Open Diabetes Res Care ; 12(4)2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39013632

RÉSUMÉ

INTRODUCTION: The Environmental Determinants of Islet Autoimmunity (ENDIA) Study is an ongoing Australian prospective cohort study investigating how modifiable prenatal and early-life exposures drive the development of islet autoimmunity and type 1 diabetes (T1D) in children. In this profile, we describe the cohort's parental demographics, maternal and neonatal outcomes and human leukocyte antigen (HLA) genotypes. RESEARCH DESIGN AND METHODS: Inclusion criteria were an unborn child, or infant aged less than 6 months, with a first-degree relative (FDR) with T1D. The primary outcome was persistent islet autoimmunity, with children followed until a T1D diagnosis or 10 years of age. Demographic data were collected at enrollment. Lifestyle, clinical and anthropometric data were collected at each visit during pregnancy and clinical pregnancy and birth data were verified against medical case notes. Data were compared between mothers with and without T1D. HLA genotyping was performed on the ENDIA child and all available FDRs. RESULTS: The final cohort comprised 1473 infants born to 1214 gestational mothers across 1453 pregnancies, with 80% enrolled during pregnancy. The distribution of familial T1D probands was 62% maternal, 28% paternal and 11% sibling. The frequency of high-risk HLA genotypes was highest in T1D probands, followed by ENDIA infants, and lowest among unaffected family members. Mothers with T1D had higher rates of pregnancy complications and perinatal intervention, and larger babies of shorter gestation. Parent demographics were comparable to the Australian population for age, parity and obesity. A greater percentage of ENDIA parents were Australian born, lived in a major city and had higher socioeconomic advantage and education. CONCLUSIONS: This comprehensive profile provides the context for understanding ENDIA's scope, methodology, unique strengths and limitations. Now fully recruited, ENDIA will provide unique insights into the roles of early-life factors in the development of islet autoimmunity and T1D in the Australian environment. TRIAL REGISTRATION NUMBER: ACTRN12613000794707.


Sujet(s)
Auto-immunité , Diabète de type 1 , Humains , Diabète de type 1/épidémiologie , Diabète de type 1/génétique , Diabète de type 1/immunologie , Diabète de type 1/étiologie , Femelle , Grossesse , Australie/épidémiologie , Études prospectives , Mâle , Enfant , Nourrisson , Nouveau-né , Facteurs de risque , Adulte , Ilots pancréatiques/immunologie , Études longitudinales , Études de suivi , Effets différés de l'exposition prénatale à des facteurs de risque/épidémiologie , Enfant d'âge préscolaire , Parents , Génotype , Antigènes HLA/génétique
4.
Immunity ; 57(7): 1448-1451, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38986438

RÉSUMÉ

Autoreactive lymphocytes that infiltrate the pancreatic islet environment and target ß cells are primary drivers of type 1 diabetes. In this issue of Immunity, Srivastava et al.1 examine the role of the islet microenvironment in autoimmunity and find that the scavenging receptor CXCL16 on islet-resident macrophages uptakes oxidized low-density lipoproteins and promotes the differentiation and survival of infiltrating pathogenic CD8+ T cells.


Sujet(s)
Auto-immunité , Lymphocytes T CD8+ , Diabète de type 1 , Ilots pancréatiques , Macrophages , Auto-immunité/immunologie , Diabète de type 1/immunologie , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Humains , Animaux , Macrophages/immunologie , Macrophages/métabolisme , Lymphocytes T CD8+/immunologie , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/immunologie
5.
Clin Nutr ESPEN ; 62: 22-27, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38901945

RÉSUMÉ

BACKGROUND & AIMS: Maternal gluten intake in relation to child's risk of type 1 diabetes has been studied in few prospective studies considering the diet during pregnancy but none during lactation. Our aim was to study whether gluten, cereals, or dietary fiber in maternal diet during pregnancy and lactation is associated with the risk of islet autoimmunity or type 1 diabetes in the offspring. METHODS: We included 4943 children with genetic susceptibility to type 1 diabetes from the Finnish Type 1 Diabetes Prediction and Prevention (DIPP) Study, born between 1996 and 2004. Maternal intake of gluten, different types of cereals, and dietary fiber were derived from a semi-quantitative validated food frequency questionnaire covering the eighth month of pregnancy and the third month of lactation. Children were monitored for islet autoantibodies up to age of 15 years and type 1 diabetes until year 2017. Risk of islet autoimmunity and clinical type 1 diabetes were estimated using Cox regression model, adjusted for energy intake, child's sex, HLA genotype, and familial diabetes. RESULTS: Altogether 312 children (6.4%) developed islet autoimmunity at median age of 3.5 (IQR 1.7, 6.6) years and 178 children (3.6%) developed type 1 diabetes at median age of 7.1 (IQR 4.3, 10.6) years. Gluten intake during pregnancy was not associated with islet autoimmunity (HR 0.96; 95% CI 0.68, 1.35), per 1 g/MJ increase in intake nor type 1 diabetes (HR 0.96; 95% CI 0.62, 1.50) in the offspring. Higher barley consumption during lactation was associated with increased risk of type 1 diabetes (HR 3.25; 95% CI 1.21, 8.70) per 1 g/MJ increase in intake. Maternal intake of other cereals or dietary fiber was not associated with the offspring outcomes. CONCLUSIONS: We observed no association between maternal intake of gluten, most consumed cereals, or dietary fiber during pregnancy or lactation and the risk of islet autoimmunity or type 1 diabetes in children from a high-risk population.


Sujet(s)
Auto-immunité , Diabète de type 1 , Fibre alimentaire , Grains comestibles , Glutens , Lactation , Humains , Diabète de type 1/immunologie , Femelle , Grossesse , Glutens/effets indésirables , Enfant , Enfant d'âge préscolaire , Mâle , Finlande , Nourrisson , Facteurs de risque , Régime alimentaire , Adolescent , Phénomènes physiologiques nutritionnels maternels , Études prospectives , Ilots pancréatiques/immunologie , Effets différés de l'exposition prénatale à des facteurs de risque , Adulte
6.
Front Immunol ; 15: 1345494, 2024.
Article de Anglais | MEDLINE | ID: mdl-38915393

RÉSUMÉ

Background: Type 1 diabetes (T1D) is preceded by a heterogenous pre-clinical phase, islet autoimmunity (IA). We aimed to identify pre vs. post-IA seroconversion (SV) changes in DNAm that differed across three IA progression phenotypes, those who lose autoantibodies (reverters), progress to clinical T1D (progressors), or maintain autoantibody levels (maintainers). Methods: This epigenome-wide association study (EWAS) included longitudinal DNAm measurements in blood (Illumina 450K and EPIC) from participants in Diabetes Autoimmunity Study in the Young (DAISY) who developed IA, one or more islet autoantibodies on at least two consecutive visits. We compared reverters - individuals who sero-reverted, negative for all autoantibodies on at least two consecutive visits and did not develop T1D (n=41); maintainers - continued to test positive for autoantibodies but did not develop T1D (n=60); progressors - developed clinical T1D (n=42). DNAm data were measured before (pre-SV visit) and after IA (post-SV visit). Linear mixed models were used to test for differences in pre- vs post-SV changes in DNAm across the three groups. Linear mixed models were also used to test for group differences in average DNAm. Cell proportions, age, and sex were adjusted for in all models. Median follow-up across all participants was 15.5 yrs. (interquartile range (IQR): 10.8-18.7). Results: The median age at the pre-SV visit was 2.2 yrs. (IQR: 0.8-5.3) in progressors, compared to 6.0 yrs. (IQR: 1.3-8.4) in reverters, and 5.7 yrs. (IQR: 1.4-9.7) in maintainers. Median time between the visits was similar in reverters 1.4 yrs. (IQR: 1-1.9), maintainers 1.3 yrs. (IQR: 1.0-2.0), and progressors 1.8 yrs. (IQR: 1.0-2.0). Changes in DNAm, pre- vs post-SV, differed across the groups at one site (cg16066195) and 11 regions. Average DNAm (mean of pre- and post-SV) differed across 22 regions. Conclusion: Differentially changing DNAm regions were located in genomic areas related to beta cell function, immune cell differentiation, and immune cell function.


Sujet(s)
Autoanticorps , Auto-immunité , Méthylation de l'ADN , Diabète de type 1 , Évolution de la maladie , Ilots pancréatiques , Humains , Diabète de type 1/immunologie , Diabète de type 1/génétique , Femelle , Mâle , Auto-immunité/génétique , Ilots pancréatiques/immunologie , Autoanticorps/sang , Autoanticorps/immunologie , Enfant , Adolescent , Études longitudinales , Enfant d'âge préscolaire , Étude d'association pangénomique , Épigenèse génétique
8.
Diabetes Care ; 47(8): 1276-1298, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38912694

RÉSUMÉ

Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programs are being increasingly emphasized. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk for (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in nonspecialized settings. To inform this monitoring, JDRF, in conjunction with international experts and societies, developed consensus guidance. Broad advice from this guidance includes the following: 1) partnerships should be fostered between endocrinologists and primary care providers to care for people who are IAb+; 2) when people who are IAb+ are initially identified, there is a need for confirmation using a second sample; 3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; 4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; 5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and 6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasizes significant unmet needs for further research on early-stage type 1 diabetes to increase the rigor of future recommendations and inform clinical care.


Sujet(s)
Autoanticorps , Diabète de type 1 , Diabète de type 1/immunologie , Diabète de type 1/diagnostic , Humains , Autoanticorps/sang , Autoanticorps/immunologie , Consensus , Ilots pancréatiques/immunologie
9.
J Clin Invest ; 134(14)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885342

RÉSUMÉ

While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects ß cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and ß cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1ß production. This aberrant pericyte-orchestrated islet inflammation was associated with ß cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1ß production. Treatment with either Cxcl1 or IL-1ß restored the mature ß cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.


Sujet(s)
Inflammation , Interleukine-1 bêta , Facteur de différenciation myéloïde-88 , Péricytes , Transduction du signal , Animaux , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Péricytes/métabolisme , Péricytes/anatomopathologie , Péricytes/immunologie , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Inflammation/immunologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Souris transgéniques , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Souris knockout , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/immunologie , Mâle , Glucose/métabolisme
10.
Nat Commun ; 15(1): 3810, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38714671

RÉSUMÉ

Previous studies have revealed heterogeneity in the progression to clinical type 1 diabetes in children who develop islet-specific antibodies either to insulin (IAA) or glutamic acid decarboxylase (GADA) as the first autoantibodies. Here, we test the hypothesis that children who later develop clinical disease have different early immune responses, depending on the type of the first autoantibody to appear (GADA-first or IAA-first). We use mass cytometry for deep immune profiling of peripheral blood mononuclear cell samples longitudinally collected from children who later progressed to clinical disease (IAA-first, GADA-first, ≥2 autoantibodies first groups) and matched for age, sex, and HLA controls who did not, as part of the Type 1 Diabetes Prediction and Prevention study. We identify differences in immune cell composition of children who later develop disease depending on the type of autoantibodies that appear first. Notably, we observe an increase in CD161 expression in natural killer cells of children with ≥2 autoantibodies and validate this in an independent cohort. The results highlight the importance of endotype-specific analyses and are likely to contribute to our understanding of pathogenic mechanisms underlying type 1 diabetes development.


Sujet(s)
Autoanticorps , Diabète de type 1 , Glutamate decarboxylase , Immunité cellulaire , Humains , Diabète de type 1/immunologie , Autoanticorps/immunologie , Autoanticorps/sang , Enfant , Femelle , Mâle , Glutamate decarboxylase/immunologie , Enfant d'âge préscolaire , Adolescent , Cellules tueuses naturelles/immunologie , Agranulocytes/immunologie , Insuline/immunologie , Ilots pancréatiques/immunologie , Évolution de la maladie
11.
J Immunol ; 212(12): 1971-1980, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38709159

RÉSUMÉ

Most pancreatic islets are destroyed immediately after intraportal transplantation by an instant blood-mediated inflammatory reaction (IBMIR) generated through activation of coagulation, complement, and proinflammatory pathways. Thus, effective mitigation of IBMIR may be contingent on the combined use of agents targeting these pathways for modulation. CD47 and thrombomodulin (TM) are two molecules with distinct functions in regulating coagulation and proinflammatory responses. We previously reported that the islet surface can be modified with biotin for transient display of novel forms of these two molecules chimeric with streptavidin (SA), that is, thrombomodulin chimeric with SA (SA-TM) and CD47 chimeric with SA (SA-CD47), as single agents with improved engraftment following intraportal transplantation. This study aimed to test whether islets can be coengineered with SA-TM and SA-CD47 molecules as a combinatorial approach to improve engraftment by inhibiting IBMIR. Mouse islets were effectively coengineered with both molecules without a detectable negative impact on their viability and metabolic function. Coengineered islets were refractory to destruction by IBMIR ex vivo and showed enhanced engraftment and sustained function in a marginal mass syngeneic intraportal transplantation model. Improved engraftment correlated with a reduction in intragraft innate immune infiltrates, particularly neutrophils and M1 macrophages. Moreover, transcripts for various intragraft procoagulatory and proinflammatory agents, including tissue factor, HMGB1 (high-mobility group box-1), IL-1ß, IL-6, TNF-α, IFN-γ, and MIP-1α, were significantly reduced in coengineered islets. These data demonstrate that the transient codisplay of SA-TM and SA-CD47 proteins on the islet surface is a facile and effective platform to modulate procoagulatory and inflammatory responses with implications for both autologous and allogeneic islet transplantation.


Sujet(s)
Antigènes CD47 , Inflammation , Transplantation d'ilots de Langerhans , Ilots pancréatiques , Souris de lignée C57BL , Thrombomoduline , Animaux , Mâle , Souris , Antigènes CD47/immunologie , Antigènes CD47/métabolisme , Inflammation/immunologie , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Transplantation d'ilots de Langerhans/méthodes , Streptavidine
12.
Diabetes ; 73(8): 1278-1284, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38701365

RÉSUMÉ

Accumulating data suggest a role for the lysosomal protease cathepsin S (CTSS) in type 1 diabetes. Circulating CTSS is increased in type 1 diabetes; however, whether CTSS has protective or deleterious effects is unclear. The study's objectives were to examine the biomarker potential of CTSS in new-onset type 1 diabetes, and to investigate the expression and secretion of CTSS in human islets and ß-cells. The CTSS level was analyzed in serum from children with new-onset type 1 diabetes and autoantibody-positive and -negative siblings by ELISA. The expression and secretion of CTSS were evaluated in isolated human islets and EndoC-ßH5 cells by real-time qPCR, immunoblotting, and ELISA. The CTSS serum level was elevated in children with new-onset type 1 diabetes and positively associated with autoantibody status in healthy siblings. Human islets and EndoC-ßH5 cells demonstrated induction and secretion of CTSS after exposure to proinflammatory cytokines, a model system of islet inflammation. Analysis of publicly available single-cell RNA sequencing data on human islets showed that elevated CTSS expression was exclusive for the ß-cells in donors with type 1 diabetes as compared with nondiabetic donors. These findings suggest a potential of CTSS as a diagnostic biomarker in type 1 diabetes.


Sujet(s)
Autoanticorps , Cathepsines , Diabète de type 1 , Ilots pancréatiques , Fratrie , Humains , Diabète de type 1/immunologie , Diabète de type 1/sang , Autoanticorps/sang , Autoanticorps/immunologie , Cathepsines/sang , Mâle , Enfant , Femelle , Ilots pancréatiques/immunologie , Adolescent , Cellules à insuline/immunologie , Cellules à insuline/métabolisme , Enfant d'âge préscolaire , Marqueurs biologiques/sang
13.
J Nutr ; 154(7): 2244-2254, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38795745

RÉSUMÉ

BACKGROUND: Gut dysbiosis and increased intestinal permeability have been reported to precede type 1 diabetes-related autoimmunity. The role of gut inflammation in autoimmunity is not understood. OBJECTIVES: This study aimed to assess whether gut inflammation markers are associated with risk of islet autoimmunity and whether diet is associated with gut inflammation markers. METHODS: A nested case-control sample of 75 case children with islet autoimmunity and 88 control children was acquired from the Finnish Type 1 Diabetes Prediction and Prevention cohort. Diet was assessed with 3-d food records, and calprotectin and human ß-defensin-2 (HBD-2) were analyzed from stool samples at 6 and 12 mo of age. Conditional logistic regression analysis was used in a matched case-control setting to assess risk of autoimmunity. Analysis of variance, independent samples t test, and a general linear model were used in secondary analyses to test associations of background characteristics and dietary factors with inflammation markers. RESULTS: In unadjusted analyses, calprotectin was not associated with risk of islet autoimmunity, whereas HBD-2 in the middle (odds ratio [OR]: 3.23; 95% confidence interval [CI]: 1.03, 10.08) or highest tertile (OR: 3.02; 95% CI: 1.05, 8.69) in comparison to the lowest at 12 mo of age showed borderline association (P-trend = 0.063) with higher risk of islet autoimmunity. Excluding children with cow milk allergy in sensitivity analyses strengthened the association of HBD-2 with islet autoimmunity, whereas adjusting for dietary factors and maternal education weakened it. At age 12 mo, higher fat intake was associated with higher HBD-2 (ß: 0.219; 95% CI: 0.110, 0.328) and higher intake of dietary fiber (ß: -0.294; 95% CI: -0.510, -0.078), magnesium (ß: -0.036; 95% CI: -0.059, -0.014), and potassium (ß: -0.003; 95% CI: -0.005, -0.001) with lower HBD-2. CONCLUSIONS: Higher HBD-2 in infancy may be associated with higher risk of islet autoimmunity. Dietary factors play a role in gut inflammatory status.


Sujet(s)
Auto-immunité , Marqueurs biologiques , Diabète de type 1 , Régime alimentaire , Ilots pancréatiques , Complexe antigénique L1 leucocytaire , bêta-Défensines , Humains , Études cas-témoins , Finlande , Femelle , Mâle , Complexe antigénique L1 leucocytaire/analyse , Diabète de type 1/immunologie , Nourrisson , Ilots pancréatiques/immunologie , Facteurs de risque , Inflammation , Fèces/composition chimique
14.
Immunity ; 57(7): 1629-1647.e8, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38754432

RÉSUMÉ

The pancreatic islet microenvironment is highly oxidative, rendering ß cells vulnerable to autoinflammatory insults. Here, we examined the role of islet resident macrophages in the autoimmune attack that initiates type 1 diabetes. Islet macrophages highly expressed CXCL16, a chemokine and scavenger receptor for oxidized low-density lipoproteins (OxLDLs), regardless of autoimmune predisposition. Deletion of Cxcl16 in nonobese diabetic (NOD) mice suppressed the development of autoimmune diabetes. Mechanistically, Cxcl16 deficiency impaired clearance of OxLDL by islet macrophages, leading to OxLDL accumulation in pancreatic islets and a substantial reduction in intra-islet transitory (Texint) CD8+ T cells displaying proliferative and effector signatures. Texint cells were vulnerable to oxidative stress and diminished by ferroptosis; PD-1 blockade rescued this population and reversed diabetes resistance in NOD.Cxcl16-/- mice. Thus, OxLDL scavenging in pancreatic islets inadvertently promotes differentiation of pathogenic CD8+ T cells, presenting a paradigm wherein tissue homeostasis processes can facilitate autoimmune pathogenesis in predisposed individuals.


Sujet(s)
Auto-immunité , Lymphocytes T CD8+ , Différenciation cellulaire , Chimiokine CXCL16 , Diabète de type 1 , Ilots pancréatiques , Lipoprotéines LDL , Macrophages , Souris de lignée NOD , Souris knockout , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Souris , Lipoprotéines LDL/métabolisme , Lipoprotéines LDL/immunologie , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Chimiokine CXCL16/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Souris de lignée C57BL
15.
J Autoimmun ; 146: 103228, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38642507

RÉSUMÉ

CD6 is a glycoprotein expressed on CD4 and CD8 T cells involved in immunoregulation. CD318 has been identified as a CD6 ligand. The role of CD318 in T cell immunity is restricted as it has only been investigated in a few mice autoimmune models but not in human diseases. CD318 expression was thought to be limited to mesenchymal-epithelial cells and, therefore, contribute to CD6-mediated T cell activation in the CD318-expressing tissue rather than through interaction with antigen-presenting cells. Here, we report CD318 expression in a subpopulation of CD318+ myeloid dendritic (mDC), whereas the other peripheral blood populations were CD318 negative. However, CD318 can be induced by activation: a subset of monocytes treated with LPS and IFNγ and in vitro monocyte derived DCs were CD318+. We also showed that recombinant CD318 inhibited T cell function. Strikingly, CD318+ DCs suppressed the proliferation of autoreactive T cells specific for GAD65, a well-known targeted self-antigen in Type 1 Diabetes (T1D). Our study provides new insight into the role of the CD318/CD6 axis in the immunopathogenesis of inflammation, suggesting a novel immunoregulatory role of CD318 in T cell-mediated autoimmune diseases and identifying a potential novel immune checkpoint inhibitor as a target for intervention in T1D which is an unmet therapeutic need.


Sujet(s)
Antigènes CD , Autoantigènes , Cellules dendritiques , Diabète de type 1 , Ilots pancréatiques , Activation des lymphocytes , Humains , Diabète de type 1/immunologie , Diabète de type 1/métabolisme , Activation des lymphocytes/immunologie , Autoantigènes/immunologie , Antigènes CD/métabolisme , Antigènes CD/immunologie , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Antigènes de différenciation des lymphocytes T/métabolisme , Antigènes de différenciation des lymphocytes T/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Cellules cultivées , Glutamate decarboxylase
16.
Cell Rep Med ; 5(5): 101535, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38677282

RÉSUMÉ

Type 1 diabetes (T1D) is a chronic condition in which beta cells are destroyed by immune cells. Despite progress in immunotherapies that could delay T1D onset, early detection of autoimmunity remains challenging. Here, we evaluate the utility of machine learning for early prediction of T1D using single-cell analysis of islets. Using gradient-boosting algorithms, we model changes in gene expression of single cells from pancreatic tissues in T1D and non-diabetic organ donors. We assess if mathematical modeling could predict the likelihood of T1D development in non-diabetic autoantibody-positive donors. While most autoantibody-positive donors are predicted to be non-diabetic, select donors with unique gene signatures are classified as T1D. Our strategy also reveals a shared gene signature in distinct T1D-associated models across cell types, suggesting a common effect of the disease on transcriptional outputs of these cells. Our study establishes a precedent for using machine learning in early detection of T1D.


Sujet(s)
Diabète de type 1 , Évolution de la maladie , Ilots pancréatiques , Apprentissage machine , Analyse sur cellule unique , Transcriptome , Humains , Diabète de type 1/génétique , Diabète de type 1/immunologie , Diabète de type 1/anatomopathologie , Analyse sur cellule unique/méthodes , Ilots pancréatiques/métabolisme , Ilots pancréatiques/immunologie , Transcriptome/génétique , Autoanticorps/immunologie , Analyse de profil d'expression de gènes/méthodes , Mâle , Femelle , Cellules à insuline/métabolisme , Adulte
17.
Front Immunol ; 15: 1375177, 2024.
Article de Anglais | MEDLINE | ID: mdl-38650946

RÉSUMÉ

Human allogeneic pancreatic islet transplantation is a life-changing treatment for patients with severe Type 1 Diabetes (T1D) who suffer from hypoglycemia unawareness and high risk of severe hypoglycemia. However, intensive immunosuppression is required to prevent immune rejection of the graft, that may in turn lead to undesirable side effects such as toxicity to the islet cells, kidney toxicity, occurrence of opportunistic infections, and malignancies. The shortage of cadaveric human islet donors further limits islet transplantation as a treatment option for widespread adoption. Alternatively, porcine islets have been considered as another source of insulin-secreting cells for transplantation in T1D patients, though xeno-transplants raise concerns over the risk of endogenous retrovirus transmission and immunological incompatibility. As a result, technological advancements have been made to protect transplanted islets from immune rejection and inflammation, ideally in the absence of chronic immunosuppression, to improve the outcomes and accessibility of allogeneic islet cell replacement therapies. These include the use of microencapsulation or macroencapsulation devices designed to provide an immunoprotective environment using a cell-impermeable layer, preventing immune cell attack of the transplanted cells. Other up and coming advancements are based on the use of stem cells as the starting source material for generating islet cells 'on-demand'. These starting stem cell sources include human induced pluripotent stem cells (hiPSCs) that have been genetically engineered to avoid the host immune response, curated HLA-selected donor hiPSCs that can be matched with recipients within a given population, and multipotent stem cells with natural immune privilege properties. These strategies are developed to provide an immune-evasive cell resource for allogeneic cell therapy. This review will summarize the immunological challenges facing islet transplantation and highlight recent bio-engineering and cell-based approaches aimed at avoiding immune rejection, to improve the accessibility of islet cell therapy and enhance treatment outcomes. Better understanding of the different approaches and their limitations can guide future research endeavors towards developing more comprehensive and targeted strategies for creating a more tolerogenic microenvironment, and improve the effectiveness and sustainability of islet transplantation to benefit more patients.


Sujet(s)
Diabète de type 1 , Rejet du greffon , Transplantation d'ilots de Langerhans , Transplantation d'ilots de Langerhans/méthodes , Humains , Animaux , Diabète de type 1/immunologie , Diabète de type 1/thérapie , Rejet du greffon/immunologie , Rejet du greffon/prévention et contrôle , Génie biomédical/méthodes , Ilots pancréatiques/immunologie
18.
Acta Diabetol ; 61(7): 897-907, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38530415

RÉSUMÉ

OBJECTIVE: To examine the association between islet autoantibodies (IAbs) and the retinal neurovascular changes in type 1 diabetes mellitus (T1DM) with no diabetic retinopathy (NDR). METHODS: This cross-sectional study measured the neural retinal structure and microvascular density of 118 NDR eyes using spectral-domain optical coherence tomography angiography. Retinal structure parameters included retinal thickness (RT), inner retinal thickness (iRT), retina never fibral layer thickness (RNFL thickness), ganglion cell complex thickness (GCC thickness), and loss volume of GCC. Microvascular parameters included vessel density of superficial capillary plexus (sVD), vessel density of deep capillary plexus, and vessel density of choroid capillary plexus. Comparison and correlation analyses of these OCTA parameters were made with various IAbs, including glutamic acid decarboxylase antibody (GADA), tyrosine phosphatase-related islet antigen 2 antibody (IA2A), and zinc transporter 8 antibody (ZnT8A). A general linear model was used to understand the association of IAbs with the retina parameters. RESULTS: The IAb positive (IAbs +) group, which included 85 patients, had thinner RT (235.20 ± 18.10 mm vs. 244.40 ± 19.90 mm at fovea, P = 0.021) and thinner iRT (120.10 ± 9.00 mm vs. 124.70 ± 6.90 mm at parafovea, P = 0.015), compared with the IAb negative (IAbs-) group comprising 33 patients. Furthermore, a more severe reduction of RT was demonstrated in the presence of multiple IAbs. Among the three IAbs, GADA was the most significant independent risk factor of all-round RT decrease (ß = -0.20 vs. -0.27 at fovea and parafovea, respectively, P < 0.05), while titers of IA2A negatively affect sVD in the parafovea (ß = -0.316, P = 0.003). CONCLUSIONS: IAbs are associated with neural retinal thinning and microcirculation reduction in T1DM patients before the clinical onset of diabetic retinopathy.


Sujet(s)
Autoanticorps , Diabète de type 1 , Rétinopathie diabétique , Microcirculation , Rétine , Humains , Autoanticorps/sang , Autoanticorps/immunologie , Diabète de type 1/immunologie , Diabète de type 1/anatomopathologie , Diabète de type 1/imagerie diagnostique , Mâle , Femelle , Études transversales , Adulte , Rétinopathie diabétique/immunologie , Rétinopathie diabétique/anatomopathologie , Rétinopathie diabétique/imagerie diagnostique , Rétine/imagerie diagnostique , Rétine/immunologie , Rétine/anatomopathologie , Adulte d'âge moyen , Tomographie par cohérence optique , Ilots pancréatiques/immunologie , Ilots pancréatiques/imagerie diagnostique , Ilots pancréatiques/anatomopathologie , Ilots pancréatiques/vascularisation , Vaisseaux rétiniens/imagerie diagnostique , Vaisseaux rétiniens/anatomopathologie , Jeune adulte
19.
Cells ; 13(5)2024 Feb 27.
Article de Anglais | MEDLINE | ID: mdl-38474380

RÉSUMÉ

Islet transplantation is a therapeutic option to replace ß-cell mass lost during type 1 or type 3c diabetes. Innate immune responses, particularly the instant blood-mediated inflammatory reaction and activation of monocytes, play a major role in the loss of transplanted islet tissue. In this study, we aimed to investigate the inhibition of toll-like receptor 4 (TLR4) on innate inflammatory responses. We first demonstrate a significant loss of graft function shortly after transplant through the assessment of miR-375 and miR-200c in plasma as biomarkers. Using in vitro models, we investigate how targeting TLR4 mitigates islet damage and immune cell activation during the peritransplant period. The results of this study support the application of TAK-242 as a therapeutic agent to reduce inflammatory and innate immune responses to islets immediately following transplantation into the hepatic portal vein. Therefore, TLR4 may serve as a target to improve islet transplant outcomes in the future.


Sujet(s)
Immunité innée , Transplantation d'ilots de Langerhans , Ilots pancréatiques , microARN , Sulfonamides , Récepteur de type Toll-4 , Immunité innée/effets des médicaments et des substances chimiques , Transplantation d'ilots de Langerhans/méthodes , Sulfonamides/pharmacologie , Récepteur de type Toll-4/antagonistes et inhibiteurs , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/immunologie , Humains
20.
Eur J Nutr ; 63(4): 1329-1338, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38413484

RÉSUMÉ

PURPOSE: The aim was to study the association between dietary intake of B vitamins in childhood and the risk of islet autoimmunity (IA) and progression to type 1 diabetes (T1D) by the age of 10 years. METHODS: We followed 8500 T1D-susceptible children born in the U.S., Finland, Sweden, and Germany in 2004 -2010 from the Environmental Determinants of Diabetes in the Young (TEDDY) study, which is a prospective observational birth cohort. Dietary intake of seven B vitamins was calculated from foods and dietary supplements based on 24-h recall at 3 months and 3-day food records collected regularly from 6 months to 10 years of age. Cox proportional hazard models were adjusted for energy, HLA-genotype, first-degree relative with T1D, sex, and country. RESULTS: A total of 778 (9.2) children developed at least one autoantibody (any IA), and 335 (3.9%) developed multiple autoantibodies. 280 (3.3%) children had IAA and 319 (3.8%) GADA as the first autoantibody. 344 (44%) children with IA progressed to T1D. We observed that higher intake of niacin was associated with a decreased risk of developing multiple autoantibodies (HR 0.95; 95% CI 0.92, 0.98) per 1 mg/1000 kcal in niacin intake. Higher intake of pyridoxine (HR 0.66; 95% CI 0.46, 0.96) and vitamin B12 (HR 0.87; 95% CI 0.77, 0.97) was associated with a decreased risk of IAA-first autoimmunity. Higher intake of riboflavin (HR 1.38; 95% CI 1.05, 1.80) was associated with an increased risk of GADA-first autoimmunity. There were no associations between any of the B vitamins and the outcomes "any IA" and progression from IA to T1D.  CONCLUSION: In this multinational, prospective birth cohort of children with genetic susceptibility to T1D, we observed some direct and inverse associations between different B vitamins and risk of IA.


Sujet(s)
Autoanticorps , Auto-immunité , Diabète de type 1 , Ilots pancréatiques , Complexe vitaminique B , Humains , Diabète de type 1/immunologie , Diabète de type 1/épidémiologie , Mâle , Femelle , Complexe vitaminique B/administration et posologie , Études prospectives , Enfant , Enfant d'âge préscolaire , Nourrisson , Ilots pancréatiques/immunologie , Autoanticorps/sang , Facteurs de risque , Régime alimentaire/méthodes , Régime alimentaire/statistiques et données numériques , Modèles des risques proportionnels , États-Unis/épidémiologie , Finlande/épidémiologie , Suède/épidémiologie , Allemagne/épidémiologie , Compléments alimentaires , Cohorte de naissance , Évolution de la maladie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE