Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
APMIS ; 2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38798081

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease, resulting in diminished islet integrity and destruction of the insulin-secreting beta cells. In this review, we investigate the intrinsic relationship between the development of T1D and the activity of the beta cells. The idea was initially hypothesized in 1982 that an increased beta-cell activity would enhance the surface antigen expression and thereby attract the immune system. Later, other findings support this idea, including increased risk of T1D development during third trimester of pregnancy, and the difference in T1D incidence in Russian and Finnish Karelia due to different lifestyles. Other implications of high beta-cell activity, such as reduced sulfatide levels, formation of non-correct insulin molecules and an increase in IFN-alpha upon virus attack, can contribute to the development of T1D. A possible way to prevent the development of T1D is to diminish beta-cell activity, which has shown promising results in animal models.

2.
Diabetes ; 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38701365

RESUMO

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 pro-inflammatory 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 to non-diabetic donors. These findings suggest a potential of CTSS as a diagnostic biomarker in type 1 diabetes.

4.
Diabetes Metab Res Rev ; 40(3): e3792, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38517704

RESUMO

AIMS: Sulfatide is a chaperone for insulin manufacturing in beta cells. Here we explore whether the blood glucose values normally could be associated with this sphingolipid and especially two of its building enzymes CERS2 and CERS6. Both T1D and T2D have low blood sulfatide levels, and insulin resistance on beta cells at clinical diagnosis. Furthermore, we examined islet pericytes for sulfatide, and beta-cell receptors for GLP-1, both of which are related to the insulin production. MATERIALS AND METHODS: We examined mRNA levels in islets from the DiViD and nPOD studies, performed genetic association analyses, and histologically investigated pericytes in the islets for sulfatide. RESULTS: Polymorphisms of the gene encoding the CERS6 enzyme responsible for synthesising dihydroceramide, a precursor to sulfatide, are associated with random blood glucose values in non-diabetic persons. This fits well with our finding of sulfatide in pericytes in the islets, which regulates the capillary blood flow in the islets of Langerhans, which is important for oxygen supply to insulin production. In the islets of newly diagnosed T1D patients, we observed low levels of GLP-1 receptors; this may explain the insulin resistance in their beta cells and their low insulin production. In T2D patients, we identified associated polymorphisms in both CERS2 and CERS6. CONCLUSIONS: Here, we describe several polymorphisms in sulfatide enzymes related to blood glucose levels and HbA1c in non-diabetic individuals. Islet pericytes from such persons contain sulfatide. Furthermore, low insulin secretion in newly diagnosed T1D may be explained by beta-cell insulin resistance due to low levels of GLP-1 receptors.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Resistência à Insulina , Ilhotas Pancreáticas , Humanos , Glicemia , Esfingolipídeos , Resistência à Insulina/genética , Pericitos , Sulfoglicoesfingolipídeos , Insulina , Insulina Regular Humana , Diabetes Mellitus Tipo 2/genética , Peptídeo 1 Semelhante ao Glucagon , Glucose
6.
PLoS One ; 18(9): e0291063, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37669305

RESUMO

Brown bears (Ursus arctos) prepare for winter by overeating and increasing adipose stores, before hibernating for up to six months without eating, drinking, and with minimal movement. In spring, the bears exit the den without any damage to organs or physiology. Recent clinical research has shown that specific lipids and lipid profiles are of special interest for diseases such as diabetes type 1 and 2. Furthermore, rodent experiments show that lipids such as sulfatide protects rodents against diabetes. As free-ranging bears experience fat accumulation and month-long physical inactivity without developing diabetes, they could possibly be affected by similar protective measures. In this study, we investigated whether lipid profiles of brown bears are related to protection against hibernation-induced damage. We sampled plasma from 10 free-ranging Scandinavian brown bears during winter hibernation and repeated sampling during active state in the summer period. With quantitative shotgun lipidomics and liquid chromatography-mass spectrometry, we profiled 314 lipid species from 26 lipid classes. A principal component analysis revealed that active and hibernation samples could be distinguished from each other based on their lipid profiles. Six lipid classes were significantly altered when comparing plasma from active state and hibernation: Hexosylceramide, phosphatidylglycerol, and lysophosphatidylglycerol were higher during hibernation, while phosphatidylcholine ether, phosphatidylethanolamine ether, and phosphatidylinositol were lower. Additionally, sulfatide species with shorter chain lengths were lower, while longer chain length sulfatides were higher during hibernation. Lipids that are altered in bears are described by others as relevant for and associated with diabetes, which strengthens their position as potential effectors during hibernation. From this analysis, a range of lipids are suggested as potential protectors of bear physiology, and of potential importance in diabetes.


Assuntos
Diabetes Mellitus Tipo 1 , Ursidae , Animais , Sulfoglicoesfingolipídeos , Adiposidade , Éteres
7.
PLoS One ; 18(9): e0289258, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37682921

RESUMO

Type 1 diabetes (T1D) is an autoimmune disease with an unexplained rising incidence for which environmental factors like gluten may play a role. Previously, we showed that a gluten-free (GF) diet provided strictly in utero reduces the autoimmune diabetes incidence in Non-Obese Diabetic (NOD) mice compared to a gluten-containing standard (STD) diet. The current study was initiated to elucidate possible mechanisms behind the diabetes-alleviating effect of the same diet intervention. NOD mice received either a GF Altromin diet or a STD Altromin diet during pregnancy. Female offspring from both groups were fed a STD diet throughout life and their diabetes incidence was recorded for 200 days. The following parameters were measured in 13-week-old female offspring: insulitis degree, glucose and insulin tolerance, and plasma insulin autoantibody titer. The diet intervention showed no reduction in autoimmune diabetes incidence, insulitis degree, glucose nor insulin tolerance and plasma insulin autoantibody titer. In conclusion, this study could not replicate the previously observed diabetes alleviative effects of a maternal gluten-free diet in NOD mouse offspring and could therefore not further elucidate potential mechanisms.


Assuntos
Diabetes Mellitus Tipo 1 , Fenômenos Fisiológicos da Nutrição Materna , Animais , Feminino , Camundongos , Gravidez , Autoanticorpos , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Tipo 1/dietoterapia , Dieta Livre de Glúten , Glucose , Glutens , Insulinas , Camundongos Endogâmicos NOD
8.
Diabetes Metab Res Rev ; 39(7): e3678, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37395313

RESUMO

AIMS: To investigate if HLA risk haplotypes and HbA1c levels are associated with the expression levels of innate anti-viral immune pathway genes in type 1 diabetes. MATERIALS AND METHODS: We investigated RNA expression levels of innate anti-viral immune pathway genes in laser-dissected islets from two to five tissue sections per donor from the Diabetes Virus Detection study and the network of Pancreatic Organ Donors in relation to HLA risk haplotypes (non-predisposed and predisposed) and HbA1c levels (normal, elevated, and high). RESULTS: The expression of innate anti-viral immune genes (TLR7, OAS1, OAS3 etc.) was significantly increased in individuals with predisposing vs non-predisposing HLA haplotypes. Also, the expression of several of the innate anti-viral immune genes from the HLA risk haplotype analysis was significantly increased in the group with high vs normal HbA1c. Furthermore, the gene expression of OAS2 was significantly increased in the group with high HbA1c vs elevated HbA1c. CONCLUSIONS: Expression of innate anti-viral immune pathway genes was increased in individuals with predisposing HLA risk haplotypes and those with high HbA1c. This indicates that type 1 diabetes might well begin with alterations in innate anti-viral immunity, and already at this stage be associated with HLA risk haplotypes.

9.
Cells ; 12(12)2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37371037

RESUMO

The incidence of the autoimmune disease type 1 diabetes is increasing, likely caused by environmental factors. A gluten-free diet has previously been shown to ameliorate autoimmune diabetes in non-obese diabetic (NOD) mice and humans. Although the exact mechanisms are not understood, interventions influencing the intestinal microbiota early in life affect the risk of type 1 diabetes. Here, we characterize how NOD mice that are fed a gluten-free (GF) diet differ from NOD mice that are fed a gluten-containing standard (STD) diet in terms of their microbiota composition by 16S rRNA gene amplicon sequencing and pancreatic immune environment by real-time quantitative PCR at the prediabetic stage at 6 and 13 weeks of age. Gut microbiota analysis revealed highly distinct microbiota compositions in both the cecum and the colon of GF-fed mice compared with STD-fed mice. The microbiotas of the GF-fed mice were characterized by an increased Firmicutes/Bacteroidetes ratio, an increased abundance of short chain fatty acid (particularly butyrate)-producing bacteria, and a reduced abundance of Lactobacilli compared with STD mice. We found that the insulitis score in the GF mice was significantly reduced compared with the STD mice and that the markers for regulatory T cells and T helper 2 cells were upregulated in the pancreas of the GF mice. In conclusion, a GF diet during pre- and early post-natal life induces shifts in the cecal and colonic microbiota compatible with a less inflammatory environment, providing a likely mechanism for the protective effect of a GF diet in humans.


Assuntos
Diabetes Mellitus Tipo 1 , Dieta Livre de Glúten , Estado Pré-Diabético , Animais , Feminino , Camundongos , Gravidez , Bactérias , Diabetes Mellitus Tipo 1/prevenção & controle , Camundongos Endogâmicos NOD , Estado Pré-Diabético/prevenção & controle , RNA Ribossômico 16S/genética , Linfócitos T Reguladores , Microbioma Gastrointestinal
10.
Diabetes Obes Metab ; 25(9): 2514-2525, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37246802

RESUMO

AIM: To study the effect of sulfatide on gene expression and proliferation of human primary fibroblasts induced by insulin, insulin-like growth factor-1 and human growth hormone. MATERIALS AND METHODS: Human primary fibroblasts were exposed to 1, 3 and 30 µM of sulfatide or its precursor galactosylceramide (GalCer). Proliferation was determined by 3 H-thymidine incorporation and gene expression via microarray analysis. RESULTS: Sulfatide and GalCer reduced the growth rate of fibroblasts by 32%-82% when exposed to 0.5 nM insulin. After challenge with 120 µM of H2 O2 , sulfatide reduced membrane leakage. Fibroblast gene expression was altered by sulfatide in gene pathways associated with cell cycle/growth, transforming growth factor-ß function, and encoding of proteins involved in intracellular signalling. NFKBIA, a key control element in NF-кB regulation, was decreased 2-fold by sulfatide. CONCLUSIONS: Sulfatide strongly inhibits fibroblast growth. We therefore suggest the addition of sulfatide to injectable commercial insulin formulations, which would reduce adverse fibroblast growth and improve well-being in patients with diabetes.


Assuntos
Insulina , Sulfoglicoesfingolipídeos , Humanos , Insulina/farmacologia , Insulina/metabolismo , Sulfoglicoesfingolipídeos/metabolismo , Sulfoglicoesfingolipídeos/farmacologia , Insulina Regular Humana , Fibroblastos/metabolismo , Estresse Oxidativo
11.
Sci Rep ; 12(1): 18149, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307540

RESUMO

Type 1 diabetes (T1D) incidence is increased after COVID-19 infection in children under 18 years of age. Interferon-α-activated oligoadenylate synthetase and downstream RNAseL activation degrade pathogen RNA, but can also damage host RNA when RNAseL activity is poorly regulated. One such regulator is PDE12 which degrades 2'-5' oligoadenylate units, thereby decreasing RNAseL activity. We analyzed PDE12 expression in islets from non-diabetic donors, individuals with newly (median disease duration 35 days) and recently (5 years) diagnosed T1D, and individuals with type 2 diabetes (T2D). We also analyzed PDE12 single-nucleotide polymorphisms (SNPs) relative to T1D incidence. PDE12 expression was decreased in individuals with recently diagnosed T1D, in three of five individuals with newly diagnosed T1D, but not in individuals with T2D. Two rare PDE12 SNPs were found to have odds ratios of 1.80 and 1.74 for T1D development. We discuss whether decreased PDE12 expression after COVID-19 infection might be part of the up to 2.5-fold increase in T1D incidence.


Assuntos
COVID-19 , Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Criança , Humanos , Adolescente , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , COVID-19/genética , Interferon-alfa , RNA
12.
Front Endocrinol (Lausanne) ; 13: 876470, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36093076

RESUMO

In this review after a lifelong research career, my personal opinion on the development of type 1 diabetes (T1D) from its very start to clinical manifestation will be described. T1D is a disease of an increased intestinal permeability and a reduced pancreas volume. I am convinced that virus might be the initiator and that this virus could persist on strategically significant locations. Furthermore, intake of gluten is important both in foetal life and at later ages. Disturbances in sphingolipid metabolism may also be of crucial importance. During certain stages of T1D, T cells take over resulting in the ultimate destruction of beta cells, which manifests T1D as an autoimmune disease. Several preventive and early treatment strategies are mentioned. All together this review has more new theories than usually, and it might also be more speculative than ordinarily. But without new ideas and theories advancement is difficult, even though everything might not hold true during the continuous discovery of the etiology and pathogenesis of T1D.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Causalidade , Diabetes Mellitus Tipo 1/etiologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Células Secretoras de Insulina/metabolismo , Linfócitos T/metabolismo
14.
PLoS One ; 17(4): e0266719, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35417506

RESUMO

The purpose of this study was to compare the effect of a gluten-free diet and/or antibiotics on tetanus vaccine induced immunoglobulin G titers and immune cell levels in BALB/c mice. The gluten-free diet was associated with a reduced anti-tetanus IgG response, and it increased the relative abundance of the anti-inflammatory Bifidobacterium significantly in some of the mice. Antibiotics also led to gut microbiota changes and lower initial vaccine titer. After a second vaccination, neither gluten-free diet nor antibiotics reduced the titers. In the spleen, the gluten-free diet significantly increased regulatory T cell (Treg) fractions, CD4+ T cell activation, and tolerogenic dendritic cell fractions and activation, which extend the downregulating effect of the Treg. Therefore, the systemic effect of the gluten-free diet seems mainly tolerogenic. Antibiotics reduced the fractions of CD4+ T and B cells in the mesenteric lymph nodes. These results suggest that vaccine response in mice is under influence of their diet, the gut microbiota and the interplay between them. However, a gluten-free diet seems to work through mechanisms different from those induced by antibiotics. Therefore, diet should be considered when testing vaccines in mice and developing vaccines for humans.


Assuntos
Microbioma Gastrointestinal , Tétano , Animais , Antibacterianos/farmacologia , Dieta Livre de Glúten , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
15.
J Autoimmun ; 127: 102795, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35101708

RESUMO

Experimental and clinical data suggest that a gluten-free diet attenuates the development of type 1 diabetes. A gluten-free diet changes the gut microbiota composition, and such microbial changes are expected to reduce the autoimmune responses. However, in experiments with laboratory mice, a gluten-free diet changes the gut microbiota differently under varying experimental settings, questioning the specific role of the gut microbes. Here we show that a maternal gluten-free diet until weaning of their pups, delayed type 1 diabetes in both dams (parent generation) and offspring (F1 generation) of untreated non-obese diabetic (NOD) mice and in mice treated with a full cocktail of antibiotics that eradicates most of the existing microbiota. Breeding a second (F2) generation of NOD mice, never exposed to the gluten-free diet or the associated microbial changes, also demonstrated a preventative effect on type 1 diabetes even though their parents (the F1 generation) had only been on a gluten-free diet very early in life. Collectively, the experimental data, thus, points towards microbiota-independent dietary protection. Furthermore, both the perinatal gluten-free diet and antibiotic treatment reduced inflammation in the salivary glands and improved glucose challenged beta cell function in the F1 offspring. However, in contrast to the autoimmune response in the pancreas, those changes appeared to be microbiota dependent, as they were missing in the antibiotic treated mice, and do, therefore, not seem to be related to the preventative effect on type 1 diabetes. Interestingly, adoptive transfer of splenocytes from gluten-free fed mice protected NOD.SCID mice from developing diabetes, demonstrating that the anti-diabetic effect of a gluten-free diet was based on early life changes in the evolving immune system. In particular, genes involved in regulation of lymphocyte activation, proliferation, and cell adhesion were highly expressed in the spleen in gluten-free fed mice at weaning compared to control fed mice of the F1 generation, which suggested that gluten promotes autoimmunity by inhibiting immune regulation, though the involvement of the specific genes needs further investigation. In conclusion, gluten-free diet reduces autoimmune inflammation in salivary glands and pancreas in NOD mice in a microbiota-dependent and -independent manner respectively, and has preventative effect on type 1 diabetes by modulating the systemic immune system.


Assuntos
Diabetes Mellitus Tipo 1 , Microbiota , Animais , Dieta Livre de Glúten , Feminino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Gravidez
16.
Oral Dis ; 28(3): 639-647, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33432638

RESUMO

OBJECTIVES: A lifelong gluten-free (GF) diet ameliorates autoimmune diabetes in non-obese diabetic (NOD) mice and most likely in humans. Besides diabetes, NOD mice develop focal sialadenitis, as seen in Sjögren's syndrome (SS). In humans, type 1 diabetes (T1D) is also linked to SS. Here, we investigated whether a lifelong GF diet influences the immune cell infiltration in the salivary glands and pancreatic islets in NOD mice. METHODS: NOD mice were fed a lifelong (i.e. 13 weeks) GF or gluten-containing standard (STD) diet. Insulitis and sialadenitis were scored on H&E-stained paraffin-embedded sections of pancreas and submandibular glands. Immune cell specificity and distribution were investigated immunohistochemically. RESULTS: There were fewer CD68+ and CD4+ cells in submandibular gland areas with focal sialadenitis as well as reduced insulitis and fewer VEGFR2+ cells in pancreatic islets in mice on GF versus STD diet. The degree of sialadenitis was not significantly lower in GF mice, but sialadenitis and insulitis correlated strongly. Lung weight was lower in GF mice. CONCLUSION: In NOD mice, a lifelong GF diet reduces infiltration of monocytes/macrophages and T cells in salivary glands and inflammation in pancreatic islets, possibly by reducing VEGFR2, indicating that the linked autoimmune diseases, T1D and SS, may be alleviated by a GF diet.


Assuntos
Ilhotas Pancreáticas , Sialadenite , Síndrome de Sjogren , Animais , Dieta Livre de Glúten , Modelos Animais de Doenças , Inflamação , Camundongos , Camundongos Endogâmicos NOD , Glândulas Salivares
17.
Front Endocrinol (Lausanne) ; 13: 1032822, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36589856

RESUMO

Background: At diagnosis of Type 1 Diabetes (T1D), 30% of the beta cells are dormant, i.e. alive, but inactive. This could reduce beta cell destruction, as cellular stress contributes to beta cell damage. However, the beta cells, that are still active, must produce more insulin and are therefore more vulnerable. The inactive beta cells represent a potential for restoring the insulin secretion. Methods: We analyzed the expression of selected genes in islets from live, newly diagnosed T1D patients from the DiViD study and organ doners with longer duration of T1D, type 2 diabetes (T2D), or no diabetes from the nPOD study. Additionally, analysis of polymorphisms was performed on all the investigated genes. Findings: Various possibilities were considered for the inactivity of the beta cells: secretion defect, fetal state, hibernation, and insulin resistance. We analyzed genes related to the ceramide and sphingomyelin synthesis and degradation, secretion, circadian rhythm and insulin action, and found changes in T1D islets that resemble fetal dedifferentiation and asynchrony. Furthermore, we found low levels of insulin receptor mRNA in the islets. No polymorphisms were found. Interpretation: Our findings suggest a secretion defect, but also fetal dedifferentiation and desynchronization in the inactive beta cells. Together with previous evidence, that predisposing factors for T2D are also present for T1D development, we raise the idea to treat individuals with ongoing T1D development prophylactically with T2D medicine like GLP-1 receptor agonists, metformin, or others, combined with anti-inflammatory compounds, in order to reactivate the dormant beta cells, and to prevent autoimmune destruction. T2D mechanisms during T1D development should be investigated further.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Humanos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo
18.
Front Biosci (Landmark Ed) ; 27(12): 331, 2022 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-36624946

RESUMO

Particular molecules play pivotal roles in the pathogenesis of many autoimmune diseases. We suggest that the C24:0 sulfatide isoform may influence the development of type 1 diabetes (T1D). C24:0 sulfatide is a sphingolipid with a long carbon-atom chain. A C16:0 sulfatide isoform is also present in the insulin-producing beta cells of the islets of Langerhans. The C16:0 isoform exhibits chaperone activity and plays an important role in insulin production. In contrast, the C24:0 isoform may suppress the autoimmune attacks on beta cells that lead to T1D. Sphingolipid levels are reduced in individuals who later develop T1D but could be increased via dietary supplements or medication.


Assuntos
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Humanos , Sulfoglicoesfingolipídeos , Insulina , Isoformas de Proteínas
19.
J Autoimmun ; 122: 102674, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34182210

RESUMO

The etiopathogenesis of the autoimmune disease type 1 diabetes (T1D) is still largely unknown, however, both genetic and environmental factors contribute to the development of the disease. A major contact surface for environmental factors is the gastrointestinal (GI) tract, where barrier defects in T1D likely cause diabetogenic antigens to enter the body tissues, contributing to beta-cell autoimmunity. Human and animal research imply that increased intestinal permeability is an important disease determinant, although the underlying methodologies, interpretations and conclusions are diverse. In this review, an updated comprehensive overview on intestinal permeability in patients with T1D and animal models of T1D is provided in the categories: in vivo permeability, ex vivo permeability, zonulin, molecular permeability and blood markers. Across categories, there is consistency pointing towards increased intestinal permeability in T1D. In animal models of T1D, the intestinal permeability varies with age and strains implying a need for careful selection of method and experimental setup. Furthermore, dietary interventions that affect diabetes incidence in animal models does also impact the intestinal permeability, suggesting an association between increased intestinal permeability and T1D development.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Animais , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Permeabilidade
20.
Diabetologia ; 64(8): 1805-1815, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33973017

RESUMO

AIMS/HYPOTHESIS: The incidence of type 1 diabetes is increasing more rapidly than can be explained by genetic drift. Viruses may play an important role in the disease, as they seem to activate the 2'-5'-linked oligoadenylate (2'-5'A) pathway of the innate antiviral immune system. Our aim was to investigate this possibility. METHODS: Innate antiviral immune pathways were searched for type 1 diabetes-associated polymorphisms using genome-wide association study data. SNPs within ±250kb flanking regions of the transcription start site of 64 genes were examined. These pathways were also investigated for type 1 diabetes-associated RNA expression profiles using laser-dissected islets from two to five tissue sections per donor from the Diabetes Virus Detection (DiViD) study and the network of Pancreatic Organ Donors (nPOD). RESULTS: We found 27 novel SNPs in genes nominally associated with type 1 diabetes. Three of those SNPs were located upstream of the 2'-5'A pathway, namely SNP rs4767000 (p = 1.03 × 10-9, OR 1.123), rs1034687 (p = 2.16 × 10-7, OR 0.869) and rs739744 (p = 1.03 × 10-9, OR 1.123). We also identified a large group of dysregulated islet genes in relation to type 1 diabetes, of which two were novel. The most aberrant genes were a group of IFN-stimulated genes. Of those, the following distinct pathways were targeted by the dysregulation (compared with the non-diabetic control group): OAS1 increased by 111% (p < 1.00 × 10-4, 95% CI -0.43, -0.15); MX1 increased by 142% (p < 1.00 × 10-4, 95% CI -0.52, -0.22); and ISG15 increased by 197% (p = 2.00 × 10-4, 95% CI -0.68, -0.18). CONCLUSIONS/INTERPRETATION: We identified a genetic predisposition in the 2'-5'A pathway that potentially contributes to dysregulation of the innate antiviral immune system in type 1 diabetes. This study describes a potential role for the 2'-5'A pathway and other components of the innate antiviral immune system in beta cell autoimmunity.


Assuntos
Nucleotídeos de Adenina/genética , Diabetes Mellitus Tipo 1/genética , Regulação da Expressão Gênica/fisiologia , Predisposição Genética para Doença , Imunidade Inata/genética , Oligorribonucleotídeos/genética , Polimorfismo de Nucleotídeo Único/genética , Viroses/imunologia , Adulto , Antivirais/uso terapêutico , Diabetes Mellitus Tipo 1/virologia , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Viroses/tratamento farmacológico , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA