Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Paediatr ; 112(3): 522-528, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36480115

RESUMO

AIM: To determine the association of HLA class II risk with the demographic and clinical characteristics of type 1 diabetes at diagnosis. METHODS: We conducted a register-based retrospective cohort study of 4993 Finnish children (2169 girls) - diagnosed with type 1 diabetes under the age of 15 years in 2003-2016. The participants were divided into six risk groups based on their HLA DR/DQ genotype. Demographic characteristics, family history of type 1 diabetes and metabolic markers at the time of diagnosis were compared between the groups. RESULTS: In total, 4056/4993 children (81.2%) carried an HLA genotype associated with an increased risk of type 1 diabetes (risk groups 3-5), whereas 937/4993 children (18.8%) carried a HLA genotype conferring no or decreased disease risk. Children with higher HLA risk were younger at diagnosis (p < 0.001) and had a shorter duration of classical symptoms before diagnosis (p = 0.016). Subjects in the high-risk group were more likely to have a family member affected by type 1 diabetes when compared to those in the neutral risk group (11.5% vs. 8.8%, p = 0.05). CONCLUSION: Children with stronger HLA disease susceptibility are younger at their disease manifestation and have a shorter period of symptoms before diagnosis, suggesting that the HLA class II genes are associated with a more aggressive disease presentation.


Assuntos
Diabetes Mellitus Tipo 1 , Criança , Feminino , Humanos , Adolescente , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/diagnóstico , Estudos Retrospectivos , Genótipo , Fatores de Risco , Família , Predisposição Genética para Doença , Autoanticorpos
2.
Pediatr Allergy Immunol ; 33(1): e13613, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34379817

RESUMO

BACKGROUND: Decreased exposure to microbial agents in industrialized countries and urban living areas is considered as a risk factor of developing immune-mediated diseases, such as allergies and asthma. Epithelial surfaces in the gastrointestinal and respiratory tracts and in the skin constitute the primary areas in contact with the environmental microbial load. METHODS: We analyzed the levels of 30 cytokines and growth factors in serum or plasma as markers of the immune maturation in the participants in the DIABIMMUNE study from Russian Karelia (n = 60), Estonia (n = 83) and Finland (n = 89), three neighboring countries with remarkable differences in the incidences of allergies, asthma and autoimmune diseases. RESULTS: We observed an upregulation of T helper cell signature cytokines during the first 12 months of life, reflecting natural development of adaptive immune responses. During the first years of life, circulating concentrations of epidermal growth factor (EGF) were significantly higher, especially in Russian children compared with Finnish children. The children who developed IgE sensitization showed lower levels of EGF than those without such responses. CONCLUSION: Our results suggest that low circulating EGF levels associate with the risk of allergies possibly via the effects on the epithelial integrity and mucosal homeostasis.


Assuntos
Asma , Hipersensibilidade , Alérgenos , Criança , Pré-Escolar , Fator de Crescimento Epidérmico , Humanos , Hipersensibilidade/epidemiologia , Imunoglobulina E
3.
Front Immunol ; 11: 468, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32265922

RESUMO

Although gut bacterial dysbiosis is recognized as a regulator of beta-cell autoimmunity, no data is available on fungal dysbiosis in the children at the risk of type 1 diabetes (T1D). We hypothesized that the co-occurrence of fungal and bacterial dysbiosis contributes to the intestinal inflammation and autoimmune destruction of insulin-producing beta-cells in T1D. Fecal and blood samples were collected from 26 children tested positive for at least one diabetes-associated autoantibody (IAA, GADA, IA-2A or ICA) and matched autoantibody-negative children with HLA-conferred susceptibility to T1D (matched for HLA-DQB1 haplotype, age, gender and early childhood nutrition). Bacterial 16S and fungal ITS2 sequencing, and analyses of the markers of intestinal inflammation, namely fecal human beta-defensin-2 (HBD2), calprotectin and secretory total IgA, were performed. Anti-Saccharomyces cerevisiae antibodies (ASCA) and circulating cytokines, IFNG, IL-17 and IL-22, were studied. After these analyses, the children were followed for development of clinical T1D (median 8 years and 8 months). Nine autoantibody positive children were diagnosed with T1D, whereas none of the autoantibody negative children developed T1D during the follow-up. Fungal dysbiosis, characterized by high abundance of fecal Saccharomyces and Candida, was found in the progressors, i.e., children with beta-cell autoimmunity who during the follow-up progressed to clinical T1D. These children showed also bacterial dysbiosis, i.e., increased Bacteroidales and Clostridiales ratio, which was, however, found also in the non-progressors, and is thus a common nominator in the children with beta-cell autoimmunity. Furthermore, the progressors showed markers of intestinal inflammation detected as increased levels of fecal HBD2 and ASCA IgG to fungal antigens. We conclude that the fungal and bacterial dysbiosis, and intestinal inflammation are associated with the development of T1D in children with beta-cell autoimmunity.


Assuntos
Candida/fisiologia , Diabetes Mellitus Tipo 1/imunologia , Fezes/microbiologia , Células Secretoras de Insulina/imunologia , Micoses/imunologia , Saccharomyces/fisiologia , Adolescente , Anticorpos Antifúngicos/sangue , Autoanticorpos/sangue , Autoimunidade , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/epidemiologia , Disbiose , Fezes/química , Feminino , Finlândia/epidemiologia , Cadeias beta de HLA-DQ/genética , Humanos , Células Secretoras de Insulina/patologia , Masculino , Micoses/epidemiologia , beta-Defensinas/análise
5.
Nat Microbiol ; 4(3): 470-479, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30559407

RESUMO

The human gut microbiome matures towards the adult composition during the first years of life and is implicated in early immune development. Here, we investigate the effects of microbial genomic diversity on gut microbiome development using integrated early childhood data sets collected in the DIABIMMUNE study in Finland, Estonia and Russian Karelia. We show that gut microbial diversity is associated with household location and linear growth of children. Single nucleotide polymorphism- and metagenomic assembly-based strain tracking revealed large and highly dynamic microbial pangenomes, especially in the genus Bacteroides, in which we identified evidence of variability deriving from Bacteroides-targeting bacteriophages. Our analyses revealed functional consequences of strain diversity; only 10% of Finnish infants harboured Bifidobacterium longum subsp. infantis, a subspecies specialized in human milk metabolism, whereas Russian infants commonly maintained a probiotic Bifidobacterium bifidum strain in infancy. Groups of bacteria contributing to diverse, characterized metabolic pathways converged to highly subject-specific configurations over the first two years of life. This longitudinal study extends the current view of early gut microbial community assembly based on strain-level genomic variation.


Assuntos
Adaptação Fisiológica , Microbioma Gastrointestinal/genética , Variação Genética , Genoma Bacteriano , Fatores Etários , Bacteriófagos/genética , Bacteroides/genética , Bacteroides/virologia , Bifidobacterium bifidum/genética , Bifidobacterium longum/genética , Desenvolvimento Infantil , Pré-Escolar , Estônia , Fezes/microbiologia , Feminino , Finlândia , Humanos , Lactente , Estudos Longitudinais , Masculino , Redes e Vias Metabólicas , Metagenômica , Polimorfismo de Nucleotídeo Único , Probióticos , Federação Russa
6.
Scand J Gastroenterol ; 53(4): 403-409, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29504486

RESUMO

OBJECTIVES: Several studies have reported that the intestinal microbiota composition of celiac disease (CD) patients differs from healthy individuals. The possible role of gut microbiota in the pathogenesis of the disease is, however, not known. Here, we aimed to assess the possible differences in early fecal microbiota composition between children that later developed CD and healthy controls matched for age, sex and HLA risk genotype. MATERIALS AND METHODS: We used 16S rRNA gene sequencing to examine the fecal microbiota of 27 children with high genetic risk of developing CD. Nine of these children developed the disease by the age of 4 years. Stool samples were collected at the age of 9 and 12 months, before any of the children had developed CD. The fecal microbiota composition of children who later developed the disease was compared with the microbiota of the children who did not have CD or associated autoantibodies at the age of 4 years. Delivery mode, early nutrition, and use of antibiotics were taken into account in the analyses. RESULTS: No statistically significant differences in the fecal microbiota composition were found between children who later developed CD (n = 9) and the control children without disease or associated autoantibodies (n = 18). CONCLUSIONS: Based on our results, the fecal microbiota composition at the age of 9 and 12 months is not associated with the development of CD. Our results, however, do not exclude the possibility of duodenal microbiota changes or a later microbiota-related trigger for the disease.


Assuntos
Doença Celíaca/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal/genética , RNA Ribossômico 16S/análise , Autoanticorpos/sangue , Autoimunidade , Estudos de Casos e Controles , Doença Celíaca/genética , Pré-Escolar , Duodeno/microbiologia , Feminino , Finlândia , Humanos , Lactente , Metagenoma
7.
Semin Immunopathol ; 39(6): 653-667, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29170800

RESUMO

Type 1 diabetes (T1D) is perceived as a chronic immune-mediated disease with a subclinical prodromal period characterized by selective loss of insulin-producing beta cells in the pancreatic islets in genetically susceptible subjects. The incidence of T1D has increased manifold in most developed countries after World War II in parallel with a series of other immune-mediated diseases. T1D results from gene-environmental interactions. The appearance of disease-associated autoantibodies into the peripheral circulation is the first detectable sign of the initiation of the disease process leading to clinical T1D. The first autoantibodies may appear already before the age of 6 months and the seroconversion rate peaks during the second year of life. This implies that exogenous factors involved in the pathogenesis of T1D must be operative in early life, some of them most likely already during pregnancy. Here, we discuss putative endogenous factors that may contribute to the development of T1D during fetal and early postnatal life. Many environmental factors operative in early life have been implicated in the pathogenesis of T1D, but relatively few have been firmly confirmed.


Assuntos
Diabetes Mellitus Tipo 1/etiologia , Autoanticorpos/imunologia , Autoimunidade , Diabetes Mellitus Tipo 1/metabolismo , Progressão da Doença , Suscetibilidade a Doenças , Meio Ambiente , Feminino , Feto , Humanos , Recém-Nascido , Ilhotas Pancreáticas/imunologia , Gravidez , Fatores de Risco
8.
Gastroenterology ; 153(4): 961-970.e3, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28687275

RESUMO

BACKGROUND & AIMS: Feeding during the first months of life might affect risk for celiac disease. Individuals with celiac disease or type 1 diabetes have been reported to have high titers of antibodies against cow's milk proteins. Avoidance of cow's milk-based formula for infants with genetic susceptibility for type 1 diabetes reduced the cumulative incidence of diabetes-associated autoantibodies. We performed a randomized controlled trial in the same population to study whether weaning to an extensively hydrolyzed formula reduced the risk of celiac disease autoimmunity or celiac disease. METHODS: We performed a double-blind controlled trial of 230 infants with HLA-defined predisposition to type 1 diabetes and at least 1 family member with type 1 diabetes. The infants were randomly assigned to groups fed a casein hydrolysate formula (n = 113) or a conventional formula (control, n = 117) whenever breast milk was not available during the first 6-8 months of life. Serum samples were collected over a median time period of 10 years and analyzed for antibodies to tissue transglutaminase (anti-TG2A) using a radiobinding assay, to endomysium using an immunofluorescence assay, and antibodies to a deamidated gliadine peptide using an immunofluorometry assay. Duodenal biopsies were collected if levels of anti-TG2A exceeded 20 relative units. Cow's milk antibodies were measured during the first 2 years of life. RESULTS: Of the 189 participants analyzed for anti-TG2A, 25 (13.2%) tested positive. Of the 230 study participants observed, 10 (4.3%) were diagnosed with celiac disease. We did not find any significant differences at the cumulative incidence of anti-TG2A positivity (hazard ratio, 1.14; 95% confidence interval, 0.51-2.54) or celiac disease (hazard ratio, 4.13; 95% confidence interval, 0.81-21.02) between the casein hydrolysate and cow's milk groups. Children who developed celiac disease had increased titers of cow's milk antibodies before the appearance of anti-TG2A or celiac disease. CONCLUSIONS: In a randomized controlled trial of 230 infants with genetic risk factors for celiac disease, we did not find evidence that weaning to a diet of extensively hydrolyzed formula compared with cow's milk-based formula would decrease the risk for celiac disease later in life. Increased titers of cow's milk antibody before anti-TG2A and celiac disease indicates that subjects with celiac disease might have increased intestinal permeability in early life. ClinicalTrials.gov Number: NCT00570102.


Assuntos
Autoanticorpos/sangue , Autoimunidade , Caseínas/uso terapêutico , Doença Celíaca/prevenção & controle , Diabetes Mellitus Tipo 1/imunologia , Proteínas de Ligação ao GTP/imunologia , Fórmulas Infantis/efeitos adversos , Hipersensibilidade a Leite/prevenção & controle , Proteínas do Leite/efeitos adversos , Transglutaminases/imunologia , Biópsia , Caseínas/efeitos adversos , Caseínas/imunologia , Doença Celíaca/diagnóstico , Doença Celíaca/imunologia , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/genética , Método Duplo-Cego , Duodeno/imunologia , Duodeno/patologia , Finlândia , Gliadina/imunologia , Humanos , Lactente , Hipersensibilidade a Leite/diagnóstico , Hipersensibilidade a Leite/imunologia , Proteínas do Leite/imunologia , Proteína 2 Glutamina gama-Glutamiltransferase , Medição de Risco , Fatores de Risco , Testes Sorológicos , Fatores de Tempo , Resultado do Tratamento
9.
J Immunol ; 194(1): 68-75, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480564

RESUMO

Upregulation of IL-17 immunity and detrimental effects of IL-17 on human islets have been implicated in human type 1 diabetes. In animal models, the plasticity of Th1/Th17 cells contributes to the development of autoimmune diabetes. In this study, we demonstrate that the upregulation of the IL-17 pathway and Th1/Th17 plasticity in peripheral blood are markers of advanced ß cell autoimmunity and impaired ß cell function in human type 1 diabetes. Activated Th17 immunity was observed in the late stage of preclinical diabetes in children with ß cell autoimmunity and impaired glucose tolerance, but not in children with early ß cell autoimmunity. We found an increased ratio of IFN-γ/IL-17 expression in Th17 cells in children with advanced ß cell autoimmunity, which correlated with HbA1c and plasma glucose concentrations in an oral glucose tolerance test, and thus impaired ß cell function. Low expression of Helios was seen in Th17 cells, suggesting that Th1/Th17 cells are not converted thymus-derived regulatory T cells. Our results suggest that the development of Th1/Th17 plasticity may serve as a biomarker of disease progression from ß cell autoantibody positivity to type 1 diabetes. These data in human type 1 diabetes emphasize the role of Th1/Th17 plasticity as a potential contributor to tissue destruction in autoimmune conditions.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Intolerância à Glucose/imunologia , Células Secretoras de Insulina/imunologia , Interleucina-17/biossíntese , Células Th1/imunologia , Células Th17/imunologia , Autoanticorpos/imunologia , Autoimunidade/imunologia , Biomarcadores/sangue , Células Cultivadas , Criança , Pré-Escolar , Progressão da Doença , Feminino , Humanos , Fator de Transcrição Ikaros/biossíntese , Interferon gama/biossíntese , Interleucina-17/imunologia , Interleucina-9/biossíntese , Interleucina-9/imunologia , Masculino , Linfócitos T Reguladores/imunologia , Regulação para Cima/imunologia
10.
Diabetes ; 62(4): 1238-44, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23274889

RESUMO

The role of the intestinal microbiota as a regulator of autoimmune diabetes in animal models is well-established, but data on human type 1 diabetes are tentative and based on studies including only a few study subjects. To exclude secondary effects of diabetes and HLA risk genotype on gut microbiota, we compared the intestinal microbiota composition in children with at least two diabetes-associated autoantibodies (n = 18) with autoantibody-negative children matched for age, sex, early feeding history, and HLA risk genotype using pyrosequencing. Principal component analysis indicated that a low abundance of lactate-producing and butyrate-producing species was associated with ß-cell autoimmunity. In addition, a dearth of the two most dominant Bifidobacterium species, Bifidobacterium adolescentis and Bifidobacterium pseudocatenulatum, and an increased abundance of the Bacteroides genus were observed in the children with ß-cell autoimmunity. We did not find increased fecal calprotectin or IgA as marker of inflammation in children with ß-cell autoimmunity. Functional studies related to the observed alterations in the gut microbiome are warranted because the low abundance of bifidobacteria and butyrate-producing species could adversely affect the intestinal epithelial barrier function and inflammation, whereas the apparent importance of the Bacteroides genus in development of type 1 diabetes is insufficiently understood.


Assuntos
Autoanticorpos/fisiologia , Bactérias/classificação , Diabetes Mellitus Tipo 1/imunologia , Fezes/microbiologia , Células Secretoras de Insulina/imunologia , Adolescente , Autoanticorpos/genética , Bactérias/isolamento & purificação , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/genética , Feminino , Regulação da Expressão Gênica/imunologia , Variação Genética , Genótipo , Cadeias beta de HLA-DQ/genética , Cadeias beta de HLA-DQ/metabolismo , Humanos , Masculino
11.
Pediatr Diabetes ; 13(5): 400-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22332874

RESUMO

BACKGROUND: Reduced risk for type 1 diabetes (T1D) has been reported in the offspring of mothers with T1D when compared with children of affected fathers. OBJECTIVE: To evaluate the hypothesis that exposure of the offspring to maternal insulin therapy induces regulatory mechanisms in utero, we compared the FOXP3 expressing regulatory T cells in cord blood (CB) of infants born to mothers with or without T1D. SUBJECTS AND METHODS: Cord blood mononuclear cells (CBMCs) from 20 infants with maternal T1D and from 20 infants with an unaffected mother were analyzed for the numbers of CD4+CD25+FOXP3+ cells ex vivo and after in vitro stimulation with human insulin by flow cytometry. The mRNA expression of FOXP3, NFATc2, STIM1, interleukin (IL)-10, and transforming growth factor (TGF)-ß was measured by real-time reverse transcription polymerase chain reaction. RESULTS: The percentage of FOXP3+ cells in CD4+CD25(high) cells was higher in the CB of the infants with maternal T1D when compared with the infants of unaffected mothers (p = 0.023). After in vitro insulin stimulation an increase in the percentage of FOXP3+ cells in CD4+CD25(high) cells (p = 0.0002) as well as upregulation of FOXP3, NFATc2, STIM1, IL-10, and TGF-ß transcripts in CBMCs (p < 0.013 for all; Wilcoxon test) was observed only in the offspring of mothers with T1D, in whom the disease-related PTPN22 allele was associated with reduced STIM1 and NFATc2 response in insulin-stimulated CBMCs (p = 0.007 and p = 0.014). CONCLUSIONS: We suggest that maternal insulin treatment induces expansion of regulatory T cells in the fetus, which might contribute to the lower risk of diabetes in children with maternal vs. paternal diabetes.


Assuntos
Antígenos CD4/sangue , Diabetes Mellitus Tipo 1/imunologia , Fatores de Transcrição Forkhead/sangue , Subunidade alfa de Receptor de Interleucina-2/sangue , Linfócitos T Reguladores/imunologia , Diabetes Mellitus Tipo 1/tratamento farmacológico , Feminino , Sangue Fetal/imunologia , Humanos , Lactente , Recém-Nascido , Insulina/uso terapêutico , Masculino , Mães
12.
J Immunol ; 185(3): 1959-67, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20592279

RESUMO

Th17 immunity has been shown to regulate autoimmune diabetes in mice. IL-17 neutralization prevented development of diabetes when given postinitiation of insulitis but not earlier, suggesting interference with the effector phase of the disease. Islet-cell Ag-specific Th17 cells converted into IFN-gamma-secreting Th1-like cells and caused diabetes in mice recipients. The role of IL-17 in human type 1 diabetes (T1D) is, however, not established. In this study, we show upregulation of Th17 immunity in peripheral blood T cells from children with T1D. This was characterized by increased IL-17 secretion and expression of IL-17, IL-22, and retinoic acid-related orphan receptor C isoform 2, but also FOXP3 transcripts upon T cell activation in vitro. Also, circulating memory CD4 cells from children with T1D showed the same pattern of IL-17, IL-22 and FOXP3 mRNA upregulation, indicating IL-17 pathway activation in vivo. IL-17-positive T cells appeared to be CD4(+) cells expressing TCR-alphabeta and CCR6, and a subpopulation showed coproduction of IFN-gamma. Given the Th17 immunity in T1D, we demonstrated that IL-17 had detrimental effects on human islet cells in vitro; it potentiated both inflammatory and proapoptotic responses. Our findings highlight the role of IL-17 immunity in the pathogenesis of human T1D and point to a potential therapeutic strategy.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Interleucina-17/fisiologia , Regulação para Cima/imunologia , Adolescente , Animais , Células Cultivadas , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/patologia , Feminino , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Humanos , Lactente , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Interleucina-17/efeitos adversos , Interleucina-17/biossíntese , Interleucina-17/metabolismo , Interleucinas/biossíntese , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , RNA Mensageiro/biossíntese , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Interleucina 22
13.
Pediatr Diabetes ; 9(5): 434-41, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18503496

RESUMO

BACKGROUND: Early exposure to cow's milk (CM) proteins have been implicated in the pathogenesis of type 1 diabetes (T1D). OBJECTIVE: We analyzed the development of the humoral immune response to dietary CM proteins in early childhood and its relation to later T1D. SUBJECTS AND METHODS: We studied a subgroup of 94 children randomized to be weaned to a CM-based infant formula in the trial to reduce insulin-dependent diabetes mellitus in the genetically at risk (TRIGR) pilot study. All subjects carried human leukocyte antigen-conferred T1D susceptibility and had an affected first-degree relative. After 7 years of follow-up, 8 subjects had progressed to T1D, 15 had at least one disease-associated autoantibody, and 71 remained autoantibody negative (controls). Immunoglobulin (Ig) G and IgA class antibodies to whole CM formula, beta-lactoglobulin (BLG), bovine serum albumin, and alpha-casein and IgG antibodies to bovine insulin (BI) were measured with enzyme-linked immunosorbent assays from sequential samples. RESULTS: The children with later T1D showed increased IgG levels to BLG from 3 to 18 months of age (p = 0.028) and enhanced IgA levels to CM formula at the age of 9 months (p = 0.022) compared with controls. In the children with an affected father or sibling, IgG antibodies to BI were higher in autoantibody-positive subjects than in autoantibody-negative subjects at 18 months of age (p = 0.022). CONCLUSION: An enhanced humoral immune response to various CM proteins in infancy is seen in a subgroup of those children who later progress to T1D. Accordingly, a dysregulated immune response to oral antigens is an early event in the pathogenesis of T1D.


Assuntos
Autoanticorpos/análise , Diabetes Mellitus Tipo 1/etiologia , Proteínas do Leite/imunologia , Leite/imunologia , Animais , Aleitamento Materno , Bovinos , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/imunologia , Feminino , Predisposição Genética para Doença , Antígenos HLA-DQ/genética , Cadeias beta de HLA-DQ , Humanos , Imunoglobulina A/análise , Imunoglobulina G/análise , Lactente , Fórmulas Infantis , Anticorpos Anti-Insulina/análise , Masculino , Fatores de Risco , Toxoide Tetânico/imunologia
14.
Immunology ; 121(2): 189-96, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17244154

RESUMO

Aberrancies in T-cell polarization including expression of chemokine receptors have been reported in human leucocyte antigen (HLA) class II associated autoimmune diseases, such as type 1 diabetes (T1D) and rheumatoid arthritis. We asked whether these aberrancies are present at birth in newborn infants carrying the HLA risk haplotypes for T1D. Sixty-seven cord blood (CB) samples from infants were screened for T1D-associated HLA risk genotypes (HLA-DR4-DQ8 and/or DR3-DQ2 without protective alleles). CB lymphocytes were stimulated with phytohaemagglutinin in type 1 (interleukin (IL)-12, anti-IL4) or type 2 (IL-4, anti-IL12) cytokine environment for 6 days. The expression of chemokine and cytokine receptors on T cells was determined by flow cytometry, secretion of cytokines was analysed with enzyme-linked immunosorbent assay, and transcription factors were analysed using real-time reverse transcriptase-polymerase chain reaction. After culture of CB lymphocytes in type 2 cytokine environment newborn infants carrying DR4-DQ8 haplotype (n=18) showed reduced percentage of CD4 T cells expressing CCR4 (P=0 x 009) and the level of CCR4 mRNA was decreased (P=0 x 008). In addition, lower secretion of IL-13 and expression of GATA-3 in CB lymphocytes cultured in type 2 cytokine environment were found in the infants with DR4-DQ8 haplotype (P=0 x 020 and P=0 x 004, respectively) in comparison to newborn infants without DR4-DQ8 and DR3-DQ2 haplotypes (n=37). Poor in vitro induction of type 2 immune responses in newborn infants with DR4-DQ8 haplotype suggests that the HLA genotype associated with risk of autoimmunity may affect the T cell polarization already at birth, which in turn may contribute to the risk for autoimmunity later in life.


Assuntos
Citocinas/imunologia , Diabetes Mellitus Tipo 1/imunologia , Sangue Fetal/imunologia , Subpopulações de Linfócitos T/imunologia , Polaridade Celular/imunologia , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Fator de Transcrição GATA3/biossíntese , Predisposição Genética para Doença , Antígenos HLA-DQ/genética , Antígeno HLA-DR4/genética , Teste de Histocompatibilidade , Humanos , Recém-Nascido , Interleucina-13/biossíntese , RNA Mensageiro/genética , Receptores CCR4 , Receptores CXCR4/biossíntese , Receptores CXCR4/sangue , Receptores de Quimiocinas/biossíntese , Receptores de Quimiocinas/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...