Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
Más filtros

Medicinas Complementárias
Intervalo de año de publicación
1.
Eur J Nutr ; 63(4): 1329-1338, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38413484

RESUMEN

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.


Asunto(s)
Autoanticuerpos , Autoinmunidad , Diabetes Mellitus Tipo 1 , Islotes Pancreáticos , Complejo Vitamínico B , Humanos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/epidemiología , Masculino , Femenino , Complejo Vitamínico B/administración & dosificación , Estudios Prospectivos , Niño , Preescolar , Lactante , Islotes Pancreáticos/inmunología , Autoanticuerpos/sangre , Factores de Riesgo , Dieta/métodos , Dieta/estadística & datos numéricos , Modelos de Riesgos Proporcionales , Estados Unidos/epidemiología , Finlandia/epidemiología , Suecia/epidemiología , Alemania/epidemiología , Suplementos Dietéticos , Cohorte de Nacimiento , Progresión de la Enfermedad
2.
Nutrients ; 14(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35057548

RESUMEN

Maternal dietary micronutrients and omega-3 fatty acids support development of the fetal and neonatal immune system. Whether supplementation is similarly beneficial for the mother during gestation has received limited attention. A scoping review of human trials was conducted looking for evidence of biochemical, genomic, and clinical effects of supplementation on the maternal immune system. The authors explored the literature on PubMed, Cochrane Library, and Web of Science databases from 2010 to the present day using PRISMA-ScR methodology. Full-length human trials in English were searched for using general terms and vitamin A, B12, C, D, and E; choline; iodine; iron; selenium; zinc; and docosahexaenoic/eicosapentaenoic acid. Of 1391 unique articles, 36 were eligible for inclusion. Diverse biochemical and epigenomic effects of supplementation were identified that may influence innate and adaptive immunity. Possible clinical benefits were encountered in malaria, HIV infections, anemia, Type 1 diabetes mellitus, and preventing preterm delivery. Only limited publications were identified that directly explored maternal immunity in pregnancy and the effects of micronutrients. None provided a holistic perspective. It is concluded that supplementation may influence biochemical aspects of the maternal immune response and some clinical outcomes, but the evidence from this review is not sufficient to justify changes to current guidelines.


Asunto(s)
Ácidos Grasos Omega-3/administración & dosificación , Sistema Inmunológico/efectos de los fármacos , Salud Materna , Micronutrientes/administración & dosificación , Fenómenos Fisiologicos de la Nutrición Prenatal , Adulto , Anemia/inmunología , Colina/administración & dosificación , Diabetes Mellitus Tipo 1/inmunología , Suplementos Dietéticos , Femenino , Infecciones por VIH/inmunología , Humanos , Yodo/administración & dosificación , Hierro/administración & dosificación , Madres , Embarazo , Selenio/administración & dosificación , Oligoelementos/administración & dosificación , Vitaminas/administración & dosificación , Zinc/administración & dosificación
3.
Am J Physiol Endocrinol Metab ; 322(1): E74-E84, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34779254

RESUMEN

Type 1 diabetes (T1D) is a chronic autoimmune disease accompanied by the immune-mediated destruction of pancreatic ß-cells. In this study, we aimed to explore the regulatory effects of vitamin D (VD) supplementation on pancreatic ß-cell function by altering the expression of bioinformatically identified cathepsin G (CatG) in T1D mice. A T1D mouse model was established in nonobese diabetic (NOD) mice, and their islets were isolated and purified. Pancreatic mononuclear cells (MNCs) were collected, from which CD4+ T cells were isolated. The levels of interleukin (IL)-2, IL-10, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) in the supernatant of mouse pancreatic tissue homogenate were assessed using ELISA. Immunohistochemistry and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labelin (TUNEL) staining were conducted to evaluate the effects of VD supplementation on pancreatic tissues of T1D mice. The pancreatic ß-cell line MIN6 was used for in vitro substantiation of findings in vivo. VD supplementation reduced glucose levels and improved glucose tolerance in T1D mice. Furthermore, VD supplementation improved pancreatic ß-cell function and suppressed immunological and inflammatory reactions in the T1D mice. We documented overexpression of CatG in diabetes tissue samples, and then showed that VD supplementation normalized the islet immune microenvironment through downregulating CatG expression in T1D mice. Experiments in vitro subsequently demonstrated that VD supplementation impeded CD4+ T activation by downregulating CatG expression and thereby enhanced pancreatic ß-cell function. Results of the present study elucidated that VD supplementation can downregulate the expression of CatG and inhibit CD4+ T cell activation, thereby improving ß-cell function in T1D.NEW & NOTEWORTHY We report that vitamin D (VD) supplementation downregulates CatG expression and inhibits CD4+ T cell activation, thereby improving ß-cell function in type 1 diabetes (T1D). This study deepens our understanding of the pathogenesis of T1D and clarifies molecular events underlying the alleviatory effect of VD for immunotherapy against T1D.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Catepsina G/metabolismo , Diabetes Mellitus Tipo 1/dietoterapia , Diabetes Mellitus Tipo 1/inmunología , Suplementos Dietéticos , Inmunosupresores/administración & dosificación , Células Secretoras de Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Vitamina D/administración & dosificación , Animales , Catepsina G/genética , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Secretoras de Insulina/inmunología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal/genética
4.
Nutrients ; 13(11)2021 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-34836227

RESUMEN

The physiological markers of autoimmune diabetes include functional disorders of the antioxidative system as well as progressing inflammation and the presence of autoantibodies. Even though people with type 1 diabetes show genetic predispositions facilitating the onset of the disease, it is believed that dietary factors can stimulate the initiation and progression of the disease. This paper analyses the possibility of using tea as an element of diet therapy in the treatment of type 1 diabetes. Based on information available in literature covering the last 10 years, the impact of regular tea consumption or diet supplements containing tea polyphenols on the oxidative status as well as inflammatory and autoimmune response of the organism was analyzed. Studies conducted on laboratory animals, human patients, and in vitro revealed positive effects of the consumption of tea or polyphenols isolated therefrom on the diabetic body. Few reports available in the literature pertain to the impact of tea on organisms affected by type 1 diabetes as most (over 85%) have focused on cases of type 2 diabetes. It has been concluded that by introducing tea into the diet, it is possible to alleviate some of the consequences of oxidative stress and inflammation, thus limiting their destructive impact on the patients' organisms, consequently improving their quality of life, regardless of the type of diabetes. Furthermore, elimination of inflammation should reduce the incidence of immune response. One should consider more widespread promotion of tea consumption by individuals genetically predisposed to diabetes, especially considering the drink's low price, easy availability, overall benefits to human health, and above all, the fact that it can be safely used over extended periods of time, regardless of the patient's age.


Asunto(s)
Antiinflamatorios no Esteroideos , Antioxidantes , Diabetes Mellitus Tipo 1/dietoterapia , Inmunomodulación , , Animales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/fisiopatología , Humanos , Inflamación/dietoterapia , Estrés Oxidativo , Polifenoles
5.
Drug Des Devel Ther ; 15: 2629-2639, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34168434

RESUMEN

BACKGROUND: Acanthopanax trifoliatus (L.) Merr. is a medicinal plant found in Southeast Asia, and its young leaves and shoots are consumed as a vegetable. The main bioactive components of this herb are polysaccharides that have significant anti-diabetic effects. The aim of this study was to evaluate the immunoregulatory effect of A. trifoliatus (L.) Merr. polysaccharide (ATMP) on a mouse model of type 1 diabetes mellitus (T1DM). METHODS: The monosaccharide composition and mean molecular mass of ATMP were determined by HPLC and HPGPC. T1DM was induced in mice using STZ, and 35, 70 and 140mg/kg ATMP was administered daily via the intragastric route for six weeks. Untreated and metformin-treated positive control groups were also included. The body weight of the mice, food and water intake and fasting glucose levels were monitored throughout the 6-week regimen. Histological changes in the pancreas and spleen were analyzed by H&E staining. Oral glucose tolerance was evaluated with the appropriate test. Peroxisome proliferator-activated receptor γ (PPARγ) mRNA and protein levels in the spleen were measured by quantitative real time PCR and Western blotting. IL-10, IFN-γ and insulin levels in the sera were determined by ELISA. The CD4+ and CD8+T cells in spleen tissues were detected by immunohistochemistry (IHC). RESULTS: ATMP and metformin significantly decreased fasting blood glucose, and the food and water intake after 6 weeks of treatment. In contrast, serum insulin levels, glucose tolerance and body weight improved considerably in the high and medium-dose ATMP and metformin groups. T1DM was associated with pancreatic and splenic tissue damage. The high dose (140mg/kg) of ATMP reduced infiltration of inflammatory cells into the pancreas and restored the structure of islet ß-cells in the diabetic mice. Consistent with this, 35, 70 and 140mg/kg ATMP increased IL-10 levels and decreased that of IFN-γ, thereby restoring the CD4+/CD8+ and Th1/Th2 cytokine ratio. At the molecular level, high-dose ATMP up-regulated PPARγ in the splenic cells. CONCLUSION: ATMP exerts a hypoglycemic effect in diabetic mice by restoring the immune balance in the spleen.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Eleutherococcus/química , Polisacáridos/farmacología , Animales , Cromatografía Líquida de Alta Presión , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inmunología , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/aislamiento & purificación , Hipoglucemiantes/farmacología , Insulina/metabolismo , Masculino , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , Polisacáridos/aislamiento & purificación , Bazo/efectos de los fármacos , Bazo/inmunología
6.
Nutrients ; 13(3)2021 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-33805588

RESUMEN

Our aim was to study the associations between maternal vitamin C and iron intake during pregnancy and the offspring's risk of developing islet autoimmunity and type 1 diabetes. The study was a part of the Finnish Type 1 Diabetes Prediction and Prevention (DIPP) prospective birth cohort including children genetically at risk of type 1 diabetes born between 1997-2004. The diets of 4879 mothers in late pregnancy were assessed with a validated food frequency questionnaire. The outcomes were islet autoimmunity and type 1 diabetes. Cox proportional hazards regression analysis adjusted for energy, family history of diabetes, human leukocyte antigen (HLA) genotype and sex was used for statistical analyses. Total intake of vitamin C or iron from food and supplements was not associated with the risk of islet autoimmunity (vitamin C: HR 0.91: 95% CI (0.80, 1.03), iron: 0.98 (0.87, 1.10)) or type 1 diabetes (vitamin C: 1.01 (0.87, 1.17), iron: 0.92 (0.78, 1.08)), neither was the use of vitamin C or iron supplements associated with the outcomes. In conclusion, no association was found between maternal vitamin C or iron intake during pregnancy and the risk of islet autoimmunity or type 1 diabetes in the offspring.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diabetes Mellitus Tipo 1/inmunología , Dieta/efectos adversos , Exposición Materna/efectos adversos , Fenómenos Fisiologicos Nutricionales Maternos/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Adulto , Ácido Ascórbico/análisis , Enfermedades Autoinmunes/genética , Preescolar , Diabetes Mellitus Tipo 1/genética , Dieta/estadística & datos numéricos , Encuestas sobre Dietas , Suplementos Dietéticos , Femenino , Finlandia , Genotipo , Antígenos HLA/inmunología , Humanos , Lactante , Hierro de la Dieta/análisis , Islotes Pancreáticos/inmunología , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Análisis de Regresión
7.
Clin Sci (Lond) ; 135(1): 19-34, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33399849

RESUMEN

Type 1 diabetes mellitus (T1D) is a chronic autoimmune disease characterized by insulin-producing pancreatic ß-cell destruction and hyperglycemia. While monocytes and NOD-like receptor family-pyrin domain containing 3 (NLRP3) are associated with T1D onset and development, the specific receptors and factors involved in NLRP3 inflammasome activation remain unknown. Herein, we evaluated the inflammatory state of resident peritoneal macrophages (PMs) from genetically modified non-obese diabetic (NOD), NLRP3-KO, wild-type (WT) mice and in peripheral blood mononuclear cells (PBMCs) from human T1D patients. We also assessed the effect of docosahexaenoic acid (DHA) on the inflammatory status. Macrophages from STZ-induced T1D mice exhibited increased inflammatory cytokine/chemokine levels, nitric oxide (NO) secretion, NLRP3 and iNOS protein levels, and augmented glycolytic activity compared to control animals. In PMs from NOD and STZ-induced T1D mice, DHA reduced NO production and attenuated the inflammatory state. Furthermore, iNOS and IL-1ß protein expression levels and NO production were lower in the PMs from diabetic NLRP3-KO mice than from WT mice. We also observed increased IL-1ß secretion in PBMCs from T1D patients and immortalized murine macrophages treated with advanced glycation end products and palmitic acid. The present study demonstrated that the resident PMs are in a proinflammatory state characterized by increased NLRP3/iNOS pathway-mediated NO production, up-regulated proinflammatory cytokine/chemokine receptor expression and altered glycolytic activity. Notably, ex vivo treatment with DHA reverted the diabetes-induced changes and attenuated the macrophage inflammatory state. It is plausible that DHA supplementation could be employed as adjuvant therapy for treating individuals with T1D.


Asunto(s)
Antiinflamatorios/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Inflamación/tratamiento farmacológico , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Adulto , Animales , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Femenino , Humanos , Inflamación/inducido químicamente , Inflamación/enzimología , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Embarazo , Transducción de Señal , Estreptozocina
8.
Best Pract Res Clin Endocrinol Metab ; 34(1): 101412, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32265102

RESUMEN

An adjuvant is an immunological or pharmacological substance or group of substances that can be added to a given agent to enhance its effect in terms of efficacy, effectiveness and potency. Different mechanisms have been hypothesized underlying the action of the adjuvant, including boosting immune (innate and adaptive) response: this generally results in sparing the necessary amount of the agent and can potentially reduce the frequency of the needed number of therapeutic interventions. Adjuvants can be commonly found in vaccines, immunization products, mineral oils, cosmetics, silicone breast implants and other therapeutic/medical devices, being usually safe and effective. However, in a fraction of genetically susceptible and predisposed subjects, the administration of adjuvants may lead to the insurgence of serious side-effects, called "autoimmune/inflammatory syndrome by adjuvants" (ASIA) or Shoenfeld's syndrome. The present review is aimed at focusing on the "endocrine pebbles" of the mosaic of autoimmunity and of the ASIA syndrome, collecting together 54 cases of sub-acute thyroiditis, 2 cases of Hashimoto's thyroiditis, 11 cases of primary ovarian failure/primary ovarian insufficiency, 13 cases of autoimmune diabetes type 1, and 1 case of autoimmune adrenal gland insufficiency occurred after exposure to adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/efectos adversos , Enfermedades Autoinmunes/inducido químicamente , Autoinmunidad/efectos de los fármacos , Enfermedades del Sistema Endocrino/inducido químicamente , Enfermedades Autoinmunes/genética , Autoinmunidad/genética , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Enfermedades del Sistema Endocrino/genética , Enfermedades del Sistema Endocrino/inmunología , Predisposición Genética a la Enfermedad , Humanos , Factores de Riesgo , Síndrome
9.
Methods Mol Biol ; 2128: 69-85, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180186

RESUMEN

Rat models of human type 1 diabetes have been shown to be of great importance for the elucidation of the mechanisms underlying the development of autoimmune diabetes. The three major well-established spontaneous rat models are the BioBreeding (BB) diabetes-prone rat, the Komeda diabetes-prone (KDP) rat, and the IDDM (LEW.1AR1-iddm) rat. Their distinctive features are described with special reference to their pathology, immunology, and genetics and compared with the situation in patients with type 1 diabetes mellitus. For all three established rat models, a distinctive genetic mutation has been identified that is responsible for the manifestation of the diabetic syndrome in these rat strains.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Edad de Inicio , Animales , Citocinas/inmunología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Sistema Digestivo/inmunología , Sistema Digestivo/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Masculino , Ratas , Ratas Endogámicas , Selección Artificial/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Int J Mol Sci ; 21(5)2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32111075

RESUMEN

Autoantigen treatment has been tried for the prevention of type 1 diabetes (T1D) and to preserve residual beta-cell function in patients with a recent onset of the disease. In experimental animal models, efficacy was good, but was insufficient in human subjects. Besides the possible minor efficacy of peroral insulin in high-risk individuals to prevent T1D, autoantigen prevention trials have failed. Other studies on autoantigen prevention and intervention at diagnosis are ongoing. One problem is to select autoantigen/s; others are dose and route. Oral administration may be improved by using different vehicles. Proinsulin peptide therapy in patients with T1D has shown possible minor efficacy. In patients with newly diagnosed T1D, subcutaneous injection of glutamic acid decarboxylase (GAD) bound to alum hydroxide (GAD-alum) can likely preserve beta-cell function, but the therapeutic effect needs to be improved. Intra-lymphatic administration may be a better alternative than subcutaneous administration, and combination therapy might improve efficacy. This review elucidates some actual problems of autoantigen therapy in the prevention and/or early intervention of type 1 diabetes.


Asunto(s)
Autoantígenos/administración & dosificación , Autoantígenos/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Administración Oral , Animales , Autoantígenos/inmunología , Quimioterapia Adyuvante , Quimioterapia Combinada , Glutamato Descarboxilasa/uso terapéutico , Humanos , Inyecciones Intralinfáticas , Inyecciones Subcutáneas , Insulina/metabolismo , Proinsulina/uso terapéutico , Vitamina D/uso terapéutico
11.
Diabetologia ; 63(3): 597-610, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31915895

RESUMEN

AIMS/HYPOTHESIS: The pathophysiology of type 1 diabetes has been linked to altered gut microbiota and more specifically to a shortage of intestinal production of the short-chain fatty acid (SCFA) butyrate, which may play key roles in maintaining intestinal epithelial integrity and in human and gut microbial metabolism. Butyrate supplementation can protect against autoimmune diabetes in mouse models. We thus set out to study the effect of oral butyrate vs placebo on glucose regulation and immune variables in human participants with longstanding type 1 diabetes. METHODS: We administered a daily oral dose of 4 g sodium butyrate or placebo for 1 month to 30 individuals with longstanding type 1 diabetes, without comorbidity or medication use, in a randomised (1:1), controlled, double-blind crossover trial, with a washout period of 1 month in between. Participants were randomly allocated to the 'oral sodium butyrate capsules first' or 'oral placebo capsules first' study arm in blocks of five. The clinical investigator received blinded medication from the clinical trial pharmacy. All participants, people doing measurements or examinations, or people assessing the outcomes were blinded to group assignment. The primary outcome was a change in the innate immune phenotype (monocyte subsets and in vitro cytokine production). Secondary outcomes were changes in blood markers of islet autoimmunity (cell counts, lymphocyte stimulation indices and CD8 quantum dot assays), glucose and lipid metabolism, beta cell function (by mixed-meal test), gut microbiota and faecal SCFA. The data was collected at the Amsterdam University Medical Centers. RESULTS: All 30 participants were analysed. Faecal butyrate and propionate levels were significantly affected by oral butyrate supplementation and butyrate treatment was safe. However, this modulation of intestinal SCFAs did not result in any significant changes in adaptive or innate immunity, or in any of the other outcome variables. In our discussion, we elaborate on this important discrepancy with previous animal work. CONCLUSIONS/INTERPRETATION: Oral butyrate supplementation does not significantly affect innate or adaptive immunity in humans with longstanding type 1 diabetes. TRIAL REGISTRATION: Netherlands Trial Register: NL4832 (www.trialregister.nl). DATA AVAILABILITY: Raw sequencing data are available in the European Nucleotide Archive repository (https://www.ebi.ac.uk/ena/browse) under study PRJEB30292. FUNDING: The study was funded by a Le Ducq consortium grant, a CVON grant, a personal ZONMW-VIDI grant and a Dutch Heart Foundation grant.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Ácido Butírico/administración & dosificación , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inmunidad Innata/efectos de los fármacos , Islotes Pancreáticos/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Administración Oral , Adulto , Ácido Butírico/efectos adversos , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Progresión de la Enfermedad , Femenino , Humanos , Islotes Pancreáticos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Países Bajos , Factores de Tiempo , Adulto Joven
12.
Diabetologia ; 63(4): 780-787, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31912198

RESUMEN

AIMS/HYPOTHESIS: Our aim was to study the association between serum 25-hydroxyvitamin D (25OHD) concentration and islet autoimmunity and type 1 diabetes in children with an increased genetic risk of type 1 diabetes. METHODS: Serum samples for 25OHD measurements were obtained in the Trial to Reduce IDDM in the Genetically at Risk (TRIGR) ancillary study (Divia) from children in 15 countries. Case children (n = 244) were defined as having positivity for at least two out of four diabetes-associated autoantibodies measured at any one sample. For each case child, two control children were selected matched for country and date of birth (±1 year) (n = 488). Of the case children, 144 developed type 1 diabetes. Serum 25OHD was measured repeatedly in infancy and childhood and was compared according to age at the first seroconversion (at 6, 12 and 18 months prior to and at seroconversion) and calendar age (0, 6, 12 and 18 months). RESULTS: In children with islet autoimmunity, mean serum 25OHD concentration was lower 18 months prior to the age of first seroconversion of the case children compared with the control children (57.7 vs 64.8 nmol/l, p = 0.007). In children with type 1 diabetes (n = 144), mean serum 25OHD concentration was lower 18 months prior to the age of the first seroconversion (58.0 vs 65.0 nmol/l, p = 0.018) and at the calendar age of 12 months (70.1 vs 75.9 nmol/l, p = 0.031) than in their control counterparts. Analyses were adjusted for month of sample collection, human leucocyte antigen genotype, maternal type 1 diabetes and sex. CONCLUSIONS/INTERPRETATION: The results suggest that early postnatal vitamin D may confer protection against the development of type 1 diabetes. TRIAL REGISTRATION: ClinicalTrials.gov NCT00179777.


Asunto(s)
Autoinmunidad , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/etiología , Islotes Pancreáticos/inmunología , Vitamina D/análogos & derivados , Edad de Inicio , Autoanticuerpos/sangre , Autoanticuerpos/genética , Autoinmunidad/genética , Estudios de Casos y Controles , Caseínas/administración & dosificación , Niño , Preescolar , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Femenino , Predisposición Genética a la Enfermedad , Prueba de Histocompatibilidad , Humanos , Lactante , Fórmulas Infantiles/química , Fórmulas Infantiles/normas , Fenómenos Fisiológicos Nutricionales del Lactante , Masculino , Estado Nutricional , Factores de Riesgo , Vitamina D/sangre
13.
Nat Biotechnol ; 37(12): 1446-1451, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31712773

RESUMEN

Vagus nerve stimulation can ameliorate autoimmune diseases such as rheumatoid arthritis by modulation of the immune system. Its efficacy for the treatment of type 1 diabetes has not been explored, in part because the nerves projecting to the pancreatic lymph nodes (pLNs) in mice are unmapped. Here, we map the nerve projecting to the pancreas and pLNs in mice and use a minimally invasive surgical procedure to implant micro-cuff electrodes onto the nerve. Pancreatic nerve electrical stimulation (PNES) resulted in ß-adrenergic receptor-mediated-accumulation of B and T cells in pLNs and reduced production of pro-inflammatory cytokines following lipopolysaccharide stimulation. Autoreactive T cells showed reduced proliferation in pLNs of mice receiving PNES as compared to sham controls. In a spontaneous mouse model of autoimmune diabetes, PNES inhibited disease progression in diabetic mice.


Asunto(s)
Diabetes Mellitus Tipo 1 , Terapia por Estimulación Eléctrica , Páncreas , Animales , Linfocitos B/inmunología , Glucemia/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Femenino , Insulina/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Páncreas/inmunología , Páncreas/inervación , Páncreas/metabolismo , Linfocitos T/inmunología
14.
Nutrients ; 11(9)2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31514368

RESUMEN

Type 1 diabetes (T1D) is a chronic autoimmune disease leading to immune-mediated destruction of pancreatic beta cells, resulting in the need for insulin therapy. The incidence of T1D is increasing worldwide, thus prompting researchers to investigate novel immunomodulatory strategies to halt autoimmunity and modify disease progression. T1D is considered as a multifactorial disease, in which genetic predisposition and environmental factors interact to promote the triggering of autoimmune responses against beta cells. Over the last decades, it has become clear that vitamin D exerts anti-inflammatory and immunomodulatory effects, apart from its well-established role in the regulation of calcium homeostasis and bone metabolism. Importantly, the global incidence of vitamin D deficiency is also dramatically increasing and epidemiologic evidence suggests an involvement of vitamin D deficiency in T1D pathogenesis. Polymorphisms in genes critical for vitamin D metabolism have also been shown to modulate the risk of T1D. Moreover, several studies have investigated the role of vitamin D (in different doses and formulations) as a potential adjuvant immunomodulatory therapy in patients with new-onset and established T1D. This review aims to present the current knowledge on the immunomodulatory effects of vitamin D and summarize the clinical interventional studies investigating its use for prevention or treatment of T1D.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Deficiencia de Vitamina D/tratamiento farmacológico , Vitamina D/uso terapéutico , Adolescente , Adulto , Animales , Niño , Diabetes Mellitus Tipo 1/epidemiología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Factores de Riesgo , Vitamina D/inmunología , Vitamina D/metabolismo , Deficiencia de Vitamina D/epidemiología , Deficiencia de Vitamina D/inmunología , Deficiencia de Vitamina D/metabolismo
15.
Mol Nutr Food Res ; 63(21): e1900307, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31423661

RESUMEN

SCOPE: This study aims to examine the protective effects of specific low-methoxyl pectin (LMP) on the development of type 1 diabetes (T1D). METHODS AND RESULTS: Female non-obese diabetic (NOD) mice are weaned onto either control or 5% LMP supplemented diets for up to 22 weeks of age. T1D incidence, gut barrier function, and pancreatic-gut immune responses are analyzed. LMP supplementation significantly dampened the onset of T1D in NOD mice. LMP supplementation induces caecal homeostasis, as indicated by the increasing SCFAs production, higher expression of tight junction proteins claudin 1, zonula occludens-2 in caecum. Furthermore, LMP-mediated caecal homeostasis impacts gut-pancreatic immunity, as evidenced by increased regulatory T cell population, modulated inflammatory cytokine expression, and suppressed NOD like receptor protein 3 (NLRP3) inflammasome activation in both caecum and pancreas. CONCLUSION: The data demonstrate that LMP limits T1D development by inducing caecal homeostasis to shape pancreatic immune environment, providing a scientific basis for using LMP as a novel functional supplementation to intervene T1D.


Asunto(s)
Ciego/efectos de los fármacos , Diabetes Mellitus Tipo 1/prevención & control , Hipoglucemiantes/farmacología , Pectinas/farmacología , Animales , Ciego/inmunología , Diabetes Mellitus Tipo 1/inmunología , Suplementos Dietéticos , Ácidos Grasos Volátiles/metabolismo , Femenino , Hipoglucemiantes/química , Factores Inmunológicos/farmacología , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones Endogámicos NOD , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Páncreas/efectos de los fármacos , Páncreas/inmunología , Páncreas/patología , Pectinas/química , Linfocitos T Reguladores/efectos de los fármacos
16.
Front Immunol ; 10: 1733, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417546

RESUMEN

Intestinal homeostasis underpins the development of type 1 diabetes (T1D), and dietary manipulations to enhance intestinal homeostasis have been proposed to prevent T1D. The current study aimed to investigate the efficacy of supplementing a novel specific low-methoxyl pectin (LMP) dietary fiber in preventing T1D development. Female NOD mice were weaned onto control or 5% (wt/wt) LMP supplemented diets for up to 40 weeks of age, overt diabetes incidence and blood glucose were monitored. Then broad-spectrum antibiotics (ABX) treatment per os for 7 days followed by gut microbiota transfer was performed to demonstrate gut microbiota-dependent effects. Next-generation sequencing was used for analyzing the composition of microbiota in caecum. Concentration of short chain fatty acids were determined by GC-MS. The barrier reinforcing tight junction proteins zonula occludens-2 (ZO-2), claudin-1 and NOD like receptor protein 3 (NLRP3) inflammasome activation were determined by Western blot. The proportion of CD25+Foxp3+CD4+ regulatory T cell (Foxp3+ Treg) in the pancreas, pancreatic and mesenteric lymph nodes was analyzed by flow cytometry. We found that LMP supplementation ameliorated T1D development in non-obese diabetic (NOD) mice, as evidenced by decreasing diabetes incidence and fasting glucose levels in LMP fed NOD mice. Further microbiota analysis revealed that LMP supplementation prevented T1D-associated caecal dysbiosis and selectively enriched caecal bacterial species to produce more SCFAs. The LMP-mediated microbial balance further enhanced caecal barrier function and shaped gut-pancreatic immune environment, as characterized by higher expression of tight junction proteins claudin-1, ZO-2 in caecum, increased Foxp3+ Treg population and decreased NLRP3 inflammasome activation in both caecum and pancreas. The microbiota-dependent beneficial effect of LMP on T1D was further proven by the fact that aberration of caecal microbiota by ABX treatment worsened T1D autoimmunity and could be restored with transfer of feces of LMP-fed NOD mice. These data demonstrate that this novel LMP limits T1D development by inducing caecal homeostasis to shape pancreatic immune environment. This finding opens a realistic option for gut microbiota manipulation and prevention of T1D in humans.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Pectinas/farmacología , Animales , Ciego/inmunología , Ciego/microbiología , Ciego/patología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Diabetes Mellitus Tipo 1/patología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Humanos , Ratones , Ratones Endogámicos NOD , Páncreas/inmunología , Páncreas/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
17.
Biofactors ; 45(6): 835-843, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31430413

RESUMEN

The immune system when acts against selfmolecules results in an imbalance in immunologic tolerance leading to the development of several autoimmune diseases (ADs) such as rheumatoid arthritis, asthma, ulcerative colitis, type 1 diabetes, and multiple sclerosis. Improved recognition of the mechanisms of ADs has led to the advancement of the management of these diseases. The principal mediators of ADs are inflammatory molecules. The herbal medicines due to their antioxidant and antiinflammatory properties have an important role in the management of ADs. Crocin is the principal chemical component extracted from saffron, which is a medicinal plant. This review focuses on the therapeutic effects of Crocin in various ADs.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Carotenoides/uso terapéutico , Crocus/química , Antioxidantes/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/inmunología , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico
18.
Immunohorizons ; 3(6): 236-253, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-31356169

RESUMEN

Dendritic cells (DCs) are crucial for the production of adaptive immune responses to disease-causing microbes. However, in the steady state (i.e., in the absence of an infection or when Ags are experimentally delivered without a DC-activating adjuvant), DCs present Ags to T cells in a tolerogenic manner and are important for the establishment of peripheral tolerance. Delivery of islet Ags to DCs using Ag-linked Abs to the DC endocytic receptor CD205 has shown promise in the NOD mouse model of type 1 diabetes (T1D). It is important to note, however, that all myeloid DCs express CD205 in humans, whereas in mice, only one of the classical DC subsets does (classical DC1; CD8α+ in spleen). Thus, the evaluation of CD205-targeted treatments in mice will likely not accurately predict the results observed in humans. To overcome this challenge, we have developed and characterized a novel NOD mouse model in which all myeloid DCs transgenically express human CD205 (hCD205). This NOD.hCD205 strain displays a similar T1D incidence profile to standard NOD mice. The presence of the transgene does not alter DC development, phenotype, or function. Importantly, the DCs are able to process and present Ags delivered via hCD205. Because Ags taken up via hCD205 can be presented on both class I and class II MHC, both CD4+ and CD8+ T cells can be modulated. As both T cell subsets are important for T1D pathogenesis, NOD.hCD205 mice represent a unique, patient-relevant tool for the development and optimization of DC-directed T1D therapies.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Inmunoterapia/métodos , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Presentación de Antígeno , Antígenos CD/genética , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Tolerancia Inmunológica , Lectinas Tipo C/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor/genética , Receptores de Superficie Celular/genética
19.
J Vis Exp ; (145)2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30958463

RESUMEN

Plant molecular farming is the use of plants to produce molecules of interest. In this perspective, plants may be used both as bioreactors for the production and subsequent purification of the final product and for the direct oral delivery of heterologous proteins when using edible plant species. In this work, we present the development of a candidate oral vaccine against Type 1 Diabetes (T1D) in edible plant systems using deconstructed plant virus-based recombinant DNA technology, delivered with vacuum infiltration. Our results show that a red beet is a suitable host for the transient expression of a human derived autoantigen associated to T1D, considered to be a promising candidate as a T1D vaccine. Leaves producing the autoantigen were thoroughly characterized for their resistance to gastric digestion, for the presence of residual bacterial charge and for their secondary metabolic profile, giving an overview of the process production for the potential use of plants for direct oral delivery of a heterologous protein. Our analysis showed almost complete degradation of the freeze-dried candidate oral vaccine following a simulated gastric digestion, suggesting that an encapsulation strategy in the manufacture of the plant-derived GAD vaccine is required.


Asunto(s)
Beta vulgaris/genética , Productos Biológicos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Vacunas/inmunología , Administración Oral , Animales , Glutamato Descarboxilasa/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Metaboloma , Mutación/genética , Hojas de la Planta/metabolismo , Plantas Modificadas Genéticamente , Spinacia oleracea/metabolismo , Porcinos , Vacunas/administración & dosificación
20.
FASEB J ; 33(7): 8241-8248, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30916998

RESUMEN

Type 1 diabetes (T1D) is an autoimmune disease characterized by the immune-mediated destruction of insulin-producing ß cells. Recent studies showed that in addition to malaria, artemisinin and its derivative, artesunate (AS), could alleviate several autoimmune diseases. However, whether AS has a role in the prevention or treatment of T1D is still unknown. Therefore, in this study we administrated AS or DMSO in the drinking water of nonobese diabetic (NOD) mice, a mouse model of T1D. We found that AS administration significantly prevented the incidence of T1D. The frequency of IL-4-producing CD4+ single-positive T cells and CD8+ T cells was significantly elevated, and IFN-γ-producing T cells were reduced in the spleen and pancreatic lymph nodes. In the pancreas, the skewing to IL-4-producing T cells was also observed. In addition, more regulatory T cells were found in the pancreas. mRNA levels of proinflammatory cytokines, including TNF-α and IL-6, were decreased. In addition, AS administration promoted the functional maturity of ß cells in vitro. Our findings demonstrate that AS administration can prevent T1D in NOD mice mainly by reducing autoimmune T cells and increasing protective T cells. Our data constitute the first functional study of AS in T1D, which may provide a new rationale for future translational studies.-Li, Z., Shi, X., Liu, J., Shao, F., Huang, G., Zhou, Z., Zheng, P. Artesunate prevents type 1 diabetes in NOD mice mainly by inducing protective IL-4-producing T cells and regulatory T cells.


Asunto(s)
Artesunato/farmacología , Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 1/prevención & control , Células Secretoras de Insulina/inmunología , Interleucina-4/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Células Secretoras de Insulina/patología , Interferón gamma/inmunología , Interleucina-6/inmunología , Ratones , Ratones Endogámicos NOD , Linfocitos T Reguladores/patología , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA