Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Clin Immunol ; 244: 109118, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36084852

RESUMEN

To enable accurate, high-throughput and longer-term studies of the immunopathogenesis of type 1 diabetes (T1D), we established three in-vitro islet-immune injury models by culturing spheroids derived from primary human islets with proinflammatory cytokines, activated peripheral blood mononuclear cells or HLA-A2-restricted preproinsulin-specific cytotoxic T lymphocytes. In all models, ß-cell function declined as manifested by increased basal and decreased glucose-stimulated insulin release (GSIS), and decreased intracellular insulin content. Additional hallmarks of T1D progression such as loss of the first-phase insulin response (FFIR), increased proinsulin-to-insulin ratios, HLA-class I expression, and inflammatory cytokine release were also observed. Using these models, we show that liraglutide, a glucagon-like peptide 1 receptor agonist, prevented loss of GSIS under T1D-relevant stress, by preserving the FFIR and decreasing immune cell infiltration and cytokine secretion. Our results corroborate that liraglutide mediates an anti-inflammatory effect that aids in protecting ß-cells from the immune-mediated attack that leads to T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Islotes Pancreáticos , Antiinflamatorios/farmacología , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucosa/metabolismo , Antígeno HLA-A2 , Humanos , Insulina , Células Secretoras de Insulina/metabolismo , Leucocitos Mononucleares/metabolismo , Liraglutida/metabolismo , Liraglutida/farmacología , Proinsulina/metabolismo
2.
Front Immunol ; 12: 683091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220832

RESUMEN

In human type 1 diabetes and animal models of the disease, a diverse assortment of immune cells infiltrates the pancreatic islets. CD8+ T cells are well represented within infiltrates and HLA multimer staining of pancreas sections provides clear evidence that islet epitope reactive T cells are present within autoimmune lesions. These bona fide effectors have been a key research focus because these cells represent an intellectually attractive culprit for ß cell destruction. However, T cell receptors are highly diverse in human insulitis. This suggests correspondingly broad antigen specificity, which includes a majority of T cells for which there is no evidence of islet-specific reactivity. The presence of "non-cognate" T cells in insulitis raises suspicion that their role could be beyond that of an innocent bystander. In this perspective, we consider the potential pathogenic contribution of non-islet-reactive T cells. Our intellectual framework will be that of a criminal investigation. Having arraigned islet-specific CD8+ T cells for the murder of pancreatic ß cells, we then turn our attention to the non-target immune cells present in human insulitis and consider the possible regulatory, benign, or effector roles that they may play in disease. Considering available evidence, we overview the case that can be made that non-islet-reactive infiltrating T cells should be suspected as co-conspirators or accessories to the crime and suggest some possible routes forward for reaching a better understanding of their role in disease.


Asunto(s)
Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/etiología , Susceptibilidad a Enfermedades , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Autoinmunidad/genética , Biomarcadores , Comunicación Celular/genética , Comunicación Celular/inmunología , Microambiente Celular/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Humanos , Islotes Pancreáticos/patología , Subgrupos de Linfocitos T/patología
3.
Clin Transl Immunology ; 10(7): e1309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34267894

RESUMEN

OBJECTIVES: The detection of a peripheral immune cell signature that specifically reflects autoimmunity in type 1 diabetes would enable the prediction and staging of disease on an individual basis. However, defining such a signature is technically challenging. Reliable interpretation of immune cell-related biomarkers depends on their inherent variability and, to understand this variability, longitudinal analyses are required. METHODS: We performed a longitudinal observational study in which 40 individuals with elevated genetic risk of type 1 diabetes and persistent islet autoantibodies provided a blood sample every 4-6 weeks for > 1 year. RESULTS: Peripheral immune cell composition (T cells, NK cells and monocytes) was assessed using well-validated flow cytometry panels and demonstrated that, while non-antigen-specific immune cell subsets were stable over time, autoantigen-reactive T-cell frequencies were highly variable in and between individuals. Neither the frequency nor phenotype of non-antigen-specific subsets or autoreactive CD8+ T cells associated with clinical onset of T1D. CONCLUSION: The findings from the Type 1 Diabetes Longitudinal BIomarker Trial underscore the inherent challenge of evaluating changes in peripheral immune cell populations as surrogates of organ-specific disease activity. The variability of peripheral antigen-specific T cells precludes their use as a prognostic marker and clearly demonstrates that a reliable prognostic cell signature remains elusive.

4.
J Autoimmun ; 119: 102628, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33706238

RESUMEN

Human leukocyte antigens of class-I (HLA-I) molecules are hyper-expressed in insulin-containing islets (ICI) of type 1 diabetic (T1D) donors. This study investigated the HLA-I expression in autoantibody positive (AAB+) donors and defined its intra-islet and intracellular localization as well as proximity to infiltrating CD8 T cells with high-resolution confocal microscopy. We found HLA-I hyper-expression had already occurred prior to clinical diagnosis of T1D in islets of AAB+ donors. Interestingly, throughout all stages of disease, HLA-I was mostly expressed by alpha cells. Hyper-expression in AAB+ and T1D donors was associated with intra-cellular accumulation in the Golgi. Proximity analysis showed a moderate but significant correlation between HLA-I and infiltrating CD8 T cells only in ICI of T1D donors, but not in AAB+ donors. These observations not only demonstrate a very early, islet-intrinsic immune-independent increase of HLA-I during diabetes pathogenesis, but also point towards a role for alpha cells in T1D.


Asunto(s)
Expresión Génica , Células Secretoras de Glucagón/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Estado Prediabético/etiología , Estado Prediabético/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Autoinmunidad , Biomarcadores , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Susceptibilidad a Enfermedades/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Transporte de Proteínas , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
5.
Sci Adv ; 6(42)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33067232

RESUMEN

Preproinsulin (PPI) is presumably a crucial islet autoantigen found in patients with type 1 diabetes (T1D) but is also recognized by CD8+ T cells from healthy individuals. We quantified PPI-specific CD8+ T cells within different areas of the human pancreas from nondiabetic controls, autoantibody-positive donors, and donors with T1D to investigate their role in diabetes development. This spatial cellular quantitation revealed unusually high frequencies of autoreactive CD8+ T cells supporting the hypothesis that PPI is indeed a key autoantigen. To our surprise, PPI-specific CD8+ T cells were already abundantly present in the nondiabetic pancreas, thus questioning the dogma that T1D is caused by defective thymic deletion or systemic immune dysregulation. During T1D development, these cells accumulated in and around islets, indicating that an islet-specific trigger such as up-regulation of major histocompatibility complex class I might be essential to unmask beta cells to the immune system.


Asunto(s)
Diabetes Mellitus Tipo 1 , Páncreas Exocrino , Autoantígenos , Linfocitos T CD8-positivos , Humanos , Insulina , Precursores de Proteínas
7.
Sci Adv ; 6(35)2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33052874

RESUMEN

The notably lobular distribution of immune lesions in type 1 diabetes (T1D) has been hypothesized to be the result of innervation within the pancreas. To investigate whether neuroimmune interactions could explain this phenomenon, we explored the impact of sympathetic signaling in the RIP-LCMV-GP mouse model of autoimmune diabetes. In this model, the CD8+ T cell attack on ß cells replicates a key pathogenic feature of human T1D. We found that inhibition of α1 adrenoceptors, ablation of sympathetic nerves, and surgical denervation all had a protective effect in this model, without affecting the systemic presence of ß cell-reactive CD8+ T cells. In vivo multiphoton imaging revealed a local effect within pancreatic islets including limited infiltration of both macrophages and ß cell-specific CD8+ T cells. Islet-resident macrophages expressed adrenoceptors and were responsive to catecholamines. Islet macrophages may therefore constitute a pivotal neuroimmune signaling relay and could be a target for future interventions in T1D.


Asunto(s)
Diabetes Mellitus Tipo 1 , Animales , Linfocitos T CD8-positivos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Páncreas/patología , Receptores Adrenérgicos
8.
Front Endocrinol (Lausanne) ; 11: 606434, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33469446

RESUMEN

Since the establishment of the network for pancreatic organ donors with diabetes (nPOD), we have gained unprecedented insight into the pathology of human type 1 diabetes. Many of the pre-existing "dogmas", mostly derived from studies of animal models and sometimes limited human samples, have to be revised now. For example, we have learned that autoreactive CD8 T cells are present even in healthy individuals within the exocrine pancreas. Furthermore, their "attraction" to islets probably relies on beta-cell intrinsic events, such as the over-expression of MHC class I and resulting presentation of autoantigens such as (prepro)insulin. In addition, we are discovering other signs of beta-cell dysfunction, possibly at least in part due to stress, such as the over-expression of certain cytokines. This review summarizes the latest developments focusing on cytokines and autoreactive CD8 T cells in human type 1 diabetes pathogenesis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Animales , Diabetes Mellitus Tipo 1/patología , Humanos , Páncreas/inmunología , Páncreas/patología
9.
J Autoimmun ; 107: 102378, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31818546

RESUMEN

Human herpesvirus-6 (HHV-6) is a ubiquitous pathogen associated with nervous and endocrine autoimmune disorders. The aim of this study was to investigate the presence of HHV-6 in pancreatic tissue sections from non-diabetic, auto-antibody positive (AAB+), and donors with type 1 diabetes (T1D) and explore whether there is any association between HHV-6 and MHC class I hyperexpression and CD8 T cell infiltration. HHV-6 DNA was detected by PCR and its protein was examined by indirect immunofluorescence assay followed by imaging using high-resolution confocal microscopy. Viral DNA (U67) was found in most pancreata of non-diabetic (3 out of 4), AAB+ (3 out of 5) and T1D donors (6 out of 7). Interestingly, HHV-6 glycoprotein B (gB) was more expressed in islets and exocrine pancreas of donors with T1D. However, gB expression was not directly associated with other pathologies. Out of 20 islets with high gB expression, only 3 islets (15%) showed MHC class I hyperexpression. Furthermore, no correlation was found between gB expression and CD8 T cell infiltration on a per-islet basis in any of the groups. Our observations indicate that HHV-6 DNA and protein are present in the pancreas of non-diabetic subjects but gB expression is higher in the pancreas of donors with T1D. The possible role of HHV-6 as a contributory factor for T1D should therefore be further investigated.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Susceptibilidad a Enfermedades , Herpesvirus Humano 6 , Páncreas/virología , Infecciones por Roseolovirus/complicaciones , Autoinmunidad , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/metabolismo , Expresión Génica , Herpesvirus Humano 6/genética , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/virología , Páncreas/inmunología , Páncreas/metabolismo , Infecciones por Roseolovirus/virología
10.
Diabetes ; 67(9): 1858-1866, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945890

RESUMEN

Indoleamine 2,3 dioxygenase-1 (IDO1) is a powerful immunoregulatory enzyme that is deficient in patients with type 1 diabetes (T1D). In this study, we present the first systematic evaluation of IDO1 expression and localization in human pancreatic tissue. Although IDO1 was constitutively expressed in ß-cells from donors without diabetes, less IDO1 was expressed in insulin-containing islets from double autoantibody-positive donors and patients with recent-onset T1D, although it was virtually absent in insulin-deficient islets from donors with T1D. Scatter plot analysis suggested that IDO1 decay occurred in individuals with multiple autoantibodies, prior to ß-cell demise. IDO1 impairment might therefore contribute to ß-cell demise and could potentially emerge as a promising therapeutic target.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Diabetes Mellitus Tipo 1/metabolismo , Regulación hacia Abajo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Células Secretoras de Insulina/enzimología , Estado Prediabético/metabolismo , Adolescente , Adulto , Anciano , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Cadáver , Estudios de Cohortes , Estudios Transversales , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Progresión de la Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Persona de Mediana Edad , Estado Prediabético/inmunología , Estado Prediabético/patología , Estado Prediabético/fisiopatología , Transporte de Proteínas , Adulto Joven
11.
Front Immunol ; 9: 1069, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29892281

RESUMEN

Obesity is associated with adipose tissue inflammation, insulin resistance, and the development of type 2 diabetes (T2D). However, our knowledge is mostly based on conventional murine models and promising preclinical studies rarely translated into successful therapies. There is a growing awareness of the limitations of studies in laboratory mice, housed in abnormally hygienic specific pathogen-free (SPF) conditions, as relevant aspects of the human immune system remain unappreciated. Here, we assessed the impact of housing conditions on adaptive immunity and metabolic disease processes during high-fat diet (HFD). We therefore compared diet-induced obesity in SPF mice with those housed in non-SPF, so-called "antigen exposed" (AE) conditions. Surprisingly, AE mice fed a HFD maintained increased insulin levels to compensate for insulin resistance, which was reflected in islet hyperplasia and improved glucose tolerance compared to SPF mice. By contrast, we observed higher proportions of effector/memory T cell subsets in blood and liver of HFD AE mice accompanied by the development of non-alcoholic steatohepatitis-like liver pathology. Thus, our data demonstrate the impact of housing conditions on metabolic alterations. Studies in AE mice, in which physiological microbial exposure was restored, could provide a tool for revealing therapeutic targets for immune-based interventions for T2D patients.


Asunto(s)
Inmunidad Adaptativa , Diabetes Mellitus Tipo 2/etiología , Vivienda , Obesidad/complicaciones , Animales , Biomarcadores , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Vivienda para Animales , Memoria Inmunológica , Inmunofenotipificación , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/etiología , Obesidad/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
12.
Diabetes Metab Res Rev ; 34(6): e3010, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29637693

RESUMEN

BACKGROUND: Oral insulin as a preventive strategy and/or treatment of type 1 diabetes has been the target of much research. Producing oral insulins is a complex and challenging task, with numerous pitfalls, due to physiological, physical, and biochemical barriers. Our aim was to determine the impact of oral insulin on the delicate gut microbiota composition. METHODS: Female nonobese diabetic mice were given oral porcine insulin 2 times a week from 5 weeks of age for 4 weeks, and then subsequently once a week for 21 weeks, or until euthanized. The mice were divided into groups on a gluten-reduced diet or a standard diet. Gut microbiota composition was analysed based on faecal samples, and the type 1 diabetes incidence of the mice was monitored. RESULTS: We observed no influence of the oral porcine insulin on the gut microbiota composition of mice on a gluten-reduced or a standard diet at 9 weeks of age. Also, the administration of oral insulin did not influence the incidence of type 1 diabetes at 30 weeks of age. CONCLUSIONS: Oral porcine insulin does not alter the gut microbiota composition of nonobese diabetic mice on either a gluten-reduced diet or standard diet. Also, the oral porcine insulin did not influence the incidence of type 1 diabetes in the groups.


Asunto(s)
Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Tipo 1/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Insulina Regular Porcina/administración & dosificación , Administración Oral , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/patología , Disbiosis/inmunología , Disbiosis/patología , Heces/microbiología , Femenino , Insulina Regular Porcina/efectos adversos , Ratones , Ratones Endogámicos NOD , Porcinos
14.
Cell Rep ; 22(10): 2667-2676, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29514095

RESUMEN

Many patients with type 1 diabetes (T1D) have residual ß cells producing small amounts of C-peptide long after disease onset but develop an inadequate glucagon response to hypoglycemia following T1D diagnosis. The features of these residual ß cells and α cells in the islet endocrine compartment are largely unknown, due to the difficulty of comprehensive investigation. By studying the T1D pancreas and isolated islets, we show that remnant ß cells appeared to maintain several aspects of regulated insulin secretion. However, the function of T1D α cells was markedly reduced, and these cells had alterations in transcription factors constituting α and ß cell identity. In the native pancreas and after placing the T1D islets into a non-autoimmune, normoglycemic in vivo environment, there was no evidence of α-to-ß cell conversion. These results suggest an explanation for the disordered T1D counterregulatory glucagon response to hypoglycemia.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Regulación de la Expresión Génica , Células Secretoras de Glucagón/metabolismo , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Reprogramación Celular , Niño , Femenino , Glucagón/metabolismo , Células Secretoras de Glucagón/patología , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Masculino , Ratones , Persona de Mediana Edad , Fenotipo , Donantes de Tejidos , Factores de Transcripción/metabolismo , Adulto Joven
15.
Sci Immunol ; 3(21)2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29572238

RESUMEN

The inflammatory lesion at the pancreatic islet in type 1 diabetes (T1D) contains a heterogeneous infiltrate of T cells. In human and mouse studies, a large majority (98 to 99%) of the cytotoxic CD8+ T cells (CTLs) within islets are not specific to any islet antigen and are thought to passively add to tissue damage. We show by intravital confocal microscopy the opposite, immune-regulatory function of this cohort of CTLs. Diabetes did not develop in mice with islets showing high levels of infiltration of non-islet-specific CTLs not recognizing local antigens. Accumulation of such CTLs resulted in lower activation and proliferation of islet-specific CTLs, leading them to enter a state of unresponsiveness due to limited access to antigens at the inflammatory lesion. This nonspecific suppression by nonautoreactive CTLs was recapitulated in a model of viral meningitis, may explain viral interference in autoimmunity, and provides insight into the regulation of organ-specific autoimmune responses.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos/inmunología , Autoinmunidad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos
17.
J Autoimmun ; 84: 65-74, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28711285

RESUMEN

Immunotherapy for type 1 diabetes (T1D) has previously focused on suppressing the autoimmune response against pancreatic beta cells to preserve endogenous insulin production and regulate glucose levels. With increased attention toward combination therapy strategies, studies indicate the multifunctional cytokine interleukin-21 (IL-21) may be a suitable target as an immuno-modulatory arm, while glucagon-like peptide-1 receptor (GLP-1R) agonists may be appropriate as a beta cell protective arm in combination therapy for T1D. We report here that treatment with anti-IL-21 monoclonal antibody delays diabetes onset in the spontaneous non-obese diabetic (NOD) and NOD.scid adoptive transfer models, while its effect in reversing recent-onset hyperglycemia is limited. However, the combination of anti-IL-21 plus the GLP-1R agonist liraglutide is effective in reversing established disease compared to either monotherapy in both the NOD and rat insulin promotor-lymphocytic choriomeningitis virus glycoprotein (RIP-LCMV-GP) models of autoimmune diabetes. Enhanced efficacy is particularly evident in severely hyperglycemic mice, with return to normoglycemia remaining stable for the majority of mice even after therapy is withdrawn. Importantly, increased beta cell proliferation does not appear to be the predominant mechanism. In conclusion, combination therapy with anti-IL-21 and liraglutide is able to consistently reverse disease in mouse models of T1D. The observed effects rival the most effective experimental disease-modifying treatments tested in preclinical studies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Diabetes Mellitus Tipo 1/terapia , Hiperglucemia/terapia , Inmunoterapia/métodos , Células Secretoras de Insulina/inmunología , Interleucinas/inmunología , Liraglutida/uso terapéutico , Animales , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Hiperglucemia/inmunología , Insulina/genética , Insulina/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos
18.
J Autoimmun ; 81: 68-73, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28325643

RESUMEN

Interleukin-1ß (IL-1ß) is known to trigger beta cell dysfunction in vitro and could potentially play a role during the pathogenesis of type 1 diabetes and type 2 diabetes. However, several clinical trials attempting to block IL-1ß function have had minimal success. We therefore re-investigated local expression of IL-1ß in human diabetic and non-diabetic pancreata. We obtained pancreatic tissue sections from the Network for Pancreatic Organ Donors with Diabetes (nPOD) including non-diabetic (n = 9), non-diabetic auto-antibody positive (AAb+, n = 5), type 1 diabetes (n = 6), and type 2 diabetes (n = 6) donors. Islets were systematically investigated for the presence of IL-1ß mRNA by in situ hybridization and IL-1ß protein by indirect immunofluorescence. We found that intra-islet IL-1ß was produced at comparable level in both non-diabetic and diabetic donors. Interestingly, the main source for IL-1ß was alpha cells but not beta cells. Our findings call into question the role of IL-1ß in the diabetic pancreas as it has been proposed in previous literature. Additionally, our results regarding the localization of IL-1ß should lead to further investigation into the role of IL-1ß in the physiology of pancreatic alpha cells.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Interleucina-1beta/metabolismo , Páncreas/citología , Páncreas/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Expresión Génica , Humanos , Interleucina-1beta/genética , Páncreas/patología
19.
Diabetes ; 66(5): 1334-1345, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28137793

RESUMEN

Type 1 diabetes is characterized by the loss of insulin production caused by ß-cell dysfunction and/or destruction. The hypothesis that ß-cell loss occurs early during the prediabetic phase has recently been challenged. Here we show, for the first time in situ, that in pancreas sections from autoantibody-positive (Ab+) donors, insulin area and ß-cell mass are maintained before disease onset and that production of proinsulin increases. This suggests that ß-cell destruction occurs more precipitously than previously assumed. Indeed, the pancreatic proinsulin-to-insulin area ratio was also increased in these donors with prediabetes. Using high-resolution confocal microscopy, we found a high accumulation of vesicles containing proinsulin in ß-cells from Ab+ donors, suggesting a defect in proinsulin conversion or an accumulation of immature vesicles caused by an increase in insulin demand and/or a dysfunction in vesicular trafficking. In addition, islets from Ab+ donors were larger and contained a higher number of ß-cells per islet. Our data indicate that ß-cell mass (and function) is maintained until shortly before diagnosis and declines rapidly at the time of clinical onset of disease. This suggests that secondary prevention before onset, when ß-cell mass is still intact, could be a successful therapeutic strategy.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo , Estado Prediabético/metabolismo , Proinsulina/metabolismo , Adulto , Autoanticuerpos , Estudios de Casos y Controles , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Células Secretoras de Insulina/patología , Masculino , Microscopía Confocal , Persona de Mediana Edad , Páncreas/patología , Estado Prediabético/patología , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/patología , Adulto Joven
20.
Diabetes Obes Metab ; 19(5): 705-712, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28094469

RESUMEN

AIMS: Glucagon-like peptide-1 (GLP-1) is an incretin hormone which stimulates insulin release and inhibits glucagon secretion from the pancreas in a glucose-dependent manner. Incretin-based therapies, consisting of GLP-1 receptor (GLP-1R) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors, are used for the treatment of type 2 diabetes (T2D). Immunohistochemical studies for GLP-1R expression have been hampered previously by the use of unspecific polyclonal antibodies. This study aimed to assess the expression levels of GLP-1R in a set of T2D donor samples obtained via nPOD. METHODS: This study used a new monoclonal antibody to assess GLP-1R expression in pancreatic tissue from 23 patients with T2D, including 7 with a DPP-4 inhibitor and 1 with a history of GLP-1R agonist treatment. A software-based automated image analysis algorithm was used for quantitating intensities and area fractions of GLP-1R positive compartments. RESULTS: The highest intensity GLP-1R immunostaining was seen in beta-cells in islets (average signal intensity, 76.1 [±8.1]). GLP-1R/insulin double-labelled single cells or small clusters of cells were also frequently located within or in close vicinity of ductal epithelium in all samples and with the same GLP-1R immunostaining intensity as found in beta-cells in islets. In the exocrine pancreas a large proportion of acinar cells expressed GLP-1R with a 3-fold lower intensity of immunoreactivity as compared to beta-cells (average signal intensity 25.5 [±3,3]). Our studies did not unequivocally demonstrate GLP-1R immunoreactivity on normal-appearing ductal epithelium. Pancreatic intraepithelial neoplasia (PanINs; a form of non-invasive pancreatic ductular neoplasia) was seen in most samples, and a minority of these expressed low levels of GLP-1R. CONCLUSION: These data confirm the ubiquity of early stage PanIN lesions in patients with T2D and do not support the hypothesis that incretin-based therapies are associated with progression towards the more advanced stage PanIN lesions.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales , Especificidad de Anticuerpos , Biomarcadores/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Femenino , Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Humanos , Hipoglucemiantes/uso terapéutico , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Incretinas/uso terapéutico , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Masculino , Persona de Mediana Edad , Páncreas/efectos de los fármacos , Páncreas/patología , Bancos de Tejidos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...