Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetologia ; 66(5): 884-896, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36884057

RESUMEN

AIMS/HYPOTHESIS: Transcriptome analyses revealed insulin-gene-derived transcripts in non-beta endocrine islet cells. We studied alternative splicing of human INS mRNA in pancreatic islets. METHODS: Alternative splicing of insulin pre-mRNA was determined by PCR analysis performed on human islet RNA and single-cell RNA-seq analysis. Antisera were generated to detect insulin variants in human pancreatic tissue using immunohistochemistry, electron microscopy and single-cell western blot to confirm the expression of insulin variants. Cytotoxic T lymphocyte (CTL) activation was determined by MIP-1ß release. RESULTS: We identified an alternatively spliced INS product. This variant encodes the complete insulin signal peptide and B chain and an alternative C-terminus that largely overlaps with a previously identified defective ribosomal product of INS. Immunohistochemical analysis revealed that the translation product of this INS-derived splice transcript was detectable in somatostatin-producing delta cells but not in beta cells; this was confirmed by light and electron microscopy. Expression of this alternatively spliced INS product activated preproinsulin-specific CTLs in vitro. The exclusive presence of this alternatively spliced INS product in delta cells may be explained by its clearance from beta cells by insulin-degrading enzyme capturing its insulin B chain fragment and a lack of insulin-degrading enzyme expression in delta cells. CONCLUSIONS/INTERPRETATION: Our data demonstrate that delta cells can express an INS product derived from alternative splicing, containing both the diabetogenic insulin signal peptide and B chain, in their secretory granules. We propose that this alternative INS product may play a role in islet autoimmunity and pathology, as well as endocrine or paracrine function or islet development and endocrine destiny, and transdifferentiation between endocrine cells. INS promoter activity is not confined to beta cells and should be used with care when assigning beta cell identity and selectivity. DATA AVAILABILITY: The full EM dataset is available via www.nanotomy.org (for review: http://www.nanotomy.org/OA/Tienhoven2021SUB/6126-368/ ). Single-cell RNA-seq data was made available by Segerstolpe et al [13] and can be found at https://sandberglab.se/pancreas . The RNA and protein sequence of INS-splice was uploaded to GenBank (BankIt2546444 INS-splice OM489474).


Asunto(s)
Insulisina , Islotes Pancreáticos , Humanos , Células Secretoras de Somatostatina/metabolismo , Insulisina/metabolismo , Insulina/genética , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , ARN , Señales de Clasificación de Proteína
2.
Expert Opin Ther Targets ; 25(4): 269-281, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33896351

RESUMEN

INTRODUCTION: Aberrant citrullination and excessive peptidylarginine deiminase (PAD) activity are detected in numerous challenging autoimmune diseases such as rheumatoid arthritis, inflammatory bowel diseases, systemic lupus erythematosus, multiple sclerosis, and type 1 diabetes. Because excessive PAD activity is a common denominator in these diseases, PADs are interesting potential therapeutic targets for future therapies. AREAS COVERED: This review summarizes the advances made in the design of PAD inhibitors, their utilization and therapeutic potential in preclinical mouse models of autoimmunity. Relevant literature encompasses studies from 1994 to 2021 that are available on PubMed.gov. EXPERT OPINION: Pan-PAD inhibition is a promising therapeutic strategy for autoimmune diseases. Drugs achieving pan-PAD inhibition were capable of ameliorating, reversing, and preventing clinical symptoms in preclinical mouse models. However, the implications for PADs in key biological processes potentially present a high risk for clinical complications and could hamper the translation of PAD inhibitors to the clinic. We envisage that PAD isozyme-specific inhibitors will improve the understanding the role of PAD isozymes in disease pathology, reduce the risk of side-effects and enhance prospects for future clinical translation.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Terapia Molecular Dirigida , Desiminasas de la Arginina Proteica/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/fisiopatología , Autoinmunidad , Citrulinación/efectos de los fármacos , Modelos Animales de Enfermedad , Diseño de Fármacos , Humanos , Isoenzimas , Ratones , Desiminasas de la Arginina Proteica/metabolismo
3.
Diabetes ; 69(4): 670-680, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31896552

RESUMEN

The signal peptide of preproinsulin is a major source for HLA class I autoantigen epitopes implicated in CD8 T cell (CTL)-mediated ß-cell destruction in type 1 diabetes (T1D). Among them, the 10-mer epitope located at the C-terminal end of the signal peptide was found to be the most prevalent in patients with recent-onset T1D. While the combined action of signal peptide peptidase and endoplasmic reticulum (ER) aminopeptidase 1 (ERAP1) is required for processing of the signal peptide, the mechanisms controlling signal peptide trimming and the contribution of the T1D inflammatory milieu on these mechanisms are unknown. Here, we show in human ß-cells that ER stress regulates ERAP1 gene expression at posttranscriptional level via the IRE1α/miR-17-5p axis and demonstrate that inhibition of the IRE1α activity impairs processing of preproinsulin signal peptide antigen and its recognition by specific autoreactive CTLs during inflammation. These results underscore the impact of ER stress in the increased visibility of ß-cells to the immune system and position the IRE1α/miR-17 pathway as a central component in ß-cell destruction processes and as a potential target for the treatment of autoimmune T1D.


Asunto(s)
Aminopeptidasas/metabolismo , Linfocitos T CD8-positivos/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Endorribonucleasas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Aminopeptidasas/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/inmunología , Interferón gamma/farmacología , Interleucina-1beta/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Proteínas Serina-Treonina Quinasas/genética , Regulación hacia Arriba
4.
Sci Rep ; 8(1): 17738, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30532033

RESUMEN

During type 1 diabetes development, cells in the islets of Langerhans engage adaptive mechanisms in response to inflammatory signals to cope with stress, to restore cellular homeostasis, and to preserve cell function. Disruption of these mechanisms may induce the formation of a repertoire of stress-induced neoantigens, which are critical in the loss of tolerance to beta cells and the development of autoimmunity. While multiple lines of evidence argue for a critical role of the endoplasmic reticulum in these processes, the lack of tools to specifically monitor beta cell stress hampers the development of therapeutic interventions focusing on maintaining endoplasmic reticulum homeostasis. Here we designed and evaluated a stress-induced reporter in which induction of stress correlates with increased light emission. This Gaussia luciferase-based reporter system employs the unconventional cytoplasmic splicing of XBP1 to report ER stress in cells exposed to known ER-stress inducers. Linking this reporter to a human beta cell-specific promotor allows tracing ER-stress in isolated human beta cells as well as in the EndoC-ßH1 cell line. This reporter system represents a valuable tool to assess ER stress in human beta cells and may aid the identification of novel therapeutics that can prevent beta cell stress in human pancreatic islets.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/fisiología , Células Secretoras de Insulina/fisiología , Islotes Pancreáticos/fisiología , Células Cultivadas , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Células HEK293 , Humanos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos
5.
Nat Med ; 23(4): 501-507, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28263308

RESUMEN

Identification of epitopes that are recognized by diabetogenic T cells and cause selective beta cell destruction in type 1 diabetes (T1D) has focused on peptides originating from native beta cell proteins. Translational errors represent a major potential source of antigenic peptides to which central immune tolerance is lacking. Here, we describe an alternative open reading frame within human insulin mRNA encoding a highly immunogenic polypeptide that is targeted by T cells in T1D patients. We show that cytotoxic T cells directed against the N-terminal peptide of this nonconventional product are present in the circulation of individuals diagnosed with T1D, and we provide direct evidence that such CD8+ T cells are capable of killing human beta cells and thereby may be diabetogenic. This study reveals a new source of nonconventional polypeptides that act as self-epitopes in clinical autoimmune disease.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Diabetes Mellitus Tipo 1/inmunología , Insulina/genética , Péptidos/inmunología , ARN Mensajero/genética , Linfocitos T Citotóxicos/inmunología , Adolescente , Adulto , Autoantígenos/genética , Autoinmunidad/genética , Linfocitos T CD8-positivos/inmunología , Niño , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/genética , Femenino , Antígenos HLA-DQ/inmunología , Humanos , Inmunohistoquímica , Células Secretoras de Insulina/inmunología , Masculino , Sistemas de Lectura Abierta , Péptidos/genética , Biosíntesis de Proteínas , Adulto Joven
6.
Trends Endocrinol Metab ; 27(6): 353-362, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27094501

RESUMEN

Type 1 diabetes (T1D) is characterized by the selective and progressive destruction of insulin-producing beta cells by the immune system. An incomplete thymic selection against self-reactive islet antigens partly explains how these T cells reach the periphery and become diabetogenic. Increasing evidence suggest that beta cells themselves also participate to their own demise by generating neoepitopes that could be recognized by the immune surveillance machinery. In this regard, these T cells eradicate self-tissue by mechanisms analogous to a classical antitumor response. Cancer immunotherapy has exploited mutations and transcriptional and translational errors to trigger a specific antitumor response. In this opinion article, we aim at merging insight in antitumor immunology and autoimmunity to reveal processes that had previously been ignored to create beta cell-specific neoantigens.


Asunto(s)
Autoinmunidad/fisiología , Diabetes Mellitus Tipo 1/inmunología , Neoplasias/inmunología , Animales , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Inmunoterapia , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...