Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 176(4): 790-804.e13, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30661759

RESUMEN

The pancreatic islets of Langerhans regulate glucose homeostasis. The loss of insulin-producing ß cells within islets results in diabetes, and islet transplantation from cadaveric donors can cure the disease. In vitro production of whole islets, not just ß cells, will benefit from a better understanding of endocrine differentiation and islet morphogenesis. We used single-cell mRNA sequencing to obtain a detailed description of pancreatic islet development. Contrary to the prevailing dogma, we find islet morphology and endocrine differentiation to be directly related. As endocrine progenitors differentiate, they migrate in cohesion and form bud-like islet precursors, or "peninsulas" (literally "almost islands"). α cells, the first to develop, constitute the peninsular outer layer, and ß cells form later, beneath them. This spatiotemporal collinearity leads to the typical core-mantle architecture of the mature, spherical islet. Finally, we induce peninsula-like structures in differentiating human embryonic stem cells, laying the ground for the generation of entire islets in vitro.


Asunto(s)
Islotes Pancreáticos/citología , Islotes Pancreáticos/embriología , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias Humanas/citología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/citología , Islotes Pancreáticos/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Morfogénesis , Páncreas/citología
2.
Am J Physiol Endocrinol Metab ; 327(1): E27-E41, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690938

RESUMEN

Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and ß-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to ß-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal ß-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced ß-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve ß-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic ß-cells and controls islet glutamate clearance and ß-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents ß-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Transportador 2 de Aminoácidos Excitadores , Ácido Glutámico , Hiperglucemia , Islotes Pancreáticos , Transportador 2 de Aminoácidos Excitadores/metabolismo , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Ácido Glutámico/metabolismo , Hiperglucemia/metabolismo , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Femenino , Transporte de Proteínas , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Anciano , Adulto , Animales , Fosfatidilinositol 3-Quinasas/metabolismo
3.
FASEB J ; 37(8): e23106, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37498234

RESUMEN

The axon guidance proteins, Roundabout (Robo) receptors play a critical role in morphogenesis of the islets of Langerhans. Mice with a ß cell-selective deletion of Robo (Robo ßKO), show severely disrupted spatial architecture of their islets, without defects in ß cell differentiation or maturity. We have recently shown that Robo ßKO mice have reduced synchronous glucose-stimulated ß cell calcium oscillations in their islets in vivo, likely disrupting their pulsatile insulin secretion. Here, we analyze whole-body metabolic regulation in Robo ßKO mice. We show that Robo ßKO mice have mild defects in glucose homeostasis, and altered glucagon and insulin secretion. However, we did not observe any severe whole-body glucoregulatory phenotype following the disruption of islet architecture in Robo ßKO. Our data suggest that islet architecture plays only a mild role in overall glucoregulation.


Asunto(s)
Glucagón , Islotes Pancreáticos , Animales , Ratones , Glucagón/metabolismo , Secreción de Insulina , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Glucosa/metabolismo , Homeostasis
4.
Diabet Med ; 41(6): e15279, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38185936

RESUMEN

AIMS: Evidence is accumulating of the therapeutic benefits of mesenchymal stromal cells (MSCs) in diabetes-related conditions. We have identified a novel population of stromal cells within islets of Langerhans - islet stellate cells (ISCs) - which have a similar morphology to MSCs. In this study we characterize mouse ISCs and compare their morphology and function to MSCs to determine whether ISCs may also have therapeutic potential in diabetes. METHODS: ISCs isolated from mouse islets were compared to mouse bone marrow MSCs by analysis of cell morphology; expression of cell-surface markers and extracellular matrix (ECM) components; proliferation; apoptosis; paracrine activity; and differentiation into adipocytes, chondrocytes and osteocytes. We also assessed the effects of co-culture with ISCs or MSCs on the insulin secretory capacity of islet beta cells. RESULTS: Although morphological similar, ISCs were functionally distinct from MSCs. Thus, ISCs were less proliferative and more apoptotic; they had different expression levels of important paracrine factors; and they were less efficient at differentiation down multiple lineages. Co-culture of mouse islets with ISCs enhanced glucose induced insulin secretion more effectively than co-culture with MSCs. CONCLUSIONS: ISCs are a specific sub-type of islet-derived stromal cells that possess biological behaviors distinct from MSCs. The enhanced beneficial effects of ISCs on islet beta cell function suggests that they may offer a therapeutic target for enhancing beta cell functional survival in diabetes.


Asunto(s)
Diferenciación Celular , Técnicas de Cocultivo , Células Secretoras de Insulina , Islotes Pancreáticos , Células Madre Mesenquimatosas , Animales , Ratones , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Células Secretoras de Insulina/citología , Diferenciación Celular/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/fisiología , Proliferación Celular/fisiología , Insulina/metabolismo , Células Cultivadas , Secreción de Insulina/fisiología , Ratones Endogámicos C57BL , Masculino , Apoptosis/fisiología
5.
Diabet Med ; 41(2): e15257, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37968808

RESUMEN

AIMS: Pancreatic islet allotransplantation is an effective therapy for type 1 diabetes mellitus, restoring glycaemic control and hypoglycaemic awareness in patients with recurrent severe hypoglycaemia. Insulin independence following transplant is being increasingly reported; however, this is not a primary endpoint in the UK. Having surpassed 10 years of islet transplantation in Scotland, we aimed to evaluate the impact of insulin independence following transplant on metabolic outcomes and graft survival. METHODS: We conducted a retrospective analysis on data collected prospectively between 2011 and 2022. Patients who underwent islet transplantation in Scotland up to the 31st January 2020 were included. Primary endpoint was graft survival (stimulated C-peptide >50 pmol/L). Secondary endpoints included GOLD score, HbA1c, C-peptide and insulin requirement. Outcomes were compared between patients who achieved insulin independence at any point following transplant versus those who did not. RESULTS: 60 patients were included. 74.5% experienced >50 severe hypoglycaemic episodes in the year preceding transplant. There was a 55.0% decrease in insulin requirement following transplant and 30.0% achieved insulin independence. Mean graft survival time was 9.0 years (95% CI 7.2-10.9) in patients who achieved insulin independence versus 4.4 years (95% CI 3.4-5.3) in patients who did not. Insulin independence was associated with significantly improved graft function, glycaemic control and hypoglycaemic awareness at 1 year. CONCLUSIONS: This is the largest UK single-centre study on islet transplant to date. Our findings demonstrate significantly improved outcomes in patients who achieved insulin independence following islet transplantation.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hipoglucemia , Trasplante de Islotes Pancreáticos , Humanos , Insulina/uso terapéutico , Estudios Retrospectivos , Péptido C , Diabetes Mellitus Tipo 1/cirugía , Hipoglucemiantes/uso terapéutico , Hipoglucemia/prevención & control , Glucemia/metabolismo
6.
Int J Mol Sci ; 25(7)2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38612880

RESUMEN

Islets of Langerhans are anatomically dispersed within the pancreas and exhibit regulatory coordination between islets in response to nutritional and inflammatory stimuli. However, within individual islets, there is also multi-faceted coordination of function between individual beta-cells, and between beta-cells and other endocrine and vascular cell types. This is mediated partly through circulatory feedback of the major secreted hormones, insulin and glucagon, but also by autocrine and paracrine actions within the islet by a range of other secreted products, including somatostatin, urocortin 3, serotonin, glucagon-like peptide-1, acetylcholine, and ghrelin. Their availability can be modulated within the islet by pericyte-mediated regulation of microvascular blood flow. Within the islet, both endocrine progenitor cells and the ability of endocrine cells to trans-differentiate between phenotypes can alter endocrine cell mass to adapt to changed metabolic circumstances, regulated by the within-islet trophic environment. Optimal islet function is precariously balanced due to the high metabolic rate required by beta-cells to synthesize and secrete insulin, and they are susceptible to oxidative and endoplasmic reticular stress in the face of high metabolic demand. Resulting changes in paracrine dynamics within the islets can contribute to the emergence of Types 1, 2 and gestational diabetes.


Asunto(s)
Diabetes Gestacional , Islotes Pancreáticos , Femenino , Humanos , Embarazo , Insulina , Comunicación , Páncreas , Insulina Regular Humana
7.
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
8.
Am J Physiol Endocrinol Metab ; 325(5): E595-E609, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37729025

RESUMEN

Simultaneous activation of the incretin G-protein-coupled receptors (GPCRs) via unimolecular dual-receptor agonists (UDRA) has emerged as a new therapeutic approach for type 2 diabetes. Recent studies also advocate triple agonism with molecules also capable of binding the glucagon receptor. In this scoping review, we discuss the cellular mechanisms of action (MOA) underlying the actions of these novel and therapeutically important classes of peptide receptor agonists. Clinical efficacy studies of several UDRAs have demonstrated favorable results both as monotherapies and when combined with approved hypoglycemics. Although the additive insulinotropic effects of dual glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic peptide receptor (GIPR) agonism were anticipated based on the known actions of either glucagon-like peptide-1 (GLP-1) or glucose-dependent insulinotropic peptide (GIP) alone, the additional benefits from GCGR were largely unexpected. Whether additional synergistic or antagonistic interactions among these G-protein receptor signaling pathways arise from simultaneous stimulation is not known. The signaling pathways affected by dual- and tri-agonism require more trenchant investigation before a comprehensive understanding of the cellular MOA. This knowledge will be essential for understanding the chronic efficacy and safety of these treatments.


Asunto(s)
Diabetes Mellitus Tipo 2 , Islotes Pancreáticos , Humanos , Incretinas/farmacología , Incretinas/metabolismo , Polipéptido Inhibidor Gástrico/farmacología , Polipéptido Inhibidor Gástrico/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptores de Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo
9.
Biomed Microdevices ; 25(3): 35, 2023 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-37646842

RESUMEN

Microelectrode arrays (MEAs) have proven to be a powerful tool to study electrophysiological processes over the last decades with most technology developed for investigation of the heart or brain. Other targets in the field of bioelectronic medicine are the peripheral nervous system and its innervation of various organs. Beyond the heart and nervous systems, the beta cells of the pancreatic islets of Langerhans generate action potentials during the production of insulin. In vitro experiments have demonstrated that their activity is a biomarker for blood glucose levels, suggesting that recording their activity in vivo could support patients suffering from diabetes mellitus with long-term automated read-out of blood glucose concentrations. Here, we present a flexible polymer-based implant having 64 low impedance microelectrodes designed to be implanted to a depth of 10 mm into the pancreas. As a first step, the implant will be used in acute experiments in pigs to explore the electrophysiological processes of the pancreas in vivo. Beyond use in the pancreas, our flexible implant and simple implantation method may also be used in other organs such as the brain.


Asunto(s)
Glucemia , Islotes Pancreáticos , Animales , Porcinos , Insulina , Encéfalo , Electrofisiología
10.
J Periodontal Res ; 58(6): 1290-1299, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37723987

RESUMEN

BACKGROUND AND OBJECTIVE: The purpose of this study was to determine if chronic periodontitis (CP) may induce hyperinsulinemia and may have the effect of on pancreatic ß-cell proliferation in a rat model. MATERIALS AND METHODS: Twelve male Sprague-Dawley rats were divided into two groups: the CP group and the control group (Con group). The following contents were evaluated: pathological changes in periodontal soft and hard tissues; serum lipopolysaccharide (LPS) level, serum fasting insulin (FINS) level, fasting blood glucose (FBG) level, and homeostasis model assessment (HOMA) ß (HOMA-ß) index; histopathological examination of islets; immunohistochemistry of insulin and p-Smad2 expression in islets; immunofluorescence of changes in the relative number of ß-cells and the number of Ki67-positive ß-cells. Western blotting was used to analyze p-Smad2/Smad2 levels. Results were analyzed by two independent samples t tests. RESULTS: Increased serum LPS level, FINS level, and HOMA-ß index were observed in the rats of the CP group; FBG level did not change significantly; histological assessments showed an enlarged islet area, increased insulin content, relatively increased ß-cells, increased Ki67-positive ß-cells, and decreased p-Smad2 expression in islets in the rats of the CP group. CONCLUSION: Our study results link CP-induced hyperinsulinemia with changes in islets, such as islet hyperplasia and compensatory ß-cell proliferation, by using a CP rat model.


Asunto(s)
Periodontitis Crónica , Hiperinsulinismo , Islotes Pancreáticos , Ratas , Masculino , Animales , Islotes Pancreáticos/patología , Ratas Sprague-Dawley , Periodontitis Crónica/metabolismo , Antígeno Ki-67/metabolismo , Lipopolisacáridos/farmacología , Hiperinsulinismo/complicaciones , Hiperinsulinismo/metabolismo , Insulina , Glucemia/metabolismo
11.
Ter Arkh ; 95(10): 896-902, 2023 Nov 23.
Artículo en Ruso | MEDLINE | ID: mdl-38159024

RESUMEN

Leonid Vasilyevich Sobolev is an outstanding pathologist, whose name definitely should occupy an honorable place in the galaxy of great scientists of Russia. However, his name was undeservedly forgotten, and the role of his work was underestimated. This Russian scientist made the most important discovery: he proved in an experiment that it is the islets of Langerhans in the pancreas that secrete a humoral regulator, "factor X", the deficiency of which leads to diabetes mellitus. This mysterious islet factor will be isolated from the pancreas of dogs in 1921 by the future Nobel laureate Frederick G. Banting and will become part of the medicine as insulin. However, there is every reason to believe that the discovery of F. G. Banting, who repeated a series of experiments by Leonid Vasilyevich Sobolev, is, in fact, secondary, which is the research subject in this paper.


Asunto(s)
Diabetes Mellitus , Premio Nobel , Animales , Perros , Insulina , Páncreas , Federación de Rusia
12.
J Proteome Res ; 21(4): 1167-1174, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35293755

RESUMEN

Filter-aided sample preparation (FASP) is widely used in bottom-up proteomics for tryptic digestion. However, the sample recovery yield of this method is limited by the amount of the starting material. While ∼100 ng of digested protein is sufficient for thorough protein identification, proteomic information gets lost with a protein content <10 µg due to incomplete peptide recovery from the filter. We developed and optimized a flexible well-plate µFASP device and protocol that is suitable for an ∼1 µg protein sample. In 1 µg of HeLa digest, we identified 1295 ± 10 proteins with µFASP followed by analysis with liquid chromatography-mass spectrometry. In contrast, only 524 ± 5 proteins were identified with the standard FASP protocol, while 1395 ± 4 proteins were identified in 20 µg after standard FASP as a benchmark. Furthermore, we conducted a combined peptidomic and proteomic study of single pancreatic islets with well-plate µFASP. Here, we separated neuropeptides and digested the remaining on-filter proteins for bottom-up proteomic analysis. Our results indicate inter-islet heterogeneity for the expression of proteins involved in glucose catabolism, pancreatic hormone processing, and secreted peptide hormones. We consider our method to provide a useful tool for proteomic characterization of samples where the biological material is scarce. All proteomic data are available under DOI: 10.6019/PXD029039.


Asunto(s)
Islotes Pancreáticos , Proteómica , Cromatografía Liquida/métodos , Humanos , Islotes Pancreáticos/química , Espectrometría de Masas , Proteínas/análisis , Proteómica/métodos
13.
Diabetologia ; 65(9): 1519-1533, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35616696

RESUMEN

AIMS/HYPOTHESIS: Pancreatic islets depend on cytosolic calcium (Ca2+) to trigger the secretion of glucoregulatory hormones and trigger transcriptional regulation of genes important for islet response to stimuli. To date, there has not been an attempt to profile Ca2+-regulated gene expression in all islet cell types. Our aim was to construct a large single-cell transcriptomic dataset from human islets exposed to conditions that would acutely induce or inhibit intracellular Ca2+ signalling, while preserving biological heterogeneity. METHODS: We exposed intact human islets from three donors to the following conditions: (1) 2.8 mmol/l glucose; (2) 16 mmol/l glucose and 40 mmol/l KCl to maximally stimulate Ca2+ signalling; and (3) 16 mmol/l glucose, 40 mmol/l KCl and 5 mmol/l EGTA (Ca2+ chelator) to inhibit Ca2+ signalling, for 1 h. We sequenced 68,650 cells from all islet cell types, and further subsetted the cells to form an endocrine cell-specific dataset of 59,373 cells expressing INS, GCG, SST or PPY. We compared transcriptomes across conditions to determine the differentially expressed Ca2+-regulated genes in each endocrine cell type, and in each endocrine cell subcluster of alpha and beta cells. RESULTS: Based on the number of Ca2+-regulated genes, we found that each alpha and beta cell cluster had a different magnitude of Ca2+ response. We also showed that polyhormonal clusters expressing both INS and GCG, or both INS and SST, are defined by Ca2+-regulated genes specific to each cluster. Finally, we identified the gene PCDH7 from the beta cell clusters that had the highest number of Ca2+-regulated genes, and showed that cells expressing cell surface PCDH7 protein have enhanced glucose-stimulated insulin secretory function. CONCLUSIONS/INTERPRETATION: Here we use our large-scale, multi-condition, single-cell dataset to show that human islets have cell-type-specific Ca2+-regulated gene expression profiles, some of them specific to subpopulations. In our dataset, we identify PCDH7 as a novel marker of beta cells having an increased number of Ca2+-regulated genes and enhanced insulin secretory function. DATA AVAILABILITY: A searchable and user-friendly format of the data in this study, specifically designed for rapid mining of single-cell RNA sequencing data, is available at https://lynnlab.shinyapps.io/Human_Islet_Atlas/ . The raw data files are available at NCBI Gene Expression Omnibus (GSE196715).


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Calcio/metabolismo , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo
14.
Diabetes Obes Metab ; 24 Suppl 1: 5-16, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431589

RESUMEN

When, in 1869, Paul Langerhans detected the "islands of tissue" in the pancreas, he took the first step on a journey towards islet transplantation as a treatment for type 1 diabetes. The route has embraced developments across biosciences, surgery, gene therapy and clinical research. This review highlights major milestones along that journey involving whole pancreas transplantation, islet transplantation, the creation of surrogate insulin-secreting cells and novel islet-like structures using genetic and bio-engineering technologies. To obviate the paucity of human tissue, pluripotent stem cells and non-ß-cells within the pancreas have been modified to create physiologically responsive insulin-secreting cells. Before implantation, these can be co-cultured with endothelial cells to promote vascularisation and with immune defence cells such as placental amnion cells to reduce immune rejection. Scaffolds to contain grafts and facilitate surgical placement provide further opportunities to achieve physiological insulin delivery. Alternatively, xenotransplants such as porcine islets might be reconsidered as opportunities exist to circumvent safety concerns and immune rejection. Thus, despite a long and arduous journey, the prospects for increased use of tissue transplantation to provide physiological insulin replacement are drawing ever closer.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Animales , Diabetes Mellitus Tipo 1/cirugía , Células Endoteliales , Femenino , Humanos , Insulina , Trasplante de Islotes Pancreáticos/efectos adversos , Masculino , Placenta , Embarazo , Porcinos
15.
Int J Mol Sci ; 23(12)2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35742878

RESUMEN

Gestational diabetes (GDM) is characterized by a glucose tolerance disorder. This may first appear during pregnancy or pre-exist before conception as a form of prediabetes, but there are few data on the pathogenesis of the latter subtype. Female New Zealand obese (NZO) mice serve as a model for this subpopulation of GDM. It was recently shown that GDM is associated with elevated urinary serotonin (5-hydroxytryptamine, 5-HT) levels, but the role of the biogenic amine in subpopulations with prediabetes remains unclear. 5-HT is synthesized in different tissues, including the islets of Langerhans during pregnancy. Furthermore, 5-HT receptors (HTRs) are expressed in tissues important for the regulation of glucose homeostasis, such as liver and pancreas. Interestingly, NZO mice showed elevated plasma and islet 5-HT concentrations as well as impaired glucose-stimulated 5-HT secretion. Incubation of isolated primary NZO islets with 5-HT revealed an inhibitory effect on insulin and glucagon secretion. In primary NZO hepatocytes, 5-HT aggravated hepatic glucose production (HGP), decreased glucose uptake (HGU), glycogen content, and modulated AKT activation as well as cyclic adenosine monophosphate (cAMP) increase, indicating 5-HT downstream modulation. Treatment with an HTR2B antagonist reduced this 5-HT-mediated deterioration of the metabolic state. With its strong effect on glucose metabolism, these data indicate that 5-HT is already a potential indicator of GDM before conception in mice.


Asunto(s)
Diabetes Gestacional , Islotes Pancreáticos , Estado Prediabético , Animales , Diabetes Gestacional/metabolismo , Femenino , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Hígado/metabolismo , Ratones , Ratones Obesos , Estado Prediabético/metabolismo , Embarazo , Serotonina/metabolismo
16.
Am J Transplant ; 21(11): 3725-3733, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33961335

RESUMEN

To describe the 10-year outcomes of islet transplantation within the Swiss-French GRAGIL Network, in patients with type 1 diabetes experiencing high glucose variability associated with severe hypoglycemia and/or with functional kidney graft. We conducted a retrospective analysis of all subjects transplanted in the GRAGIL-1c and GARGIL-2 islet transplantation trials and analyzed components of metabolic control, graft function and safety outcomes over the 10-year period of follow-up. Forty-four patients were included between September 2003 and April 2010. Thirty-one patients completed a 10-year follow-up. Ten years after islet transplantation, median HbA1c was 7.2% (6.2-8.0) (55 mmol/mol [44-64]) versus 8.0% (7.1-9.1) (64 mmol/mol [54-76]) before transplantation (p < .001). Seventeen of 23 (73.9%) recipients were free of severe hypoglycemia, 1/21 patients (4.8%) was insulin-independent and median C-peptide was 0.6 ng/ml (0.2-1.2). Insulin requirements (UI/kg/day) were 0.3 (0.1-0.5) versus 0.5 (0.4-0.6) before transplantation (p < .001). Median (IQR) ß-score was 1 (0-4) (p < .05 when comparing with pre-transplantation values) and 51.9% recipients had a functional islet graft at 10 years. With a 10-year follow-up in a multicentric network, islet transplantation provided sustained improvement of glycemic control and was efficient to prevent severe hypoglycemia in almost 75% of the recipients.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Glucemia , Diabetes Mellitus Tipo 1/cirugía , Humanos , Estudios Retrospectivos , Suiza , Resultado del Tratamiento
17.
Am J Transplant ; 21(1): 297-306, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32524720

RESUMEN

In order to assess ß cell secretory capacity after islet transplantation, standardized mixed meal stimulation tests are often used. But these tests are cumbersome and the effect of exogenous insulin on the test results is unclear. The aim of our study was to determine to what extent fasting glycemic indices can estimate stimulated ß cell function in islet transplant recipients with and without basal insulin. In total 100 mixed meal stimulation tests, including 31 with concurrent basal insulin treatment, were performed in 36 islet transplant recipients. In a multivariate model, fasting C-peptide and fasting glucose together estimated peak C-peptide with R2  = .87 and area under the curve (AUC) C-peptide with a R2  = .93. There was a larger increase of glucose during tests in which exogenous insulin was used (+7.9 vs +5.3 mmol/L, P < .001) and exogenous insulin use was associated with a slightly lower estimated peak C-peptide (relative change: -15%, P = .02). In islet transplant recipients the combination of fasting C-peptide and glucose can be used to accurately estimate stimulated ß cell function after a mixed meal stimulation test, whether exogenous basal insulin is present or not. These data indicate that graft function can be reliably determined during exogenous insulin treatment and that regular islet graft stimulation tests can be minimized.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos , Glucemia , Péptido C , Ayuno , Humanos , Insulina
18.
Am J Transplant ; 21(10): 3411-3420, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33754431

RESUMEN

In this single-center, retrospective cohort study, we aimed to elucidate simple metabolic markers or surrogate indices of ß-cell function that best predict long-term insulin independence and goal glycemic HbA1c control (HbA1c ≤ 6.5%) after total pancreatectomy with islet autotransplantation (TP-IAT). Patients who underwent TP-IAT (n = 371) were reviewed for metabolic measures before TP-IAT and for insulin independence and glycemic control at 1, 3, and 5 years after TP-IAT. Insulin independence and goal glycemic control were achieved in 33% and 68% at 1 year, respectively. Although the groups who were insulin independent and dependent overlap substantially on baseline measures, an individual who has abnormal glycemia (prediabetes HbA1c or fasting glucose) or estimated IEQs/kg < 2500 has a very high likelihood of remaining insulin dependent after surgery. In multivariate logistic regression modelling, metabolic measures correctly predicted insulin independence in about 70% of patients at 1, 3, and 5 years after TP-IAT. In conclusion, metabolic testing measures before surgery are highly associated with diabetes outcomes after TP-IAT at a population level and correctly predict outcomes in approximately two out of three patients. These findings may aid in prognostic counseling for chronic pancreatitis patients who are likely to eventually need TP-IAT.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Pancreatitis Crónica , Humanos , Pancreatectomía , Pancreatitis Crónica/cirugía , Estudios Retrospectivos , Trasplante Autólogo , Resultado del Tratamiento
19.
Am J Transplant ; 21(11): 3714-3724, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34033222

RESUMEN

Several cytokines and chemokines are elevated after islet infusion in patients undergoing total pancreatectomy with islet autotransplantation (TPIAT), including CXCL8 (also known as interleukin-8), leading to islet loss. We investigated whether use of reparixin for blockade of the CXCL8 pathway would improve islet engraftment and insulin independence after TPIAT. Adults without diabetes scheduled for TPIAT at nine academic centers were randomized to a continuous infusion of reparixin or placebo (double-blinded) for 7 days in the peri-transplant period. Efficacy measures included insulin independence (primary), insulin dose, hemoglobin A1c (HbA1c ), and mixed meal tolerance testing. The intent-to-treat population included 102 participants (age 39.5 ± 12.2 years, 69% female), n = 50 reparixin-treated, n = 52 placebo-treated. The proportion insulin-independent at Day 365 was similar in reparixin and placebo: 20% vs. 21% (p = .542). Twenty-seven of 42 (64.3%) in the reparixin group and 28/45 (62.2%) in the placebo group maintained HbA1c ≤6.5% (p = .842, Day 365). Area under the curve C-peptide from mixed meal testing was similar between groups, as were adverse events. In conclusion, reparixin infusion did not improve diabetes outcomes. CXCL8 inhibition alone may be insufficient to prevent islet damage from innate inflammation in islet autotransplantation. This first multicenter clinical trial in TPIAT highlights the potential for future multicenter collaborations.


Asunto(s)
Diabetes Mellitus , Trasplante de Islotes Pancreáticos , Pancreatitis Crónica , Receptores de Interleucina-8A/antagonistas & inhibidores , Receptores de Interleucina-8B/antagonistas & inhibidores , Adulto , Péptido C , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pancreatectomía , Pancreatitis Crónica/cirugía , Receptores de Trasplantes , Trasplante Autólogo , Resultado del Tratamiento
20.
Am J Transplant ; 21(3): 1027-1038, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32865886

RESUMEN

Islet transplantation is an effective therapy for life-threatening hypoglycemia, but graft function gradually declines over time in many recipients. We characterized islet-specific T cells in recipients within an islet transplant program favoring alemtuzumab (ATZ) lymphodepleting induction and examined associations with graft function. Fifty-eight recipients were studied: 23 pretransplant and 40 posttransplant (including 5 with pretransplant phenotyping). The proportion with islet-specific T cell responses was not significantly different over time (pre-Tx: 59%; 1-6 m posttransplant: 38%; 7-12 m: 44%; 13-24 m: 47%; and >24 m: 45%). However, phenotype shifted significantly, with IFN-γ-dominated response in the pretransplant group replaced by IL-10-dominated response in the 1-6 m posttransplant group, reverting to predominantly IFN-γ-oriented response in the >24 m group. Clustering analysis of posttransplant responses revealed two main agglomerations, characterized by IFN-γ and IL-10 phenotypes, respectively. IL-10-oriented posttransplant response was associated with relatively low graft function. Recipients within the IL-10+ cluster had a significant decline in C-peptide levels in the period preceding the IL-10 response, but stable graft function following the response. In contrast, an IFN-γ response was associated with subsequently decreased C-peptide. Islet transplantation favoring ATZ induction is associated with an initial altered islet-specific T cell phenotype but reversion toward pretransplant profiles over time. Posttransplant autoreactive T cell phenotype may be a predictor of subsequent graft function.


Asunto(s)
Diabetes Mellitus Tipo 1 , Trasplante de Células Madre Hematopoyéticas , Trasplante de Islotes Pancreáticos , Alemtuzumab/uso terapéutico , Supervivencia de Injerto , Humanos , Fenotipo , Linfocitos T
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA