Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 299
Filtrar
1.
Trends Endocrinol Metab ; 35(6): 466-477, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38861922

RESUMO

The current paradigm for the insulin system focuses on the phenomenon of glucose-stimulated insulin secretion and insulin action on blood glucose control. This historical glucose-centric perspective may have introduced a conceptual bias in our understanding of insulin regulation. A body of evidence demonstrating that in vivo variations in blood glucose and insulin secretion can be largely dissociated motivated us to reconsider the fundamental design of the insulin system as a control system for metabolic homeostasis. Here, we propose that a minimal glucose-centric model does not accurately describe the physiological behavior of the insulin system and propose a new paradigm focusing on the effects of incretins, arguing that under fasting conditions, insulin is regulated by an adipoincretin effect.


Assuntos
Secreção de Insulina , Insulina , Humanos , Secreção de Insulina/fisiologia , Insulina/metabolismo , Animais , Homeostase/fisiologia , Incretinas/metabolismo , Glicemia/metabolismo
2.
Front Endocrinol (Lausanne) ; 15: 1402880, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38883608

RESUMO

Background: Pancreatic islets are important in nutrient homeostasis and improved cellular models of clonal origin may very useful especially in view of relatively scarce primary material. Close 3D contact and coupling between ß-cells are a hallmark of physiological function improving signal/noise ratios. Extracellular electrophysiology using micro-electrode arrays (MEA) is technically far more accessible than single cell patch clamp, enables dynamic monitoring of electrical activity in 3D organoids and recorded multicellular slow potentials (SP) provide unbiased insight in cell-cell coupling. Objective: We have therefore asked whether 3D spheroids enhance clonal ß-cell function such as electrical activity and hormone secretion using human EndoC-ßH1, EndoC-ßH5 and rodent INS-1 832/13 cells. Methods: Spheroids were formed either by hanging drop or proprietary devices. Extracellular electrophysiology was conducted using multi-electrode arrays with appropriate signal extraction and hormone secretion measured by ELISA. Results: EndoC-ßH1 spheroids exhibited increased signals in terms of SP frequency and especially amplitude as compared to monolayers and even single cell action potentials (AP) were quantifiable. Enhanced electrical signature in spheroids was accompanied by an increase in the glucose stimulated insulin secretion index. EndoC-ßH5 monolayers and spheroids gave electrophysiological profiles similar to EndoC-ßH1, except for a higher electrical activity at 3 mM glucose, and exhibited moreover a biphasic profile. Again, physiological concentrations of GLP-1 increased AP frequency. Spheroids also exhibited a higher secretion index. INS-1 cells did not form stable spheroids, but overexpression of connexin 36, required for cell-cell coupling, increased glucose responsiveness, dampened basal activity and consequently augmented the stimulation index. Conclusion: In conclusion, spheroid formation enhances physiological function of the human clonal ß-cell lines and these models may provide surrogates for primary islets in extracellular electrophysiology.


Assuntos
Células Secretoras de Insulina , Esferoides Celulares , Humanos , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citologia , Fenômenos Eletrofisiológicos , Secreção de Insulina/fisiologia , Glucose/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Potenciais de Ação/fisiologia , Animais
3.
Rev Med Suisse ; 20(876): 1069-1073, 2024 May 29.
Artigo em Francês | MEDLINE | ID: mdl-38812338

RESUMO

C-peptide measurement allows an estimation of the residual endogenous insulin secretion in diabetic patients. Nowadays plasmatic testing is convenient and unexpensive, but we lack standardized tests. Therefore, there are no official recommendation regarding its use. As an indication, in some circumstances, C-peptide measurement could be used to specify the type of diabetes, help guide the treatment strategy and potentially assess the risk for complications. Its use is still limited and not recommended on a routine base for all patients living with diabetes, but in the future, tests standardization and establishment of reference ranges could give more insight on the clinical relevance of C-peptide measurement.


Le dosage du peptide-C est une mesure permettant d'évaluer la sécrétion endogène résiduelle d'insuline chez les patients diabétiques. Le dosage plasmatique est facilement réalisable actuellement, pour un coût modeste, mais l'absence de standardisation des tests ne permet pas d'émettre des recommandations officielles par rapport à son utilisation. À titre indicatif, dans certaines situations, le dosage du peptide-C peut être utilisé pour préciser le type de diabète, guider les traitements médicamenteux et potentiellement évaluer les risques de complications. Son utilisation est pour le moment limitée et n'est pas recommandée en routine pour tous les patients atteints de diabète, mais à l'avenir, la formalisation du dosage et l'établissement de valeurs de référence pourraient permettre de définir son utilisation clinique.


Assuntos
Peptídeo C , Secreção de Insulina , Insulina , Humanos , Peptídeo C/sangue , Peptídeo C/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Diabetes Mellitus/sangue , Diabetes Mellitus/metabolismo , Diabetes Mellitus/diagnóstico
5.
Diabetes ; 73(6): 856-863, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38768366

RESUMO

An agreed-upon consensus model of glucose-stimulated insulin secretion from healthy ß-cells is essential for understanding diabetes pathophysiology. Since the discovery of the KATP channel in 1984, an oxidative phosphorylation (OxPhos)-driven rise in ATP has been assumed to close KATP channels to initiate insulin secretion. This model lacks any evidence, genetic or otherwise, that mitochondria possess the bioenergetics to raise the ATP/ADP ratio to the triggering threshold, and conflicts with genetic evidence demonstrating that OxPhos is dispensable for insulin secretion. It also conflates the stoichiometric yield of OxPhos with thermodynamics, and overestimates OxPhos by failing to account for established features of ß-cell metabolism, such as leak, anaplerosis, cataplerosis, and NADPH production that subtract from the efficiency of mitochondrial ATP production. We have proposed an alternative model, based on the spatial and bioenergetic specializations of ß-cell metabolism, in which glycolysis initiates insulin secretion. The evidence for this model includes that 1) glycolysis has high control strength over insulin secretion; 2) glycolysis is active at the correct time to explain KATP channel closure; 3) plasma membrane-associated glycolytic enzymes control KATP channels; 4) pyruvate kinase has favorable bioenergetics, relative to OxPhos, for raising ATP/ADP; and 5) OxPhos stalls before membrane depolarization and increases after. Although several key experiments remain to evaluate this model, the 1984 model is based purely on circumstantial evidence and must be rescued by causal, mechanistic experiments if it is to endure.


Assuntos
Glucose , Secreção de Insulina , Células Secretoras de Insulina , Insulina , Canais KATP , Fosforilação Oxidativa , Células Secretoras de Insulina/metabolismo , Humanos , Glucose/metabolismo , Canais KATP/metabolismo , Canais KATP/genética , Secreção de Insulina/fisiologia , Animais , Insulina/metabolismo , Glicólise/fisiologia , Modelos Biológicos , Trifosfato de Adenosina/metabolismo
6.
Diabetes ; 73(6): 849-855, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38768365

RESUMO

The canonical model of glucose-induced increase in insulin secretion involves the metabolism of glucose via glycolysis and the citrate cycle, resulting in increased ATP synthesis by the respiratory chain and the closure of ATP-sensitive K+ (KATP) channels. The resulting plasma membrane depolarization, followed by Ca2+ influx through L-type Ca2+ channels, then induces insulin granule fusion. Merrins and colleagues have recently proposed an alternative model whereby KATP channels are controlled by pyruvate kinase, using glycolytic and mitochondrial phosphoenolpyruvate (PEP) to generate microdomains of high ATP/ADP immediately adjacent to KATP channels. This model presents several challenges. First, how mitochondrially generated PEP, but not ATP produced abundantly by the mitochondrial F1F0-ATP synthase, can gain access to the proposed microdomains is unclear. Second, ATP/ADP fluctuations imaged immediately beneath the plasma membrane closely resemble those in the bulk cytosol. Third, ADP privation of the respiratory chain at high glucose, suggested to drive alternating, phased-locked generation by mitochondria of ATP or PEP, has yet to be directly demonstrated. Finally, the approaches used to explore these questions may be complicated by off-target effects. We suggest instead that Ca2+ changes, well known to affect both ATP generation and consumption, likely drive cytosolic ATP/ADP oscillations that in turn regulate KATP channels and membrane potential. Thus, it remains to be demonstrated that a new model is required to replace the existing, mitochondrial bioenergetics-based model.


Assuntos
Glucose , Células Secretoras de Insulina , Canais KATP , Células Secretoras de Insulina/metabolismo , Canais KATP/metabolismo , Glucose/metabolismo , Humanos , Animais , Trifosfato de Adenosina/metabolismo , Mitocôndrias/metabolismo , Insulina/metabolismo , Difosfato de Adenosina/metabolismo , Modelos Biológicos , Secreção de Insulina/fisiologia
7.
PLoS Comput Biol ; 20(5): e1012130, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38739680

RESUMO

Within the islets of Langerhans, beta cells orchestrate synchronized insulin secretion, a pivotal aspect of metabolic homeostasis. Despite the inherent heterogeneity and multimodal activity of individual cells, intercellular coupling acts as a homogenizing force, enabling coordinated responses through the propagation of intercellular waves. Disruptions in this coordination are implicated in irregular insulin secretion, a hallmark of diabetes. Recently, innovative approaches, such as integrating multicellular calcium imaging with network analysis, have emerged for a quantitative assessment of the cellular activity in islets. However, different groups use distinct experimental preparations, microscopic techniques, apply different methods to process the measured signals and use various methods to derive functional connectivity patterns. This makes comparisons between findings and their integration into a bigger picture difficult and has led to disputes in functional connectivity interpretations. To address these issues, we present here a systematic analysis of how different approaches influence the network representation of islet activity. Our findings show that the choice of methods used to construct networks is not crucial, although care is needed when combining data from different islets. Conversely, the conclusions drawn from network analysis can be heavily affected by the pre-processing of the time series, the type of the oscillatory component in the signals, and by the experimental preparation. Our tutorial-like investigation aims to resolve interpretational issues, reconcile conflicting views, advance functional implications, and encourage researchers to adopt connectivity analysis. As we conclude, we outline challenges for future research, emphasizing the broader applicability of our conclusions to other tissues exhibiting complex multicellular dynamics.


Assuntos
Ilhotas Pancreáticas , Ilhotas Pancreáticas/fisiologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/citologia , Animais , Biologia Computacional/métodos , Camundongos , Insulina/metabolismo , Humanos , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/citologia , Secreção de Insulina/fisiologia , Modelos Biológicos , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia
8.
Am J Physiol Endocrinol Metab ; 326(5): E663-E672, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38568150

RESUMO

Despite the fact that genes and the environment are known to play a central role in islet function, our knowledge of how these parameters interact to modulate insulin secretory function remains relatively poor. Presently, we performed ex vivo glucose-stimulated insulin secretion and insulin content assays in islets of 213 mice from 13 inbred mouse strains on chow, Western diet (WD), and a high-fat, carbohydrate-free (KETO) diet. Strikingly, among these 13 strains, islets from the commonly used C57BL/6J mouse strain were the least glucose responsive. Using matched metabolic phenotyping data, we performed correlation analyses of isolated islet parameters and found a positive correlation between basal and glucose-stimulated insulin secretion, but no relationship between insulin secretion and insulin content. Using in vivo metabolic measures, we found that glucose tolerance determines the relationship between ex vivo islet insulin secretion and plasma insulin levels. Finally, we showed that islet glucose-stimulated insulin secretion decreased with KETO in almost all strains, concomitant with broader phenotypic changes, such as increased adiposity and glucose intolerance. This is an important finding as it should caution against the application of KETO diet for beta-cell health. Together these data offer key insights into the intersection of diet and genetic background on islet function and whole body glucose metabolism.NEW & NOTEWORTHY Thirteen strains of mice on chow, Western diet, and high-fat, carbohydrate-free (KETO), correlating whole body phenotypes to ex vivo pancreatic islet functional measurements, were used. The study finds a huge spectrum of functional islet responses and insulin phenotypes across all strains and diets, with the ubiquitous C57Bl/6J mouse exhibiting the lowest secretory response of all strains, highlighting the overall importance of considering genetic background when investigating islet function. Ex vivo basal and stimulated insulin secretion are correlated in the islet, and KETO imparts widescale downregulation of islet insulin secretion.


Assuntos
Dieta Hiperlipídica , Secreção de Insulina , Insulina , Ilhotas Pancreáticas , Camundongos Endogâmicos C57BL , Animais , Camundongos , Ilhotas Pancreáticas/metabolismo , Secreção de Insulina/fisiologia , Insulina/metabolismo , Insulina/sangue , Masculino , Dieta Ocidental , Glucose/metabolismo , Dieta com Restrição de Carboidratos , Camundongos Endogâmicos , Glicemia/metabolismo , Intolerância à Glucose/metabolismo , Intolerância à Glucose/genética
9.
Diabetologia ; 67(6): 1079-1094, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38512414

RESUMO

AIMS/HYPOTHESIS: Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility, which we explore here by focusing on the imprinted gene Nnat (encoding neuronatin [NNAT]), which is required for normal insulin synthesis and secretion. METHODS: Single-cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing enhanced GFP under the control of the Nnat enhancer/promoter regions were generated for FACS of beta cells and downstream analysis of CpG methylation by bisulphite sequencing and RNA-seq, respectively. Animals deleted for the de novo methyltransferase DNA methyltransferase 3 alpha (DNMT3A) from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 AM and Cal-590 AM. Insulin secretion was measured using homogeneous time-resolved fluorescence imaging. RESULTS: Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic datasets demonstrated the early establishment of Nnat-positive and -negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a subpopulation specialised for insulin production, and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialisation. CONCLUSIONS/INTERPRETATION: These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes. DATA AVAILABILITY: The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD048465.


Assuntos
Ilhas de CpG , Metilação de DNA , Células Secretoras de Insulina , Células Secretoras de Insulina/metabolismo , Animais , Camundongos , Ilhas de CpG/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Camundongos Transgênicos , DNA Metiltransferase 3A/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina/fisiologia
10.
Diabet Med ; 41(6): e15279, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38185936

RESUMO

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.


Assuntos
Diferenciação Celular , Técnicas de Cocultura , Células Secretoras de Insulina , Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Animais , Camundongos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/citologia , Diferenciação Celular/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/fisiologia , Proliferação de Células/fisiologia , Insulina/metabolismo , Células Cultivadas , Secreção de Insulina/fisiologia , Camundongos Endogâmicos C57BL , Masculino , Apoptose/fisiologia
11.
Front Endocrinol (Lausanne) ; 14: 1144834, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36909323

RESUMO

Aim: Clinical heterogeneity exists in overall obesity and abdominal obesity in terms of insulin secretion and sensitivity. Further, the impact of visceral fat (VF) on the first- and second-phase insulin secretion (FPIS and SPIS) is controversial. We aim to investigate insulin secretion and sensitivity in Chinese patients with T2DM according to different BMI and VF levels. Methods: This study enrolled 300 participants. A dual bioelectrical impedance analyzer was used to assess the visceral and subcutaneous fat area (VFA and SFA). VF levels were categorized as normal or high, with the cutoff value of 100 cm2. FPIS and SPIS were evaluated by arginine stimulation test and standardized steamed bread meal tolerance test, respectively. ß-cell function (HOMA2-ß), insulin resistance (HOMA2-IR), and Gutt's insulin sensitivity index (Gutt-ISI) were also calculated. Spearman's correlation analysis and multivariate linear regression analysis were adopted for statistical analysis. Results: Participants were categorized into four groups: normal weight-normal VF, normal weight-high VF, overweight/obese-normal VF and overweight/obese-high VF. Multivariate linear regression showed that both VFA and SFA were correlated with FPIS, HOMA2-IR and Gutt-ISI after controlling for gender and diabetes duration. After further adjustment for BMI and VFA, some associations of SFA with insulin secretion and sensitivity disappeared. After adjustment for gender, diabetes duration, BMI and SFA, VFA was positively correlated with FPIS, SPIS and HOMA2-IR. Subjects with overweight/obese-high VF were more likely to have higher FPIS, HOMA2-IR and lower Gutt-ISI (all p < 0.05). Conclusion: VF affects both FPIS and SPIS, and worsens insulin sensitivity independent of BMI and subcutaneous fat in Chinese patients with T2DM. Clinical trial registration: http://www.chictr.org.cn, identifier ChiCTR2200062884.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Humanos , Diabetes Mellitus Tipo 2/complicações , Resistência à Insulina/fisiologia , Secreção de Insulina/fisiologia , Gordura Intra-Abdominal/química , Sobrepeso/complicações , Índice de Massa Corporal , População do Leste Asiático , Glicemia/análise , Obesidade/complicações , Gordura Subcutânea/química
12.
Methods Mol Biol ; 2592: 101-111, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36507988

RESUMO

The role of Zn2+ ions in proper storage of insulin in ß-cell granules is well-established so when insulin is secreted from ß-cells stimulated by an increase in plasma glucose, free Zn2+ ions are also released. This local increase in Zn2+ can be detected in the pancreas of rodents in real time by the use of a zinc-responsive MR contrast agent. This method offers the opportunity to monitor ß-cell function longitudinally in live rodents. The methods used in our lab are fully described in this short report and some MR images of a rat pancreas showing clearly enhanced hot spots in the tail are presented.


Assuntos
Células Secretoras de Insulina , Roedores , Ratos , Animais , Secreção de Insulina/fisiologia , Roedores/metabolismo , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Pâncreas/diagnóstico por imagem , Pâncreas/metabolismo
13.
Mol Metab ; 66: 101625, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36374631

RESUMO

OBJECTIVE: The endocrine pancreatic ß-cells play a pivotal role in maintaining whole-body glucose homeostasis and its dysregulation is a consistent feature in all forms of diabetes. However, knowledge of intracellular regulators that modulate ß-cell function remains incomplete. We investigated the physiological role of ROCK1 in the regulation of insulin secretion and glucose homeostasis. METHODS: Mice lacking ROCK1 in pancreatic ß-cells (RIP-Cre; ROCK1loxP/loxP, ß-ROCK1-/-) were studied. Glucose and insulin tolerance tests as well as glucose-stimulated insulin secretion (GSIS) were measured. An insulin secretion response to a direct glucose or pyruvate or pyruvate kinase (PK) activator stimulation in isolated islets from ß-ROCK1-/- mice or ß-cell lines with knockdown of ROCK1 was also evaluated. A proximity ligation assay was performed to determine the physical interactions between PK and ROCK1. RESULTS: Mice with a deficiency of ROCK1 in pancreatic ß-cells exhibited significantly increased blood glucose levels and reduced serum insulin without changes in body weight. Interestingly, ß-ROCK1-/- mice displayed a progressive impairment of glucose tolerance while maintaining insulin sensitivity mostly due to impaired GSIS. Consistently, GSIS markedly decreased in ROCK1-deficient islets and ROCK1 knockdown INS-1 cells. Concurrently, ROCK1 blockade led to a significant decrease in intracellular calcium and ATP levels and oxygen consumption rates in isolated islets and INS-1 cells. Treatment of ROCK1-deficient islets or ROCK1 knockdown ß-cells either with pyruvate or a PK activator rescued the impaired GSIS. Mechanistically, we observed that glucose stimulation in ß-cells greatly enhanced ROCK1 binding to PK. CONCLUSIONS: Our findings demonstrate that ß-cell ROCK1 is essential for glucose-stimulated insulin secretion and for glucose homeostasis and that ROCK1 acts as an upstream regulator of glycolytic pyruvate kinase signaling.


Assuntos
Secreção de Insulina , Insulina , Piruvato Quinase , Quinases Associadas a rho , Animais , Camundongos , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Piruvato Quinase/antagonistas & inibidores , Piruvato Quinase/metabolismo , Piruvatos
14.
Mol Metab ; 61: 101488, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35470093

RESUMO

OBJECTIVE: IL-6 is an important contributor to glucose and energy homeostasis through changes in whole-body glucose disposal, insulin sensitivity, food intake and energy expenditure. However, the relative contributions of peripheral versus central IL-6 signaling to these metabolic actions are presently unclear. A conditional mouse model with reduced brain IL-6Ra expression was used to explore how blunted central IL-6 signaling alters metabolic status in lean and obese mice. METHODS: Transgenic mice with reduced levels of central IL-6 receptor alpha (IL-6Ra) (IL-6Ra KD mice) and Nestin Cre controls (Cre+/- mice) were fed standard chow or high-fat diet for 20 weeks. Obese and lean mouse cohorts underwent metabolic phenotyping with various measures of energy and glucose homeostasis determined. Glucose-stimulated insulin secretion was assessed in vivo and ex vivo in both mouse groups. RESULTS: IL-6Ra KD mice exhibited altered body fat mass, liver steatosis, plasma insulin, IL-6 and NEFA levels versus Cre+/- mice in a diet-dependent manner. IL-6Ra KD mice had increased food intake, higher RER, decreased energy expenditure with diminished cold tolerance compared to Cre+/- controls. Standard chow-fed IL-6Ra KD mice displayed reduced plasma insulin and glucose-stimulated insulin secretion with impaired glucose disposal and unchanged insulin sensitivity. Isolated pancreatic islets from standard chow-fed IL-6Ra KD mice showed comparable morphology and glucose-stimulated insulin secretion to Cre+/- controls. The diminished in vivo insulin secretion exhibited by IL-6Ra KD mice was recovered by blockade of autonomic ganglia. CONCLUSIONS: This study shows that central IL-6Ra signaling contributes to glucose and energy control mechanisms by regulating food intake, energy expenditure, fuel flexibility and insulin secretion. A plausible mechanism linking central IL-6Ra signaling and pancreatic insulin secretion is through the modulation of autonomic output activity. Thus, brain IL-6 signaling may contribute to the central adaptive mechanisms engaged in response to metabolic stress.


Assuntos
Glucose , Resistência à Insulina , Receptores de Interleucina-6 , Animais , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Interleucina-6/metabolismo , Camundongos , Receptores de Interleucina-6/deficiência , Receptores de Interleucina-6/metabolismo
15.
Diabetes ; 71(3): 367-375, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35196393

RESUMO

Secretion of insulin from pancreatic ß-cells is complex, but physiological glucose-dependent secretion is dominated by electrical activity, in turn controlled by ATP-sensitive potassium (KATP) channel activity. Accordingly, loss-of-function mutations of the KATP channel Kir6.2 (KCNJ11) or SUR1 (ABCC8) subunit increase electrical excitability and secretion, resulting in congenital hyperinsulinism (CHI), whereas gain-of-function mutations cause underexcitability and undersecretion, resulting in neonatal diabetes mellitus (NDM). Thus, diazoxide, which activates KATP channels, and sulfonylureas, which inhibit KATP channels, have dramatically improved therapies for CHI and NDM, respectively. However, key findings do not fit within this simple paradigm: mice with complete absence of ß-cell KATP activity are not hyperinsulinemic; instead, they are paradoxically glucose intolerant and prone to diabetes, as are older human CHI patients. Critically, despite these advances, there has been little insight into any role of KATP channel activity changes in the development of type 2 diabetes (T2D). Intriguingly, the CHI progression from hypersecretion to undersecretion actually mirrors the classical response to insulin resistance in the progression of T2D. In seeking to explain the progression of CHI, multiple lines of evidence lead us to propose that underlying mechanisms are also similar and that development of T2D may involve loss of KATP activity.


Assuntos
Hiperinsulinismo Congênito/genética , Hiperinsulinismo Congênito/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Canais KATP/fisiologia , Animais , Glicemia , Cálcio/farmacologia , Humanos , Resistência à Insulina , Secreção de Insulina/genética , Secreção de Insulina/fisiologia , Canais KATP/genética , Camundongos , Camundongos Knockout , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Receptores de Sulfonilureias/genética , Receptores de Sulfonilureias/fisiologia
16.
Cell Rep ; 38(1): 110179, 2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34986353

RESUMO

G protein-coupled receptors (GPCRs) in intestinal enteroendocrine cells (EECs) respond to nutritional, neural, and microbial cues and modulate the release of gut hormones. Here we show that Gpr17, an orphan GPCR, is co-expressed in glucagon-like peptide-1 (GLP-1)-expressing EECs in human and rodent intestinal epithelium. Acute genetic ablation of Gpr17 in intestinal epithelium improves glucose tolerance and glucose-stimulated insulin secretion (GSIS). Importantly, inducible knockout (iKO) mice and Gpr17 null intestinal organoids respond to glucose or lipid ingestion with increased secretion of GLP-1, but not the other incretin glucose-dependent insulinotropic polypeptide (GIP). In an in vitro EEC model, overexpression or agonism of Gpr17 reduces voltage-gated calcium currents and decreases cyclic AMP (cAMP) production, and these are two critical factors regulating GLP-1 secretion. Together, our work shows that intestinal Gpr17 signaling functions as an inhibitory pathway for GLP-1 secretion in EECs, suggesting intestinal GPR17 is a potential target for diabetes and obesity intervention.


Assuntos
Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/metabolismo , Mucosa Intestinal/metabolismo , Proteínas do Tecido Nervoso/genética , Receptores Acoplados a Proteínas G/genética , Animais , Glicemia/análise , Cálcio/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Diabetes Mellitus/patologia , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Teste de Tolerância a Glucose , Células HEK293 , Células HeLa , Humanos , Incretinas/metabolismo , Insulina/metabolismo , Secreção de Insulina/fisiologia , Mucosa Intestinal/citologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/patologia , Receptores dos Hormônios Gastrointestinais/metabolismo
17.
Endocrinology ; 163(3)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35086144

RESUMO

During development of type 2 diabetes (T2D), excessive nutritional load is thought to expose pancreatic islets to toxic effects of lipids and reduce ß-cell function and mass. However, lipids also play a positive role in cellular metabolism and function. Thus, proper trafficking of lipids is critical for ß cells to maximize the beneficial effects of these molecules while preventing their toxic effects. Lipid droplets (LDs) are organelles that play an important role in the storage and trafficking of lipids. In this review, we summarize the discovery of LDs in pancreatic ß cells, LD lifecycle, and the effect of LD catabolism on ß-cell insulin secretion. We discuss factors affecting LD formation such as age, cell type, species, and nutrient availability. We then outline published studies targeting critical LD regulators, primarily in rat and human ß-cell models, to understand the molecular effect of LD formation and degradation on ß-cell function and health. Furthermore, based on the abnormal LD accumulation observed in human T2D islets, we discuss the possible role of LDs during the development of ß-cell failure in T2D. Current knowledge indicates that proper formation and clearance of LDs are critical to normal insulin secretion, endoplasmic reticulum homeostasis, and mitochondrial integrity in ß cells. However, it remains unclear whether LDs positively or negatively affect human ß-cell demise in T2D. Thus, we discuss possible research directions to address the knowledge gap regarding the role of LDs in ß-cell failure.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Células Secretoras de Insulina/fisiologia , Células Secretoras de Insulina/ultraestrutura , Gotículas Lipídicas/fisiologia , Animais , Morte Celular , Senescência Celular , Diabetes Mellitus Tipo 2/patologia , Estresse do Retículo Endoplasmático , Humanos , Secreção de Insulina/fisiologia , Perilipina-2/fisiologia , Perilipina-5/fisiologia , Ratos
18.
Artigo em Inglês | MEDLINE | ID: mdl-34303334

RESUMO

BACKGROUND: Insulin resistance is a well-known predictor and risk factor for type 2 diabetes mellitus (T2DM). Higher hematocrit induced by higher insulin resistance affects blood rheology. OBJECTIVE: This study intended to reveal the association between indices of insulin resistance and hemorheological parameters during a 75 g oral glucose tolerance test (75-g OGTT). METHODS: A total of 575 healthy young Japanese participants took 75-g OGTT. We then analyzed the association between insulin resistance indices and hematological parameters. RESULTS: The Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) was significantly correlated with hematocrit (Ht), hemoglobin (Hb), red blood cell (RBC), white blood cell (WBC), platelet count, lipid parameters and body mass index (BMI). The Matsuda index was negatively correlated with RBC count, WBC count, platelet count, total cholesterol (TC), low-density lipoprotein- cholesterol (LDL-C), triglyceride (TG), and positively correlated with high-density lipoprotein- cholesterol (HDL-C). The disposition index was negatively correlated with Hb, RBC count, LDL-C and BMI, and positively correlated with HDL-C. The Homeostasis Model Assessment of beta cell (HOMA-ß) was positively correlated with WBC count, platelet count, TC, LDL-C and TG. The insulinogenic index was positively correlated with WBC count, platelet count and TC. Multiple regression analysis revealed that HOMA-IR was independently associated with TG, and the Matsuda index was independently associated with TG, WBC count, and platelet count. The insulinogenic index was independently associated with WBC count. CONCLUSION: Cardinal rheological parameters reflected insulin resistance and release even in young healthy Japanese individuals within the physiological range of glycemic control.


Assuntos
Circulação Sanguínea/fisiologia , Glicemia/metabolismo , Resistência à Insulina/fisiologia , Adulto , Contagem de Eritrócitos , Feminino , Teste de Tolerância a Glucose , Voluntários Saudáveis , Hematócrito , Humanos , Secreção de Insulina/fisiologia , Japão , Contagem de Leucócitos , Masculino , Contagem de Plaquetas , Reologia , Adulto Jovem
19.
Mol Metab ; 55: 101414, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34871777

RESUMO

OBJECTIVE: Age is a risk factor for type 2 diabetes (T2D). We aimed to elucidate whether ß-cell glucose metabolism is altered with aging and contributes to T2D. METHODS: We used senescence-accelerated mice (SAM), C57BL/6J (B6) mice, and ob/ob mice as aging models. As a diabetes model, we used db/db mice. The glucose responsiveness of insulin secretion and the [U-13C]-glucose metabolic flux were examined in isolated islets. We analyzed the expression of ß-cell-specific genes in isolated islets and pancreatic sections as molecular signatures of ß-cell identity. ß cells defective in the malate-aspartate (MA) shuttle were previously generated from MIN6-K8 cells by the knockout of Got1, a component of the shuttle. We analyzed Got1 KO ß cells as a model of increased glycolysis. RESULTS: We identified hyperresponsiveness to glucose and compromised cellular identity as dysfunctional phenotypes shared in common between aged and diabetic mouse ß cells. We also observed a metabolic commonality between aged and diabetic ß cells: hyperactive glycolysis through the increased expression of nicotinamide mononucleotide adenylyl transferase 2 (Nmnat2), a cytosolic nicotinamide adenine dinucleotide (NAD)-synthesizing enzyme. Got1 KO ß cells showed increased glycolysis, ß-cell dysfunction, and impaired cellular identity, phenocopying aging and diabetes. Using Got1 KO ß cells, we show that attenuation of glycolysis or Nmnat2 activity can restore ß-cell function and identity. CONCLUSIONS: Our study demonstrates that hyperactive glycolysis is a metabolic signature of aged and diabetic ß cells, which may underlie age-related ß-cell dysfunction and loss of cellular identity. We suggest Nmnat2 suppression as an approach to counteract age-related T2D.


Assuntos
Envelhecimento/fisiologia , Glicólise/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Linhagem Celular , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Controle Glicêmico/métodos , Insulina/metabolismo , Secreção de Insulina/fisiologia , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Obesidade/metabolismo
20.
Alcohol Clin Exp Res ; 46(1): 87-99, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34773280

RESUMO

BACKGROUND: Alcohol, insulin resistance (IR), and hepatitis C (HCV) are all significant contributors to adverse outcomes of chronic liver disease. Latinos are disproportionately affected by these risk factors. We investigated the relationship between alcohol use and insulin action in a prospective cohort of Latino individuals with and without HCV. METHODS: One hundred fifty-three nondiabetic Latino individuals (60 HCV+, 93 HCV-) underwent clinical evaluation and metabolic testing; 56 had repeat testing over a median follow-up of 1.5 years. Peripheral IR and hepatic IR were measured via steady-state plasma glucose (SSPG) and endogenous glucose production during a two-step, 240-min insulin suppression test. Insulin secretion (IS) was measured using the graded glucose infusion test. Alcohol use was categorized as none, moderate (≤1 drink/day for women and ≤2 drinks/day for men), and heavy (>moderate). Multivariable models including HCV status assessed associations of alcohol use with baseline SSPG, hepatic IR and IS, and changes in these parameters over time. RESULTS: Overall, the median age was 44 years, 63.4% were male, 66.7% overweight/ obese, and 31.9% had heavy lifetime alcohol use while 60.4% had moderate lifetime alcohol use. SSPG and IS were similar by levels of alcohol use at baseline and alcohol use was not statistically significantly associated with change in these measures over time. However, lifetime daily heavy alcohol use (vs. not heavy, coef 2.4 µU-mg/kg-min-ml, p = 0.04) and HCV status (coef 4.4 µU-mg/kg-min-ml, p = 0.0003) were independently associated with higher baseline hepatic IR, and current heavy alcohol use was associated with greater change in hepatic IR in follow-up (coef 5.8 µU-mg/kg-min-ml, p = 0.03). CONCLUSIONS: In this cohort of Latino individuals, lifetime and current heavy alcohol use influenced hepatic IR and its change over time. Strategies to decrease rates of heavy alcohol use or increase abstinence along with lifestyle modification and anti-HCV therapy to reduce metabolic risk are critical to prevent adverse liver and metabolic outcomes in Latino individuals.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Hepatite C/complicações , Hispânico ou Latino/estatística & dados numéricos , Resistência à Insulina/etnologia , Insulina/farmacologia , Adulto , Estudos de Coortes , Citocromo P-450 CYP2E1/genética , Etanol/administração & dosagem , Feminino , Genótipo , Hepatite C/fisiopatologia , Humanos , Secreção de Insulina/fisiologia , Fígado/efeitos dos fármacos , Fígado/fisiopatologia , Hepatopatias/epidemiologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...