Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 18(3): 283-292, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28092375

RESUMO

The deleterious effect of chronic activation of the IL-1ß system on type 2 diabetes and other metabolic diseases is well documented. However, a possible physiological role for IL-1ß in glucose metabolism has remained unexplored. Here we found that feeding induced a physiological increase in the number of peritoneal macrophages that secreted IL-1ß, in a glucose-dependent manner. Subsequently, IL-1ß contributed to the postprandial stimulation of insulin secretion. Accordingly, lack of endogenous IL-1ß signaling in mice during refeeding and obesity diminished the concentration of insulin in plasma. IL-1ß and insulin increased the uptake of glucose into macrophages, and insulin reinforced a pro-inflammatory pattern via the insulin receptor, glucose metabolism, production of reactive oxygen species, and secretion of IL-1ß mediated by the NLRP3 inflammasome. Postprandial inflammation might be limited by normalization of glycemia, since it was prevented by inhibition of the sodium-glucose cotransporter SGLT2. Our findings identify a physiological role for IL-1ß and insulin in the regulation of both metabolism and immunity.


Assuntos
Diabetes Mellitus Tipo 2/imunologia , Inflamação/imunologia , Células Secretoras de Insulina/fisiologia , Interleucina-1beta/metabolismo , Macrófagos/fisiologia , Animais , Células Cultivadas , Glucose/metabolismo , Humanos , Inflamassomos/metabolismo , Insulina/metabolismo , Interleucina-1beta/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Período Pós-Prandial , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Transportador 2 de Glucose-Sódio/metabolismo
2.
Immunity ; 47(5): 928-942.e7, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29166590

RESUMO

Pancreatic-islet inflammation contributes to the failure of ß cell insulin secretion during obesity and type 2 diabetes. However, little is known about the nature and function of resident immune cells in this context or in homeostasis. Here we show that interleukin (IL)-33 was produced by islet mesenchymal cells and enhanced by a diabetes milieu (glucose, IL-1ß, and palmitate). IL-33 promoted ß cell function through islet-resident group 2 innate lymphoid cells (ILC2s) that elicited retinoic acid (RA)-producing capacities in macrophages and dendritic cells via the secretion of IL-13 and colony-stimulating factor 2. In turn, local RA signaled to the ß cells to increase insulin secretion. This IL-33-ILC2 axis was activated after acute ß cell stress but was defective during chronic obesity. Accordingly, IL-33 injections rescued islet function in obese mice. Our findings provide evidence that an immunometabolic crosstalk between islet-derived IL-33, ILC2s, and myeloid cells fosters insulin secretion.


Assuntos
Insulina/metabolismo , Interleucina-33/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Células Mieloides/metabolismo , Tretinoína/metabolismo , Animais , Humanos , Inflamação/imunologia , Secreção de Insulina , Interleucina-33/biossíntese , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/patologia , Linfócitos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vitamina A/fisiologia
3.
Diabetologia ; 66(11): 2062-2074, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37650924

RESUMO

AIMS/HYPOTHESIS: Glutamate-induced cytotoxicity (excitotoxicity) has been detected in pancreatic beta cells. The cystine/glutamate antiporter System xc- exports glutamate to the extracellular space and is therefore implicated as driving excitotoxicity. As of yet, it has not been investigated whether System xc- contributes to pancreatic islet function. METHODS: This study describes the implications of deficiency of System xc- on glucose metabolism in both constitutive and myeloid cell-specific knockout mice using metabolic tests and diet-induced obesity. Pancreatic islets were isolated and analysed for beta cell function, glutathione levels and ER stress. RESULTS: Constitutive System xc- deficiency led to an approximately threefold decrease in glutathione levels in the pancreatic islets as well as cystine shortage characterised by upregulation of Chac1. This shortage further manifested as downregulation of beta cell identity genes and a tonic increase in endoplasmic reticulum stress markers, which resulted in diminished insulin secretion both in vitro and in vivo. Myeloid-specific deletion did not have a significant impact on metabolism or islet function. CONCLUSIONS/INTERPRETATION: These findings suggest that System xc- is required for glutathione maintenance and insulin production in beta cells and that the system is dispensable for islet macrophage function.


Assuntos
Cistina , Ácido Glutâmico , Camundongos , Animais , Cistina/metabolismo , Ácido Glutâmico/metabolismo , Secreção de Insulina , Antiporters/metabolismo , Camundongos Knockout , Glutationa/metabolismo
4.
Diabetologia ; 66(12): 2292-2306, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37792013

RESUMO

AIMS/HYPOTHESIS: Colony stimulating factor 1 (CSF1) promotes the proliferation, differentiation and survival of macrophages, which have been implicated in both beneficial and detrimental effects on glucose metabolism. However, the physiological role of CSF1 signalling in glucose homeostasis and the potential therapeutic implications of modulating this pathway are not known. We aimed to study the composition of tissue macrophages (and other immune cells) following CSF1 receptor (CSF1R) inhibition and elucidate the metabolic consequences of CSF1R inhibition. METHODS: We assessed immune cell populations in various organs by flow cytometry, and tissue-specific metabolic effects by hyperinsulinaemic-euglycaemic clamps and insulin secretion assays in mice fed a chow diet containing PLX5622 (a CSF1R inhibitor) or a control diet. RESULTS: CSF1R inhibition depleted macrophages in multiple tissues while simultaneously increasing eosinophils and group 2 innate lymphoid cells. These immunological changes were consistent across different organs and were sex independent and reversible after cessation of the PLX5622. CSF1R inhibition improved hepatic insulin sensitivity but concomitantly impaired insulin secretion. In healthy islets, we found a high frequency of IL-1ß+ islet macrophages. Their depletion by CSF1R inhibition led to downregulation of macrophage-related pathways and mediators of cytokine activity, including Nlrp3, suggesting IL-1ß as a candidate insulin secretagogue. Partial restoration of physiological insulin secretion was achieved by injecting recombinant IL-1ß prior to glucose stimulation in mice lacking macrophages. CONCLUSIONS/INTERPRETATION: Macrophages and macrophage-derived factors, such as IL-1ß, play an important role in physiological insulin secretion. A better understanding of the tissue-specific effects of CSF1R inhibition on immune cells and glucose homeostasis is crucial for the development of targeted immune-modulatory treatments in metabolic disease. DATA AVAILABILITY: The RNA-Seq dataset is available in the Gene Expression Omnibus (GEO) under the accession number GSE189434 ( http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE189434 ).


Assuntos
Imunidade Inata , Linfócitos , Camundongos , Animais , Macrófagos/metabolismo , Glucose/metabolismo
5.
Diabetologia ; 63(2): 362-373, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31796986

RESUMO

AIMS/HYPOTHESIS: IL-6 is a cytokine with various effects on metabolism. In mice, IL-6 improved beta cell function and glucose homeostasis via upregulation of glucagon-like peptide 1 (GLP-1), and IL-6 release from muscle during exercise potentiated this beneficial increase in GLP-1. This study aimed to identify whether exercise-induced IL-6 has a similar effect in humans. METHODS: In a multicentre, double-blind clinical trial, we randomly assigned patients with type 2 diabetes or obesity to intravenous tocilizumab (an IL-6 receptor antagonist) 8 mg/kg every 4 weeks, oral sitagliptin (a dipeptidyl peptidase-4 inhibitor) 100 mg daily or double placebos (a placebo saline infusion every 4 weeks and a placebo pill once daily) during a 12 week training intervention. The primary endpoints were the difference in change of active GLP-1 response to an acute exercise bout and change in the AUC for the concentration-time curve of active GLP-1 during mixed meal tolerance tests at baseline and after the training intervention. RESULTS: Nineteen patients were allocated to tocilizumab, 17 to sitagliptin and 16 to placebos. During the acute exercise bout active GLP-1 levels were 26% lower with tocilizumab (multiplicative effect: 0.74 [95% CI 0.56, 0.98], p = 0.034) and 53% higher with sitagliptin (1.53 [1.15, 2.03], p = 0.004) compared with placebo. After the 12 week training intervention, the active GLP-1 AUC with sitagliptin was about twofold that with placebo (2.03 [1.56, 2.62]; p < 0.001), while GLP-1 AUC values showed a small non-significant decrease of 13% at 4 weeks after the last tocilizumab infusion (0.87 [0.67, 1.12]; p = 0.261). CONCLUSIONS/INTERPRETATION: IL-6 is implicated in the regulation of GLP-1 in humans. IL-6 receptor blockade lowered active GLP-1 levels in response to a meal and an acute exercise bout in a reversible manner, without lasting effects beyond IL-6 receptor blockade. TRIAL REGISTRATION: Clinicaltrials.gov NCT01073826. FUNDING: Danish National Research Foundation. Danish Council for Independent Research. Novo Nordisk Foundation. Danish Centre for Strategic Research in Type 2 Diabetes. European Foundation for the Study of Diabetes. Swiss National Research Foundation.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Exercício Físico/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Anticorpos Monoclonais Humanizados/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Método Duplo-Cego , Feminino , Humanos , Interleucina-6/metabolismo , Masculino , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Receptores de Interleucina-6/metabolismo , Fosfato de Sitagliptina/uso terapêutico
6.
Appetite ; 155: 104792, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32707265

RESUMO

The initial release of insulin in response to food stimuli acting on receptors in the head and oropharynx is called the cephalic phase of insulin secretion. Insulin has been shown to act centrally to regulate food intake and glucose metabolism and the cephalic phase of insulin secretion may contribute to these functions. Though well documented in laboratory animals, the existence of cephalic phase insulin release in humans has recently come into question. We therefore performed a systematic review and meta-analysis of studies of cephalic phase insulin release in humans. Efficacy outcomes included any change in circulating insulin levels in healthy human volunteers post any food stimulus as compared to baseline or control in a time period of no longer than 10 min. Primary outcome: The overall pooled effect size estimate for cephalic phase insulin release was 0.47 [0.36, 0.58] p-value <0.0001. Secondary outcomes: A random effects meta-analysis with an added moderator for type of stimulus presentation (one, two, four or five sensory qualities) and type of stimulus offered (liquid, solid formulation) also significantly influenced results p = 0.0116 and p = 0.0024 respectively, while sex had no significant effect. Sensitivity Analysis: More restrictive analyses only including studies that used non-ingestive stimuli (p = 0.0001), or studies that reported insulin values within 5 min post stimulus presentation (p < 0.0001) still showed significant positive overall effect size estimates. In summary, our analysis shows that there is evidence for the presence of cephalic phase insulin secretion in humans. Secondary analyses suggest that the type and presentation of stimulus may significantly influence cephalic phase insulin secretion, while sex had no significant effect on cephalic phase insulin secretion.


Assuntos
Glicemia , Insulina , Animais , Alimentos , Humanos , Insulina/metabolismo , Secreção de Insulina
7.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33137873

RESUMO

Selective inhibition of histone deacetylase 3 (HDAC3) prevents glucolipotoxicity-induced ß-cell dysfunction and apoptosis by alleviation of proapoptotic endoplasmic reticulum (ER) stress-signaling, but the precise molecular mechanisms of alleviation are unexplored. By unbiased microarray analysis of the ß-cell gene expression profile of insulin-producing cells exposed to glucolipotoxicity in the presence or absence of a selective HDAC3 inhibitor, we identified Enhancer of zeste homolog 2 (EZH2) as the sole target candidate. ß-Cells were protected against glucolipotoxicity-induced ER stress and apoptosis by EZH2 attenuation. Small molecule inhibitors of EZH2 histone methyltransferase activity rescued human islets from glucolipotoxicity-induced apoptosis. Moreover, EZH2 knockdown cells were protected against glucolipotoxicity-induced downregulation of the protective non-canonical Nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB) pathway. We conclude that EZH2 deficiency protects from glucolipotoxicity-induced ER stress, apoptosis and downregulation of the non-canonical NFκB pathway, but not from insulin secretory dysfunction. The mechanism likely involves transcriptional regulation via EZH2 functioning as a methyltransferase and/or as a methylation-dependent transcription factor.


Assuntos
Apoptose , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Glucose/efeitos adversos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Lipídeos/efeitos adversos , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Edulcorantes/efeitos adversos
8.
Gastroenterology ; 151(1): 165-79, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26971825

RESUMO

BACKGROUND & AIMS: Glucose-dependent insulinotropic peptide (GIP) induces production of interleukin 6 (IL6) by adipocytes. IL6 increases production of glucagon-like peptide (GLP)-1 by L cells and α cells, leading to secretion of insulin from ß cells. We investigated whether GIP regulates GLP1 and glycemia via IL6. METHODS: We obtained samples of human pancreatic islets and isolated islets from mice; human α cells and ß cells were sorted by flow cytometry and incubated with GIP. Islets were analyzed by quantitative polymerase chain reaction and immunohistochemistry. BKS.Cg-Dock7m+/+ Leprdb/J db/db mice (diabetic mice) and db/+ mice, as well as C57BL/6J IL6-knockout mice (IL6-KO) and C57BL/6J mice with the full-length Il6 gene (controls), were fed a chow or a high-fat diet; some mice were given injections of recombinant GIP, IL6, GLP, a neutralizing antibody against IL6 (anti-IL6), lipopolysaccharide, and/or IL1B. Mice were given a glucose challenge and blood samples were collected and analyzed. RESULTS: Incubation of mouse and human pancreatic α cells with GIP induced their production of IL6, leading to production of GLP1 and insulin secretion from pancreatic islets. This did not occur in islets from IL6-KO mice or in islets incubated with anti-IL6. Incubation of islets with IL1B resulted in IL6 production but directly reduced GLP1 production. Incubation of mouse islets with the sodium glucose transporter 2 inhibitor dapagliflozin induced production of GLP1 and IL6. Injection of control mice with GIP increased plasma levels of GLP1, insulin, and glucose tolerance; these effects were amplified in mice given lipopolysaccharide but reduced in IL6-KO mice or in mice given anti-IL6. Islets from diabetic mice had increased levels of IL1B and IL6, compared with db/+ mice, but injection of GIP did not lead to production of GLP1 or reduce glycemia. CONCLUSIONS: In studies of pancreatic islets from human beings and mice, we found that GIP induces production of IL6 by α cells, leading to islet production of GLP1 and insulin. This process is regulated by inflammation, via IL1B, and by sodium glucose transporter 2. In diabetic mice, increased islet levels of IL6 and IL1B might increase or reduce the production of GLP1 and affect glycemia.


Assuntos
Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-6/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/etiologia , Diabetes Mellitus Experimental/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Camundongos , Camundongos Endogâmicos C57BL
9.
Am J Physiol Regul Integr Comp Physiol ; 306(11): R861-7, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24694381

RESUMO

Contracting muscle releases interleukin-6 (IL-6) enabling the metabolic switch from carbohydrate to fat utilization. Similarly, metabolism is switched during transition from fed to fasting state. Herein, we examined a putative role for IL-6 in the metabolic adaptation to normal fasting. In lean C57BL/6J mice, 6 h of food withdrawal increased gene transcription levels of IL-6 in skeletal muscle but not in white adipose tissue. Concomitantly, circulating IL-6 and free fatty acid (FFA) levels were significantly increased, whereas respiratory quotient (RQ) was reduced in 6-h fasted mice. In white adipose tissue, phosphorylation of hormone-sensitive lipase (HSL) was increased on fasting, indicating increased lipolysis. Intriguingly, fasting-induced increase in circulating IL-6 levels and parallel rise in FFA concentration were absent in obese and glucose-intolerant mice. A causative role for IL-6 in the physiological adaptation to fasting was further supported by the fact that fasting-induced increase in circulating FFA levels was significantly blunted in lean IL-6 knockout (KO) and lean C57BL/6J mice treated with neutralizing IL-6 antibody. Consistently, phosphorylation of HSL was significantly reduced in adipose tissue of IL-6-depleted mice. Hence, our findings suggest a novel role for IL-6 in energy supply during early fasting.


Assuntos
Jejum/psicologia , Ácidos Graxos não Esterificados/metabolismo , Interleucina-6/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Metabolismo Energético/fisiologia , Interleucina-6/deficiência , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais
10.
Br J Clin Pharmacol ; 76(2): 263-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22506644

RESUMO

Metabolic diseases are associated with activation of the innate immune system in various tissues and characterized by elevated inflammatory factors and the presence of immune cells. Type 2 diabetes develops when islet beta cells are deficient in producing sufficient insulin to overcome peripheral insulin resistance. Intra-islet IL-1ß activity diminishes beta cell function and survival and governs islet inflammation. Targeting the IL-1 system with the IL-1 receptor antagonist IL1Ra improved insulin secretion, glycaemia and reduced systemic inflammation in a proof of concept study with patients with type 2 diabetes. Currently, long lasting and specific IL-1ß blocking antibodies are being evaluated in clinical trials and this may lead to a novel cytokine-based treatment for type 2 diabetes.


Assuntos
Anticorpos Bloqueadores/uso terapêutico , Diabetes Mellitus Tipo 2/terapia , Interleucina-1/imunologia , Ensaios Clínicos como Assunto , Diabetes Mellitus Tipo 2/imunologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/imunologia
11.
STAR Protoc ; 4(4): 102664, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37889759

RESUMO

Diabetes mellitus is a disease of the hormone-secreting endocrine pancreas. However, increasing evidence suggests that the exocrine pancreas is also involved in the pathogenesis of diabetes. In this protocol, we describe how to harvest both isolated islets and exocrine tissue from one mouse pancreas, followed by a detailed explanation of how to isolate and analyze immune cells using full-spectrum flow cytometry.


Assuntos
Ilhotas Pancreáticas , Pâncreas Exócrino , Camundongos , Animais , Citometria de Fluxo
12.
PLoS One ; 18(10): e0291950, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37792774

RESUMO

System xc-, encoded by Slc7a11, is an antiporter responsible for exporting glutamate while importing cystine, which is essential for protein synthesis and the formation of thiol peptides, such as glutathione. Glutathione acts as a co-factor for enzymes responsible for scavenging reactive oxygen species. Upon exposure to bacterial products, macrophages exhibit a rapid upregulation of system xc-. This study investigates the impact of Slc7a11 deficiency on the functionality of peritoneal and bone marrow-derived macrophages. Our findings reveal that the absence of Slc7a11 results in significantly reduced glutathione levels, compromised mitochondrial flexibility, and hindered cytokine production in bone marrow-derived macrophages. Conversely, system xc- has a lesser impact on peritoneal macrophages in vivo. These results indicate that system xc- is essential for maintaining glutathione levels, mitochondrial functionality, and cytokine production, with a heightened importance under atmospheric oxygen tension.


Assuntos
Cistina , Ácido Glutâmico , Ácido Glutâmico/metabolismo , Cistina/metabolismo , Antiporters , Macrófagos Peritoneais/metabolismo , Glutationa/metabolismo , Citocinas/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo
13.
Cell Metab ; 34(7): 991-1003.e6, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35750050

RESUMO

The initial cephalic phase of insulin secretion is mediated through the vagus nerve and is not due to glycemic stimulation of pancreatic ß cells. Recently, IL-1ß was shown to stimulate postprandial insulin secretion. Here, we describe that this incretin-like effect of IL-1ß involves neuronal transmission. Furthermore, we found that cephalic phase insulin release was mediated by IL-1ß originating from microglia. Moreover, IL-1ß activated the vagus nerve to induce insulin secretion and regulated the activity of the hypothalamus in response to cephalic stimulation. Notably, cephalic phase insulin release was impaired in obesity, in both mice and humans, and in mice, this was due to dysregulated IL-1ß signaling. Our findings attribute a regulatory role to IL-1ß in the integration of nutrient-derived sensory information, subsequent neuronally mediated insulin secretion, and the dysregulation of autonomic cephalic phase responses in obesity.


Assuntos
Células Secretoras de Insulina , Insulina , Interleucina-1beta , Animais , Glicemia/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Obesidade/metabolismo
14.
Nat Commun ; 13(1): 4761, 2022 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-35963866

RESUMO

Defective insulin processing is associated with obesity and diabetes. Prohormone convertase 1/3 (PC1/3) is an endopeptidase required for the processing of neurotransmitters and hormones. PC1/3 deficiency and genome-wide association studies relate PC1/3 with early onset obesity. Here, we find that deletion of PC1/3 in obesity-related neuronal cells expressing proopiomelanocortin mildly and transiently change body weight and fail to produce a phenotype when targeted to Agouti-related peptide- or nestin-expressing tissues. In contrast, pancreatic ß cell-specific PC1/3 ablation induces hyperphagia with consecutive obesity despite uncontrolled diabetes with glucosuria. Obesity develops not due to impaired pro-islet amyloid polypeptide processing but due to impaired insulin maturation. Proinsulin crosses the blood-brain-barrier but does not induce central satiety. Accordingly, insulin therapy prevents hyperphagia. Further, islet PC1/3 expression levels negatively correlate with body mass index in humans. In this work, we show that impaired PC1/3-mediated proinsulin processing, as observed in human prediabetes, promotes hyperphagic obesity.


Assuntos
Diabetes Mellitus , Proinsulina , Estudo de Associação Genômica Ampla , Humanos , Hiperfagia/genética , Insulina/metabolismo , Obesidade/complicações , Obesidade/genética , Obesidade/metabolismo , Proinsulina/genética , Proinsulina/metabolismo , Pró-Proteína Convertase 1/genética
15.
iScience ; 24(11): 103250, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34746709

RESUMO

Aging is the prime risk factor for the development of type 2 diabetes. We investigated the role of the interleukin-1 (IL-1) system on insulin secretion in aged mice. During aging, expression of the protective IL-1 receptor antagonist decreased in islets, whereas IL-1beta gene expression increased specifically in the CD45 + islet immune cell fraction. One-year-old mice with a whole-body knockout of IL-1beta had higher insulin secretion in vivo and in isolated islets, along with enhanced proliferation marker Ki67 and elevated size and number of islets. Myeloid cell-specific IL-1beta knockout preserved glucose-stimulated insulin secretion during aging, whereas it declined in control mice. Isolated islets from aged myeloIL-1beta ko mice secreted more insulin along with increased expression of Ins2, Kir6.2, and of the cell-cycle gene E2f1. IL-1beta treatment of isolated islets reduced E2f1, Ins2, and Kir6.2 expression in beta cells. We conclude that IL-1beta contributes the age-associated decline of beta cell function.

16.
Physiology (Bethesda) ; 24: 325-31, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19996363

RESUMO

Onset of Type 2 diabetes occurs when the pancreatic beta-cell fails to adapt to the increased insulin demand caused by insulin resistance. Morphological and therapeutic intervention studies have uncovered an inflammatory process in islets of patients with Type 2 diabetes characterized by the presence of cytokines, immune cells, beta-cell apoptosis, amyloid deposits, and fibrosis. This insulitis is due to a pathological activation of the innate immune system by metabolic stress and governed by IL-1 signaling. We propose that this insulitis contributes to the decrease in beta-cell mass and the impaired insulin secretion observed in patients with Type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Insulina/patologia , Animais , Citocinas/imunologia , Diabetes Mellitus Tipo 2/imunologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Resistência à Insulina/imunologia , Células Secretoras de Insulina/imunologia
17.
Cell Rep ; 30(5): 1627-1643.e7, 2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32023474

RESUMO

The innate immune system safeguards the organism from both pathogenic and environmental stressors. Also, physiologic levels of nutrients affect organismal and intra-cellular metabolism and challenge the immune system. In the long term, over-nutrition leads to low-grade systemic inflammation. Here, we investigate tissue-resident components of the innate immune system (macrophages) and their response to short- and long-term nutritional challenges. We analyze the transcriptomes of six tissue-resident macrophage populations upon acute feeding and identify adipose tissue macrophages and the IL-1 pathway as early sensors of metabolic changes. Furthermore, by comparing functional responses between macrophage subtypes, we propose a regulatory, anti-inflammatory role of heat shock proteins of the HSP70 family in response to long- and short-term metabolic challenges. Our data provide a resource for assessing the impact of nutrition and over-nutrition on the spectrum of macrophages across tissues with a potential for identification of systemic responses.


Assuntos
Macrófagos/metabolismo , Transcrição Gênica , Tecido Adiposo/citologia , Animais , Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica , Ácidos Graxos/metabolismo , Proteínas de Choque Térmico/metabolismo , Interleucina-1/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Ratos , Transdução de Sinais , Estreptozocina , Fatores de Tempo
18.
Cell Metab ; 31(4): 699-709.e5, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32197070

RESUMO

Postprandial hypoglycemia is a disabling complication of the treatment of obesity by gastric bypass surgery. So far, no therapy exists, and the underlying mechanisms remain unclear. Here, we hypothesized that glucose-induced IL-1ß leads to an exaggerated insulin response in this condition. Therefore, we conducted a placebo-controlled, randomized, double-blind, crossover study with the SGLT2-inhibitor empagliflozin and the IL-1 receptor antagonist anakinra (clinicaltrials.govNCT03200782; n = 12). Both drugs reduced postprandial insulin release and prevented hypoglycemia (symptomatic events requiring rescue glucose: placebo = 7/12, empagliflozin = 2/12, and anakinra = 2/12, pvallikelihood ratio test (LRT) = 0.013; nadir blood glucose for placebo = 2.4 mmol/L, 95% CI 2.18-2.62, empagliflozin = 2.69 mmol/L, 95% CI 2.31-3.08, and anakinra = 2.99 mmol/L, 95% CI 2.43-3.55, pvalLRT = 0.048). Moreover, analysis of monocytes ex vivo revealed a hyper-reactive inflammatory state that has features of an exaggerated response to a meal. Our study proposes a role for glucose-induced IL-1ß in postprandial hypoglycemia after gastric bypass surgery and suggests that SGLT2-inhibitors and IL-1 antagonism may improve this condition.


Assuntos
Compostos Benzidrílicos/farmacologia , Derivação Gástrica/efeitos adversos , Glucosídeos/farmacologia , Hipoglicemia/tratamento farmacológico , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/fisiologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Adulto , Estudos Cross-Over , Método Duplo-Cego , Feminino , Glucose/metabolismo , Humanos , Hipoglicemia/etiologia , Masculino , Pessoa de Meia-Idade , Período Pós-Prandial , Estudo de Prova de Conceito
19.
Sci Rep ; 10(1): 3035, 2020 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-32080229

RESUMO

Gestational diabetes mellitus (GDM) is one of the most common diseases associated with pregnancy, however, the underlying mechanisms remain unclear. Based on the well documented role of inflammation in type 2 diabetes, the aim was to investigate the role of inflammation in GDM. We established a mouse model for GDM on the basis of its two major risk factors, obesity and aging. In these GDM mice, we observed increased Interleukin-1ß (IL-1ß) expression in the uterus and the placenta along with elevated circulating IL-1ß concentrations compared to normoglycemic pregnant mice. Treatment with an anti-IL-1ß antibody improved glucose-tolerance of GDM mice without apparent deleterious effects for the fetus. Finally, IL-1ß antagonism showed a tendency for reduced plasma corticosterone concentrations, possibly explaining the metabolic improvement. We conclude that IL-1ß is a causal driver of impaired glucose tolerance in GDM.


Assuntos
Diabetes Gestacional/metabolismo , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Interleucina-1beta/antagonistas & inibidores , Animais , Diabetes Gestacional/sangue , Modelos Animais de Doenças , Feminino , Hormônios/sangue , Hiperglicemia/sangue , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Gravidez , Esteroides/sangue
20.
Semin Immunopathol ; 41(4): 501-513, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30989320

RESUMO

Metabolic diseases including type 2 diabetes are associated with meta-inflammation. ß-Cell failure is a major component of the pathogenesis of type 2 diabetes. It is now well established that increased numbers of innate immune cells, cytokines, and chemokines have detrimental effects on islets in these chronic conditions. Recently, evidence emerged which points to initially adaptive and restorative functions of inflammatory factors and immune cells in metabolism. In the following review, we provide an overview on the features of islet inflammation in diabetes and models of prediabetes. We separately emphasize what is known on islet inflammation in humans and focus on in vivo animal models and how they are used to elucidate mechanistic aspects of islet inflammation. Further, we discuss the recently emerging physiologic signaling role of cytokines during adaptation and normal function of islet cells.


Assuntos
Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Diabetes Mellitus Tipo 2/patologia , Humanos , Inflamação/metabolismo , Inflamação/patologia , Células Secretoras de Insulina/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA