Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Diabetes Metab Res Rev ; 40(5): e3829, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38850100

ABSTRACT

AIMS: Pancreatic polypeptide (PP) is elevated in people with vascular risk factors such as type 2 diabetes or increased visceral fat. We investigated potential relationships between PP and microvascular and macrovascular complications of diabetes. MATERIALS AND METHODS: Animal study: Subcutaneous PP infusion for 4 weeks in high fat diet mouse model. Retinal mRNA submitted for Ingenuity Pathway Analysis. Human study: fasting PP measured in 1478 participants and vascular complications recorded over median 5.5 (IQR 4.9-5.8) years follow-up. RESULTS: Animal study: The retinal transcriptional response to PP was indicative of cellular stress and damage, and this footprint matched responses described in previously published studies of retinal disease. Of mechanistic importance the transcriptional landscape was consistent with upregulation of folliculin, a recently identified susceptibility gene for diabetic retinopathy. Human study: Adjusting for established risk factors, PP was associated with prevalent and incident clinically significant retinopathy (odds ratio (OR) 1.289 (1.107-1.501) p = 0.001; hazard ratio (HR) 1.259 (1.035-1.531) p = 0.0213), albuminuria (OR 1.277 (1.124-1.454), p = 0.0002; HR 1.608 (1.208-2.141) p = 0.0011), and macrovascular disease (OR 1.021 (1.006-1.037) p = 0.0068; HR 1.324 (1.089-1.61), p = 0.0049), in individuals with type 2 diabetes, and progression to diabetes in non-diabetic individuals (HR 1.402 (1.081-1.818), p = 0.0109). CONCLUSIONS: Elevated fasting PP is independently associated with vascular complications of diabetes and affects retinal pathways potentially influencing retinal neuronal survival. Our results suggest possible new roles for PP-fold peptides in the pathophysiology of diabetes complications and vascular risk stratification.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Angiopathies , Diabetic Retinopathy , Fasting , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Humans , Male , Female , Middle Aged , Diabetic Angiopathies/etiology , Diabetic Angiopathies/epidemiology , Animals , Mice , Follow-Up Studies , Diabetic Retinopathy/etiology , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/pathology , Prognosis , Incidence , Biomarkers/analysis , Risk Factors , Aged
2.
Nat Commun ; 14(1): 5431, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37669965

ABSTRACT

Zinc and plant-derived ligands of the aryl hydrocarbon receptor (AHR) are dietary components affecting intestinal epithelial barrier function. Here, we explore whether zinc and the AHR pathway are linked. We show that dietary supplementation with an AHR pre-ligand offers protection against inflammatory bowel disease in a mouse model while protection fails in mice lacking AHR in the intestinal epithelium. AHR agonist treatment is also ineffective in mice fed zinc depleted diet. In human ileum organoids and Caco-2 cells, AHR activation increases total cellular zinc and cytosolic free Zn2+ concentrations through transcription of genes for zinc importers. Tight junction proteins are upregulated through zinc inhibition of nuclear factor kappa-light-chain-enhancer and calpain activity. Our data show that AHR activation by plant-derived dietary ligands improves gut barrier function at least partly via zinc-dependent cellular pathways, suggesting that combined dietary supplementation with AHR ligands and zinc might be effective in preventing inflammatory gut disorders.


Subject(s)
Receptors, Aryl Hydrocarbon , Zinc , Humans , Animals , Mice , Caco-2 Cells , Ligands , Cytosol , Organic Chemicals
3.
Diabetes ; 71(4): 837-852, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35073578

ABSTRACT

Serum progesterone sulfates were evaluated in the etiology of gestational diabetes mellitus (GDM). Serum progesterone sulfates were measured using ultra-performance liquid chromatography-tandem mass spectrometry in four patient cohorts: 1) the Hyperglycemia and Adverse Pregnancy Outcomes study; 2) London-based women of mixed ancestry and 3) U.K.-based women of European ancestry with or without GDM; and 4) 11-13 weeks pregnant women with BMI ≤25 or BMI ≥35 kg/m2 with subsequent uncomplicated pregnancies or GDM. Glucose-stimulated insulin secretion (GSIS) was evaluated in response to progesterone sulfates in mouse islets and human islets. Calcium fluorescence was measured in HEK293 cells expressing transient receptor potential cation channel subfamily M member 3 (TRPM3). Computer modeling using Molecular Operating Environment generated three-dimensional structures of TRPM3. Epiallopregnanolone sulfate (PM5S) concentrations were reduced in GDM (P < 0.05), in women with higher fasting plasma glucose (P < 0.010), and in early pregnancy samples from women who subsequently developed GDM with BMI ≥35 kg/m2 (P < 0.05). In islets, 50 µmol/L PM5S increased GSIS by at least twofold (P < 0.001); isosakuranetin (TRPM3 inhibitor) abolished this effect. PM5S increased calcium influx in TRPM3-expressing HEK293 cells. Computer modeling and docking showed identical positioning of PM5S to the natural ligand in TRPM3. PM5S increases GSIS and is reduced in GDM serum. The activation of GSIS by PM5S is mediated by TRPM3 in both mouse and human islets.


Subject(s)
Diabetes, Gestational , TRPM Cation Channels , Animals , Blood Glucose/metabolism , Calcium/metabolism , Female , HEK293 Cells , Humans , Insulin/metabolism , Insulin Secretion , Mice , Pregnancy , Progesterone , Sulfates/metabolism
4.
PLoS One ; 16(3): e0249239, 2021.
Article in English | MEDLINE | ID: mdl-33788878

ABSTRACT

Combinatorial gut hormone therapy is one of the more promising strategies for identifying improved treatments for metabolic disease. Many approaches combine the established benefits of glucagon-like peptide-1 (GLP-1) agonism with one or more additional molecules with the aim of improving metabolic outcomes. Recent attention has been drawn to the glucose-dependent insulinotropic polypeptide (GIP) system due to compelling pre-clinical evidence describing the metabolic benefits of antagonising the GIP receptor (GIPR). We rationalised that benefit might be accrued from combining GIPR antagonism with GLP-1 agonism. Two GIPR peptide antagonists, GIPA-1 (mouse GIP(3-30)NH2) and GIPA-2 (NαAc-K10[γEγE-C16]-Arg18-hGIP(5-42)), were pharmacologically characterised and both exhibited potent antagonist properties. Acute in vivo administration of GIPA-1 during an oral glucose tolerance test (OGTT) had negligible effects on glucose tolerance and insulin in lean mice. In contrast, GIPA-2 impaired glucose tolerance and attenuated circulating insulin levels. A mouse model of diet-induced obesity (DIO) was used to investigate the potential metabolic benefits of chronic dosing of each antagonist, alone or in combination with liraglutide. Chronic administration studies showed expected effects of liraglutide, lowering food intake, body weight, fasting blood glucose and plasma insulin concentrations while improving glucose sensitivity, whereas delivery of either GIPR antagonist alone had negligible effects on these parameters. Interestingly, chronic dual therapy augmented insulin sensitizing effects and lowered plasma triglycerides and free-fatty acids, with more notable effects observed with GIPA-1 compared to GIPA-2. Thus, the co-administration of both a GIPR antagonist with a GLP1 agonist uncovers interesting beneficial effects on measures of insulin sensitivity, circulating lipids and certain adipose stores that seem influenced by the degree or nature of GIP receptor antagonism.


Subject(s)
Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucose/metabolism , Amino Acid Sequence , Animals , Blood Glucose/analysis , Body Weight/drug effects , Diet, High-Fat/veterinary , Fatty Acids/blood , Gastric Inhibitory Polypeptide/chemistry , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test , Insulin Secretion , Liraglutide/pharmacology , Male , Mice , Mice, Inbred C57BL , ROC Curve , Triglycerides/blood
5.
Methods Mol Biol ; 2128: 207-216, 2020.
Article in English | MEDLINE | ID: mdl-32180195

ABSTRACT

Type 2 diabetes is characterized by glucose intolerance, caused by insulin resistance in peripheral metabolic tissues and by impaired glucose-stimulated insulin secretion, the hallmark of beta-cell dysfunction. The glucose tolerance test is used in clinic and research to identify individuals with impaired glucose tolerance and overt type 2 diabetes. It is the most routinely used physiological test for first pass assessment of glucose homeostasis in rodents because of its simplicity. The GTT measures changes in blood glucose concentration over a 2-h period following the administration of a bolus of glucose. However, this simplicity belies several important considerations which need to be addressed, to aid reproducibility and produce interpretable data. Here, we describe in detail how to perform a GTT using four different routes of glucose administration: intraperitoneal, oral, voluntary oral, and intravenous.


Subject(s)
Blood Glucose/analysis , Glucose Intolerance/diagnosis , Glucose Tolerance Test/methods , Glucose/administration & dosage , Administration, Intravenous/methods , Administration, Oral , Animals , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/diagnosis , Fasting , Glucose Intolerance/blood , Injections, Intraperitoneal/methods , Insulin/blood , Mice , Reproducibility of Results
6.
Mol Metab ; 34: 157-173, 2020 04.
Article in English | MEDLINE | ID: mdl-32180555

ABSTRACT

OBJECTIVE: Enteroendocrine cells (EECs) survey the gut luminal environment and coordinate hormonal, immune and neuronal responses to it. They exhibit well-characterised physiological roles ranging from the control of local gut function to whole body metabolism, but little is known regarding the regulatory networks controlling their differentiation, especially in the human gut. The small molecule isoxazole-9 (ISX-9) has been shown to stimulate neuronal and pancreatic beta-cell differentiation, both closely related to EEC differentiation. Our aim was to use ISX-9 as a tool to explore EEC differentiation. METHODS: We investigated the effects of ISX-9 on EEC differentiation in mouse and human intestinal organoids, using real-time quantitative polymerase chain reaction (RT-qPCR), fluorescent-activated cell sorting, immunostaining and single-cell RNA sequencing. RESULTS: ISX-9 increased the number of neurogenin3-RFP (Ngn3)-positive endocrine progenitor cells and upregulated NeuroD1 and Pax4, transcription factors that play roles in mouse EEC specification. Single-cell analysis showed induction of Pax4 expression in a developmentally late Ngn3+ population of cells and potentiation of genes associated with progenitors biased toward serotonin-producing enterochromaffin (EC) cells. Further, we observed enrichment of organoids with functional EC cells that was partly dependent on stimulation of calcium signalling in a population of cells residing outside the crypt base. Inducible Pax4 overexpression, in ileal organoids, uncovered its importance as a component of early human endocrine specification and highlighted the potential existence of two major endocrine lineages, the early appearing enterochromaffin lineage and the later developing peptidergic lineage which contains classical gut hormone cell types. CONCLUSION: Our data provide proof-of-concept for the controlled manipulation of specific endocrine lineages with small molecules, whilst also shedding new light on human EEC differentiation and its similarity to the mouse. Given their diverse roles, understanding endocrine lineage plasticity and its control could have multiple therapeutic implications.


Subject(s)
Cell Lineage/drug effects , Enteroendocrine Cells/drug effects , Intestines/cytology , Isoxazoles/pharmacology , Organoids/cytology , Organoids/drug effects , Stem Cells/drug effects , Thiophenes/pharmacology , Animals , Cell Differentiation/drug effects , Enteroendocrine Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Organoids/growth & development , Organoids/metabolism , Stem Cells/metabolism
7.
Diabetologia ; 63(6): 1093-1102, 2020 06.
Article in English | MEDLINE | ID: mdl-32221645

ABSTRACT

Diabetes is one of the most challenging health concerns facing society. Available drugs treat the symptoms but there is no cure. This presents an urgent need to better understand human diabetes in order to develop improved treatments or target remission. New disease models need to be developed that more accurately describe the pathology of diabetes. Organoid technology provides an opportunity to fill this knowledge gap. Organoids are 3D structures, established from pluripotent stem cells or adult stem/progenitor cells, that recapitulate key aspects of the in vivo tissues they mimic. In this review we briefly introduce organoids and their benefits; we focus on organoids generated from tissues important for glucose homeostasis and tissues associated with diabetic complications. We hope this review serves as a touchstone to demonstrate how organoid technology extends the research toolbox and can deliver a step change of discovery in the field of diabetes.


Subject(s)
Diabetes Complications/pathology , Organoids/pathology , Pluripotent Stem Cells/pathology , Animals , Diabetes Mellitus , Disease Models, Animal , Humans , Obesity/pathology
8.
Sci Rep ; 10(1): 3895, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127609

ABSTRACT

Ursodeoxycholic acid (UDCA) treatment can reduce itch and lower endogenous serum bile acids in intrahepatic cholestasis of pregnancy (ICP). We sought to determine how it could influence the gut environment in ICP to alter enterohepatic signalling. The gut microbiota and bile acid content were determined in faeces from 35 pregnant women (14 with uncomplicated pregnancies and 21 with ICP, 17 receiving UDCA). Faecal bile salt hydrolase activity was measured using a precipitation assay. Serum fibroblast growth factor 19 (FGF19) and 7α-hydroxy-4-cholesten-3-one (C4) concentrations were measured following a standardised diet for 21 hours. Women with a high ratio of Bacteroidetes to Firmicutes were more likely to be treated with UDCA (Fisher's exact test p = 0.0178) than those with a lower ratio. Bile salt hydrolase activity was reduced in women with low Bacteroidetes:Firmicutes. Women taking UDCA had higher faecal lithocholic acid (p < 0.0001), with more unconjugated bile acids than women with untreated ICP or uncomplicated pregnancy. UDCA-treatment increased serum FGF19, and reduced C4 (reflecting lower bile acid synthesis). During ICP, UDCA treatment can be associated with enrichment of the gut microbiota with Bacteroidetes. These demonstrate high bile salt hydrolase activity, which deconjugates bile acids enabling secondary modification to FXR agonists, enhancing enterohepatic feedback via FGF19.


Subject(s)
Amidohydrolases/genetics , Bacteroidetes/drug effects , Bacteroidetes/genetics , Cholestasis, Intrahepatic/microbiology , Gene Expression Regulation, Bacterial , Intestines/microbiology , Pregnancy Complications/microbiology , Ursodeoxycholic Acid/pharmacology , Animals , Case-Control Studies , Female , Gastrointestinal Microbiome/drug effects , Humans , Mice , Pregnancy
9.
Gut ; 69(3): 578-590, 2020 03.
Article in English | MEDLINE | ID: mdl-31792136

ABSTRACT

OBJECTIVE: The functional role of interleukin-22 (IL22) in chronic inflammation is controversial, and mechanistic insights into how it regulates target tissue are lacking. In this study, we evaluated the functional role of IL22 in chronic colitis and probed mechanisms of IL22-mediated regulation of colonic epithelial cells. DESIGN: To investigate the functional role of IL22 in chronic colitis and how it regulates colonic epithelial cells, we employed a three-dimentional mini-gut epithelial organoid system, in vivo disease models and transcriptomic datasets in human IBD. RESULTS: As well as inducing transcriptional modules implicated in antimicrobial responses, IL22 also coordinated an endoplasmic reticulum (ER) stress response transcriptional programme in colonic epithelial cells. In the colon of patients with active colonic Crohn's disease (CD), there was enrichment of IL22-responsive transcriptional modules and ER stress response modules. Strikingly, in an IL22-dependent model of chronic colitis, targeting IL22 alleviated colonic epithelial ER stress and attenuated colitis. Pharmacological modulation of the ER stress response similarly impacted the severity of colitis. In patients with colonic CD, antibody blockade of IL12p40, which simultaneously blocks IL12 and IL23, the key upstream regulator of IL22 production, alleviated the colonic epithelial ER stress response. CONCLUSIONS: Our data challenge perceptions of IL22 as a predominantly beneficial cytokine in IBD and provide novel insights into the molecular mechanisms of IL22-mediated pathogenicity in chronic colitis. Targeting IL22-regulated pathways and alleviating colonic epithelial ER stress may represent promising therapeutic strategies in patients with colitis. TRIAL REGISTRATION NUMBER: NCT02749630.


Subject(s)
Colitis/genetics , Crohn Disease/physiopathology , Endoplasmic Reticulum Stress/genetics , Epithelial Cells/physiology , Interleukins/pharmacology , Transcription, Genetic , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Survival/drug effects , Chronic Disease , Colitis/blood , Colitis/drug therapy , Colitis/pathology , Colon/pathology , Crohn Disease/pathology , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Humans , Interleukin-17/pharmacology , Interleukin-23/antagonists & inhibitors , Interleukins/blood , Interleukins/genetics , Intestinal Mucosa/pathology , Mice , Organoids , Patient Acuity , Phenylbutyrates/pharmacology , Recombinant Proteins/pharmacology , Transcription, Genetic/drug effects , Tunicamycin/pharmacology , Unfolded Protein Response , Ustekinumab/pharmacology , Ustekinumab/therapeutic use , Interleukin-22
10.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Article in English | MEDLINE | ID: mdl-31756561

ABSTRACT

BACKGROUND & AIMS: The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis. METHODS: We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice. RESULTS: First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling. CONCLUSIONS: This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.


Subject(s)
Enteroendocrine Cells/physiology , Inhibitor of Apoptosis Proteins/genetics , Intestinal Mucosa/physiology , MicroRNAs/metabolism , Stem Cells/physiology , Animals , Cell Lineage/genetics , Cell Proliferation/genetics , Cells, Cultured , Computational Biology , ErbB Receptors/metabolism , Feeding Behavior/physiology , Female , Inhibitor of Apoptosis Proteins/metabolism , Intestinal Mucosa/cytology , Male , Mice , Mice, Transgenic , Models, Animal , Organoids , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis
12.
Dis Model Mech ; 12(1)2019 01 23.
Article in English | MEDLINE | ID: mdl-30679186

ABSTRACT

Islet inflammation and cytokine production are implicated in pancreatic ß-cell dysfunction and diabetes pathogenesis. However, we lack therapeutics to protect the insulin-producing ß-cells from inflammatory damage. Closing this clinical gap requires the establishment of new disease models of islet inflammation to facilitate screening efforts aimed at identifying new protective agents. Here, we have developed a genetic model of Interleukin-1ß (Il-1ß)-driven islet inflammation in zebrafish, a vertebrate that allows for non-invasive imaging of ß-cells and in vivo drug discovery. Live imaging of immune cells and ß-cells in our model revealed dynamic migration, increased visitation and prolonged macrophage retention in the islet, together with robust activation of NF-κB signalling in ß-cells. We find that Il-1ß-mediated inflammation does not cause ß-cell destruction but, rather, it impairs ß-cell function and identity. In vivo, ß-cells exhibit impaired glucose-stimulated calcium influx and reduced expression of genes involved in function and maturity. These defects are accompanied by α-cell expansion, glucose intolerance and hyperglycemia following a glucose challenge. Notably, we show that a medicinal plant derivative (wedelolactone) is capable of reducing the immune-cell infiltration while also ameliorating the hyperglycemic phenotype of our model. Importantly, these anti-diabetic properties in zebrafish are predictive of wedelolactone's efficacy in protecting rodent and human islets from cytokine-induced apoptosis. In summary, this new zebrafish model of diabetes opens a window to study the interactions between immune and ß-cells in vivo, while also allowing the identification of therapeutic agents for protecting ß-cells from inflammation.


Subject(s)
Biological Products/pharmacology , Coumarins/pharmacology , Inflammation/pathology , Insulin-Secreting Cells/pathology , Animals , Animals, Genetically Modified , Apoptosis/drug effects , Calcium/metabolism , Cytokines/pharmacology , Disease Models, Animal , Down-Regulation/drug effects , Glucose/pharmacology , Humans , Hyperglycemia/genetics , Hyperglycemia/pathology , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-1beta/metabolism , Larva/drug effects , Larva/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Models, Genetic , Time-Lapse Imaging , Transcription, Genetic/drug effects , Zebrafish
13.
Elife ; 72018 04 06.
Article in English | MEDLINE | ID: mdl-29624168

ABSTRACT

The pancreatic islet, a cellular community harboring the insulin-producing beta-cells, is known to undergo age-related alterations. However, only a handful of signals associated with aging have been identified. By comparing beta-cells from younger and older zebrafish, here we show that the aging islets exhibit signs of chronic inflammation. These include recruitment of tnfα-expressing macrophages and the activation of NF-kB signaling in beta-cells. Using a transgenic reporter, we show that NF-kB activity is undetectable in juvenile beta-cells, whereas cells from older fish exhibit heterogeneous NF-kB activity. We link this heterogeneity to differences in gene expression and proliferation. Beta-cells with high NF-kB signaling proliferate significantly less compared to their neighbors with low activity. The NF-kB signalinghi cells also exhibit premature upregulation of socs2, an age-related gene that inhibits beta-cell proliferation. Together, our results show that NF-kB activity marks the asynchronous decline in beta-cell proliferation with advancing age.


Subject(s)
Aging , Cell Proliferation , Inflammation Mediators/metabolism , Inflammation/pathology , Insulin-Secreting Cells/pathology , NF-kappa B/metabolism , Zebrafish/physiology , Animals , Animals, Genetically Modified , Cells, Cultured , Gene Expression Profiling , Inflammation/immunology , Inflammation/metabolism , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , NF-kappa B/genetics , Signal Transduction , Single-Cell Analysis , Transcriptional Activation , Zebrafish/immunology
14.
Diabetes Obes Metab ; 20(3): 599-609, 2018 03.
Article in English | MEDLINE | ID: mdl-28940946

ABSTRACT

AIMS: Two unmet therapeutic strategies for diabetes treatment are prevention of beta-cell death and stimulation of beta-cell replication. Our aim was to characterize the role of neuropeptide Y receptors in the control of beta-cell mass. MATERIALS AND METHODS: We used endogenous and selective agonists of the NPY receptor system to explore its role in the prevention of beta-cell apoptosis and proliferation in islets isolated from both mouse and human donors. We further explored the intra-cellular signalling cascades involved, using chemical inhibitors of key signalling pathways. As proof of principle we designed a long-acting analogue of [Leu31 Pro34 ]-NPY, an agonist of the islet-expressed Y receptors, to determine if targeting this system could preserve beta-cell mass in vivo. RESULTS: Our data reveal that NPY Y1, 4 and 5 receptor activation engages a generalized and powerful anti-apoptotic pathway that protects mouse and human islets from damage. These anti-apoptotic effects were dependent on stimulating a Gαi-PLC-PKC signalling cascade, which prevented cytokine-induced NFkB signalling. NPY receptor activation functionally protected islets by restoring glucose responsiveness following chemically induced injury in both species. NPY receptor activation attenuated beta-cell apoptosis, preserved functional beta-cell mass and attenuated the hyperglycaemic phenotype in a low-dose streptozotocin model of diabetes. CONCLUSION: Taken together, our observations identify the islet Y receptors as promising targets for the preservation of beta-cell mass. As such, targeting these receptors could help to maintain beta-cell mass in both type 1 and type 2 diabetes, and may also be useful for improving islet transplantation outcomes.


Subject(s)
Insulin-Secreting Cells/cytology , Receptors, Neuropeptide Y/physiology , Analysis of Variance , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Humans , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism , Signal Transduction/physiology
16.
Curr Opin Pharmacol ; 37: 51-58, 2017 12.
Article in English | MEDLINE | ID: mdl-28968540

ABSTRACT

The advent of near physiological organoid technology has produced a step change in our understanding of stem cells and has provided the research community with a powerful new cell based tool to model human physiology and disease. We review the pros and cons of intestinal organoid culture systems. The molecular and genetic tools to manipulate them and how they are being used to answer fundamental questions in metabolic research, including the function of enteroendocrine cells in health and disease.


Subject(s)
Intestinal Mucosa/metabolism , Organoids/metabolism , Animals , Biomedical Research , Enteroendocrine Cells/metabolism , Humans , Metabolic Diseases , Models, Biological , Pluripotent Stem Cells/metabolism
17.
Mol Metab ; 6(1): 48-60, 2017 01.
Article in English | MEDLINE | ID: mdl-28123937

ABSTRACT

OBJECTIVE: Dietary supplementation with fermentable carbohydrate protects against body weight gain. Fermentation by the resident gut microbiota produces short-chain fatty acids, which act at free fatty acid receptor 2 (FFAR2). Our aim was to test the hypothesis that FFAR2 is important in regulating the beneficial effects of fermentable carbohydrate on body weight and to understand the role of gut hormones PYY and GLP-1. METHODS: Wild-type or Ffar2-/- mice were fed an inulin supplemented or control diet. Mice were metabolically characterized and gut hormone concentrations, enteroendocrine cell density measurements were carried out. Intestinal organoids and colonic cultures were utilized to substantiate the in vivo findings. RESULTS: We provide new mechanistic insight into how fermentable carbohydrate regulates metabolism. Using mice that lack FFAR2, we demonstrate that the fermentable carbohydrate inulin acts via this receptor to drive an 87% increase in the density of cells that produce the appetite-suppressing hormone peptide YY (PYY), reduce food intake, and prevent diet-induced obesity. CONCLUSION: Our results demonstrate that FFAR2 is predominantly involved in regulating the effects of fermentable carbohydrate on metabolism and does so, in part, by enhancing PYY cell density and release. This highlights the potential for targeting enteroendocrine cell differentiation to treat obesity.


Subject(s)
Dietary Carbohydrates/metabolism , Peptide YY/metabolism , Receptors, Cell Surface/metabolism , Animals , Body Weight , Colon/cytology , Dietary Supplements , Eating , Fatty Acids, Volatile/metabolism , Fermentation , Fermented Foods , Gastrointestinal Hormones/metabolism , Gastrointestinal Microbiome/physiology , Glucagon-Like Peptide 1/metabolism , Inulin/metabolism , Male , Mice , Mice, Knockout , Obesity/metabolism , Receptors, Cell Surface/physiology , Weight Gain
18.
Obesity (Silver Spring) ; 23(6): 1194-200, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25958858

ABSTRACT

OBJECTIVE: The G-protein coupled receptor family C group 6 member A (GPRC6A) is activated by proteinogenic amino acids and may sense amino acids in the gastrointestinal tract and the brain. The study investigated whether GPRC6A was necessary for the effects of low- and high-protein diets on body weight and food intake in mice. METHODS: The role of GPRC6A in mediating the effects of a low-protein diet on body weight was investigated in GPRC6a knockout (GPRC6a-KO) and wild-type (WT) mice fed a control diet (18% protein) or a low-protein diet (6% protein) for 9 days. The role of GPRC6A in mediating the effects of a high-protein diet on body weight was investigated in GPRC6a-KO and WT mice fed a control diet (18% protein) or a high-protein diet (50% protein) for 5 weeks. RESULTS: A high-protein diet reduced body weight gain and food intake compared with a control diet in both WT and GPRC6a-KO mice. A low-protein diet decreased body weight gain in GPRC6a-KO mice. CONCLUSIONS: GPRC6A was not necessary for the effects of a low- or high-protein diet on body weight and likely does not play a role in protein-induced satiety.


Subject(s)
Body Weight/drug effects , Dietary Proteins/administration & dosage , Receptors, G-Protein-Coupled/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Weight Gain/drug effects
19.
Diabetologia ; 57(9): 1762-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24917132

ABSTRACT

Replenishment of beta cell mass is a key aim of novel therapeutic interventions for diabetes, and the implementation of new strategies will be aided by understanding the mechanisms employed to regulate beta cell mass under normal physiological conditions. We have recently identified a new role for the gut hormone peptide YY (PYY) and the neuropeptide Y (NPY) receptor systems in the control of beta cell survival. PYY is perhaps best known for its role in regulating appetite and body weight, but its production by islet cells, the presence of NPY receptors on islets and the demonstration that Y1 activation causes proliferation of beta cells and protects them from apoptosis, suggest a role for this peptide in modulating beta cell mass. This review introduces PYY and its potential role in glucose homeostasis, then focuses on evidence supporting the concept that PYY and NPY receptors are exciting new targets for the preservation of beta cells.


Subject(s)
Appetite Regulation/physiology , Peptide YY/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Diabetes Mellitus/metabolism , Glucose/metabolism , Homeostasis , Humans
20.
Biochem Med (Zagreb) ; 22(3): 283-97, 2012.
Article in English | MEDLINE | ID: mdl-23092061

ABSTRACT

Peptide hormones are released from the gastrointestinal tract in response to nutrients and communicate information regarding the current state of energy balance to the brain. These hormones regulate appetite, energy expenditure and glucose homeostasis. They can act either via the circulation at target peripheral tissues, by activation of the vagus nerve or by acting on key brain regions implicated in energy homeostasis such as the hypothalamus and brainstem. This review gives an overview of the main gut hormones implicated in the regulation of food intake and how some of these are being targeted to develop anti obesity treatments.


Subject(s)
Gastrointestinal Hormones/metabolism , Obesity/metabolism , Animals , Appetite/drug effects , Appetite/physiology , Eating/drug effects , Gastrointestinal Hormones/therapeutic use , Humans , Obesity/drug therapy , Peptide Hormones/metabolism , Peptide Hormones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...