Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 20(3): 350-361, 2019 03.
Article in English | MEDLINE | ID: mdl-30718914

ABSTRACT

Despite the known importance of zinc for human immunity, molecular insights into its roles have remained limited. Here we report a novel autosomal recessive disease characterized by absent B cells, agammaglobulinemia and early onset infections in five unrelated families. The immunodeficiency results from hypomorphic mutations of SLC39A7, which encodes the endoplasmic reticulum-to-cytoplasm zinc transporter ZIP7. Using CRISPR-Cas9 mutagenesis we have precisely modeled ZIP7 deficiency in mice. Homozygosity for a null allele caused embryonic death, but hypomorphic alleles reproduced the block in B cell development seen in patients. B cells from mutant mice exhibited a diminished concentration of cytoplasmic free zinc, increased phosphatase activity and decreased phosphorylation of signaling molecules downstream of the pre-B cell and B cell receptors. Our findings highlight a specific role for cytosolic Zn2+ in modulating B cell receptor signal strength and positive selection.


Subject(s)
Agammaglobulinemia/immunology , B-Lymphocytes/immunology , Cation Transport Proteins/immunology , Zinc/immunology , Agammaglobulinemia/genetics , Agammaglobulinemia/metabolism , Animals , B-Lymphocytes/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Child, Preschool , Cytosol/immunology , Cytosol/metabolism , Disease Models, Animal , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Female , Gene Expression Profiling , Humans , Infant , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Pedigree , Zinc/metabolism
2.
Diabetologia ; 67(6): 1079-1094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38512414

ABSTRACT

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.


Subject(s)
CpG Islands , DNA Methylation , Insulin-Secreting Cells , Insulin-Secreting Cells/metabolism , Animals , Mice , CpG Islands/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Transgenic , DNA Methyltransferase 3A/metabolism , Humans , Insulin/metabolism , Insulin Secretion/physiology
3.
Diabetologia ; 65(6): 997-1011, 2022 06.
Article in English | MEDLINE | ID: mdl-35294578

ABSTRACT

AIMS/HYPOTHESIS: Although targeted in extrapancreatic tissues by several drugs used to treat type 2 diabetes, the role of AMP-activated protein kinase (AMPK) in the control of insulin secretion is still debatable. Previous studies have used pharmacological activators of limited selectivity and specificity, and none has examined in primary pancreatic beta cells the actions of the latest generation of highly potent and specific activators that act via the allosteric drug and metabolite (ADaM) site. METHODS: AMPK was activated acutely in islets isolated from C57BL6/J mice, and in an EndoC-ßH3 cell line, using three structurally distinct ADaM site activators (991, PF-06409577 and RA089), with varying selectivity for ß1- vs ß2-containing complexes. Mouse lines expressing a gain-of-function mutation in the γ1 AMPK subunit (D316a) were generated to examine the effects of chronic AMPK stimulation in the whole body, or selectively in the beta cell. RESULTS: Acute (1.5 h) treatment of wild-type mouse islets with 991, PF-06409577 or RA089 robustly stimulated insulin secretion at high glucose concentrations (p<0.01, p<0.05 and p<0.001, respectively), despite a lowering of glucose-induced intracellular free Ca2+ dynamics in response to 991 (AUC, p<0.05) and to RA089 at the highest dose (25 µmol/l) at 5.59 min (p<0.05). Although abolished in the absence of AMPK, the effects of 991 were observed in the absence of the upstream kinase, liver kinase B1, further implicating 'amplifying' pathways. In marked contrast, chronic activation of AMPK, either globally or selectively in the beta cell, achieved using a gain-of-function mutant, impaired insulin release in vivo (p<0.05 at 15 min following i.p. injection of 3 mmol/l glucose) and in vitro (p<0.01 following incubation of islets with 17 mmol/l glucose), and lowered glucose tolerance (p<0.001). CONCLUSIONS/INTERPRETATION: AMPK activation exerts complex, time-dependent effects on insulin secretion. These observations should inform the design and future clinical use of AMPK modulators.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice
4.
Diabetologia ; 64(4): 850-864, 2021 04.
Article in English | MEDLINE | ID: mdl-33492421

ABSTRACT

AIMS/HYPOTHESIS: Variants close to the VPS13C/C2CD4A/C2CD4B locus are associated with altered risk of type 2 diabetes in genome-wide association studies. While previous functional work has suggested roles for VPS13C and C2CD4A in disease development, none has explored the role of C2CD4B. METHODS: CRISPR/Cas9-induced global C2cd4b-knockout mice and zebrafish larvae with c2cd4a deletion were used to study the role of this gene in glucose homeostasis. C2 calcium dependent domain containing protein (C2CD)4A and C2CD4B constructs tagged with FLAG or green fluorescent protein were generated to investigate subcellular dynamics using confocal or near-field microscopy and to identify interacting partners by mass spectrometry. RESULTS: Systemic inactivation of C2cd4b in mice led to marked, but highly sexually dimorphic changes in body weight and glucose homeostasis. Female C2cd4b mice displayed unchanged body weight compared with control littermates, but abnormal glucose tolerance (AUC, p = 0.01) and defective in vivo, but not in vitro, insulin secretion (p = 0.02). This was associated with a marked decrease in follicle-stimulating hormone levels as compared with wild-type (WT) littermates (p = 0.003). In sharp contrast, male C2cd4b null mice displayed essentially normal glucose tolerance but an increase in body weight (p < 0.001) and fasting blood glucose (p = 0.003) after maintenance on a high-fat and -sucrose diet vs WT littermates. No metabolic disturbances were observed after global inactivation of C2cd4a in mice, or in pancreatic beta cell function at larval stages in C2cd4a null zebrafish. Fasting blood glucose levels were also unaltered in adult C2cd4a-null fish. C2CD4B and C2CD4A were partially localised to the plasma membrane, with the latter under the control of intracellular Ca2+. Binding partners for both included secretory-granule-localised PTPRN2/phogrin. CONCLUSIONS/INTERPRETATION: Our studies suggest that C2cd4b may act centrally in the pituitary to influence sex-dependent circuits that control pancreatic beta cell function and glucose tolerance in rodents. However, the absence of sexual dimorphism in the impact of diabetes risk variants argues for additional roles for C2CD4A or VPS13C in the control of glucose homeostasis in humans. DATA AVAILABILITY: The datasets generated and/or analysed during the current study are available in the Biorxiv repository ( www.biorxiv.org/content/10.1101/2020.05.18.099200v1 ). RNA-Seq (GSE152576) and proteomics (PXD021597) data have been deposited to GEO ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152576 ) and ProteomeXchange ( www.ebi.ac.uk/pride/archive/projects/PXD021597 ) repositories, respectively.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Homeostasis/genetics , Insulin-Secreting Cells/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Biomarkers/blood , Blood Glucose/genetics , Female , Follicle Stimulating Hormone/blood , Genotype , Humans , Insulin/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pituitary Gland/metabolism , Sex Characteristics , Weight Gain , Zebrafish/blood , Zebrafish/genetics , Zebrafish Proteins/blood , Zebrafish Proteins/genetics
5.
Nature ; 524(7565): 356-60, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26258299

ABSTRACT

The typical response of the adult mammalian pulmonary circulation to a low oxygen environment is vasoconstriction and structural remodelling of pulmonary arterioles, leading to chronic elevation of pulmonary artery pressure (pulmonary hypertension) and right ventricular hypertrophy. Some mammals, however, exhibit genetic resistance to hypoxia-induced pulmonary hypertension. We used a congenic breeding program and comparative genomics to exploit this variation in the rat and identified the gene Slc39a12 as a major regulator of hypoxia-induced pulmonary vascular remodelling. Slc39a12 encodes the zinc transporter ZIP12. Here we report that ZIP12 expression is increased in many cell types, including endothelial, smooth muscle and interstitial cells, in the remodelled pulmonary arterioles of rats, cows and humans susceptible to hypoxia-induced pulmonary hypertension. We show that ZIP12 expression in pulmonary vascular smooth muscle cells is hypoxia dependent and that targeted inhibition of ZIP12 inhibits the rise in intracellular labile zinc in hypoxia-exposed pulmonary vascular smooth muscle cells and their proliferation in culture. We demonstrate that genetic disruption of ZIP12 expression attenuates the development of pulmonary hypertension in rats housed in a hypoxic atmosphere. This new and unexpected insight into the fundamental role of a zinc transporter in mammalian pulmonary vascular homeostasis suggests a new drug target for the pharmacological management of pulmonary hypertension.


Subject(s)
Cation Transport Proteins/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Animals, Congenic , Arterioles/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cattle , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Chromosomes, Mammalian/genetics , Chronic Disease , Female , Gene Knockdown Techniques , Homeostasis , Humans , Hypertension, Pulmonary/genetics , Hypoxia/genetics , Intracellular Space/metabolism , Male , Muscle, Smooth, Vascular/cytology , Rats , Rats, Inbred F344 , Rats, Inbred WKY , Zinc/metabolism
6.
Proc Natl Acad Sci U S A ; 115(32): E7642-E7649, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038024

ABSTRACT

SLC30A8 encodes a zinc transporter that is primarily expressed in the pancreatic islets of Langerhans. In ß-cells it transports zinc into insulin-containing secretory granules. Loss-of-function (LOF) mutations in SLC30A8 protect against type 2 diabetes in humans. In this study, we generated a knockin mouse model carrying one of the most common human LOF mutations for SLC30A8, R138X. The R138X mice had normal body weight, glucose tolerance, and pancreatic ß-cell mass. Interestingly, in hyperglycemic conditions induced by the insulin receptor antagonist S961, the R138X mice showed a 50% increase in insulin secretion. This effect was not associated with enhanced ß-cell proliferation or mass. Our data suggest that the SLC30A8 R138X LOF mutation may exert beneficial effects on glucose metabolism by increasing the capacity of ß-cells to secrete insulin under hyperglycemic conditions.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Zinc Transporter 8/genetics , Alleles , Animals , Blood Glucose , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Gene Knock-In Techniques , Humans , Hyperglycemia/blood , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Insulin Secretion , Loss of Function Mutation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/pharmacology , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/metabolism , Zinc Transporter 8/metabolism
7.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498234

ABSTRACT

Diabetes mellitus now affects more than 400 million individuals worldwide, with significant impacts on the lives of those affected and associated socio-economic costs. Although defects in insulin secretion underlie all forms of the disease, the molecular mechanisms which drive them are still poorly understood. Subsets of specialised beta cells have, in recent years, been suggested to play critical roles in "pacing" overall islet activity. The molecular nature of these cells, the means through which their identity is established and the changes which may contribute to their functional demise and "loss of influence" in both type 1 and type 2 diabetes are largely unknown. Genomic imprinting involves the selective silencing of one of the two parental alleles through DNA methylation and modified imprinted gene expression is involved in a number of diseases. Loss of expression, or loss of imprinting, can be shown in mouse models to lead to defects in beta cell function and abnormal insulin secretion. In the present review we survey the evidence that altered expression of imprinted genes contribute to loss of beta cell function, the importance of beta cell heterogeneity in normal and disease states, and hypothesise whether there is a direct link between the two.


Subject(s)
Diabetes Mellitus/genetics , Genomic Imprinting , Insulin-Secreting Cells/metabolism , Animals , Diabetes Mellitus/metabolism , Humans , Insulin Secretion , Single-Cell Analysis , Transcriptome
8.
Am J Hum Genet ; 100(2): 238-256, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28132686

ABSTRACT

Genetic variants near ARAP1 (CENTD2) and STARD10 influence type 2 diabetes (T2D) risk. The risk alleles impair glucose-induced insulin secretion and, paradoxically but characteristically, are associated with decreased proinsulin:insulin ratios, indicating improved proinsulin conversion. Neither the identity of the causal variants nor the gene(s) through which risk is conferred have been firmly established. Whereas ARAP1 encodes a GTPase activating protein, STARD10 is a member of the steroidogenic acute regulatory protein (StAR)-related lipid transfer protein family. By integrating genetic fine-mapping and epigenomic annotation data and performing promoter-reporter and chromatin conformational capture (3C) studies in ß cell lines, we localize the causal variant(s) at this locus to a 5 kb region that overlaps a stretch-enhancer active in islets. This region contains several highly correlated T2D-risk variants, including the rs140130268 indel. Expression QTL analysis of islet transcriptomes from three independent subject groups demonstrated that T2D-risk allele carriers displayed reduced levels of STARD10 mRNA, with no concomitant change in ARAP1 mRNA levels. Correspondingly, ß-cell-selective deletion of StarD10 in mice led to impaired glucose-stimulated Ca2+ dynamics and insulin secretion and recapitulated the pattern of improved proinsulin processing observed at the human GWAS signal. Conversely, overexpression of StarD10 in the adult ß cell improved glucose tolerance in high fat-fed animals. In contrast, manipulation of Arap1 in ß cells had no impact on insulin secretion or proinsulin conversion in mice. This convergence of human and murine data provides compelling evidence that the T2D risk associated with variation at this locus is mediated through reduction in STARD10 expression in the ß cell.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Phosphoproteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alleles , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Diabetes Mellitus, Type 2/blood , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Genetic Variation , Homeostasis , Humans , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/metabolism , Liver/metabolism , Mice , Proinsulin/blood , Proinsulin/metabolism , Quantitative Trait Loci , Transcriptome
9.
J Biol Chem ; 292(21): 8892-8906, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28377501

ABSTRACT

Heterozygous mutations in the human paired box gene PAX6 lead to impaired glucose tolerance. Although embryonic deletion of the Pax6 gene in mice leads to loss of most pancreatic islet cell types, the functional consequences of Pax6 loss in adults are poorly defined. Here we developed a mouse line in which Pax6 was selectively inactivated in ß cells by crossing animals with floxed Pax6 alleles to mice expressing the inducible Pdx1CreERT transgene. Pax6 deficiency, achieved by tamoxifen injection, caused progressive hyperglycemia. Although ß cell mass was preserved 8 days post-injection, total insulin content and insulin:chromogranin A immunoreactivity were reduced by ∼60%, and glucose-stimulated insulin secretion was eliminated. RNA sequencing and quantitative real-time PCR analyses revealed that, although the expression of key ß cell genes, including Ins2, Slc30a8, MafA, Slc2a2, G6pc2, and Glp1r, was reduced after Pax6 deletion, that of several genes that are usually selectively repressed ("disallowed") in ß cells, including Slc16a1, was increased. Assessed in intact islets, glucose-induced ATP:ADP increases were significantly reduced (p < 0.05) in ßPax6KO versus control ß cells, and the former displayed attenuated increases in cytosolic Ca2+ Unexpectedly, glucose-induced increases in intercellular connectivity were enhanced after Pax6 deletion, consistent with increases in the expression of the glucose sensor glucokinase, but decreases in that of two transcription factors usually expressed in fully differentiated ß-cells, Pdx1 and Nkx6.1, were observed in islet "hub" cells. These results indicate that Pax6 is required for the functional identity of adult ß cells. Furthermore, deficiencies in ß cell glucose sensing are likely to contribute to defective insulin secretion in human carriers of PAX6 mutations.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium Signaling , Calcium/metabolism , Gene Expression Regulation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , PAX6 Transcription Factor/biosynthesis , Adenosine Triphosphate/genetics , Animals , Humans , Mice , Mice, Knockout , PAX6 Transcription Factor/genetics
10.
Brain ; 140(4): 928-939, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28334855

ABSTRACT

A novel autosomal recessive cerebro-renal syndrome was identified in consanguineous Bedouin kindred: neurological deterioration was evident as of early age, progressing into severe intellectual disability, profound ataxia, camptocormia and oculomotor apraxia. Brain MRI was normal. Four of the six affected individuals also had early-onset nephropathy with features of tubulo-interstitial nephritis, hypertension and tendency for hyperkalemia, though none had rapid deterioration of renal function. Genome wide linkage analysis identified an ∼18 Mb disease-associated locus on chromosome 4 (maximal logarithm of odds score 4.4 at D4S2971; θ = 0). Whole exome sequencing identified a single mutation in SLC30A9 within this locus, segregating as expected within the kindred and not found in a homozygous state in 300 Bedouin controls. We showed that SLC30A9 (solute carrier family 30 member 9; also known as ZnT-9) is ubiquitously expressed with high levels in cerebellum, skeletal muscle, thymus and kidney. Confocal analysis of SH-SY5Y cells overexpressing SLC30A9 fused to enhanced green fluorescent protein demonstrated vesicular cytosolic localization associated with the endoplasmic reticulum, not co-localizing with endosomal or Golgi markers. SLC30A9 encodes a putative zinc transporter (by similarity) previously associated with Wnt signalling. However, using dual-luciferase reporter assay in SH-SY5Y cells we showed that Wnt signalling was not affected by the mutation. Based on protein modelling, the identified mutation is expected to affect SLC30A9's highly conserved cation efflux domain, putatively disrupting its transmembrane helix structure. Cytosolic Zn2+ measurements in HEK293 cells overexpressing wild-type and mutant SLC30A9 showed lower zinc concentration within mutant rather than wild-type SLC30A9 cells. This suggests that SLC30A9 has zinc transport properties affecting intracellular zinc homeostasis, and that the molecular mechanism of the disease is through defective function of this novel activity of SLC30A9 rather than by a defect in its previously described role in transcriptional activation of Wnt signalling.


Subject(s)
Cation Transport Proteins/genetics , Cell Cycle Proteins/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Homeostasis/genetics , Intellectual Disability/genetics , Kidney Diseases/genetics , Nuclear Proteins/genetics , Zinc/metabolism , Age of Onset , Arabs , Chromosome Mapping , Consanguinity , Cytosol/metabolism , Cytosol/ultrastructure , Female , Genome-Wide Association Study , HEK293 Cells , Humans , Infant , Male , Mutation , Pedigree , Syndrome , Transcription Factors , Wnt Signaling Pathway/genetics
11.
Diabetes Obes Metab ; 19 Suppl 1: 30-41, 2017 09.
Article in English | MEDLINE | ID: mdl-28466490

ABSTRACT

Ca2+ is the key intracellular regulator of insulin secretion, acting in the ß-cell as the ultimate trigger for exocytosis. In response to high glucose, ATP-sensitive K+ channel closure and plasma membrane depolarization engage a sophisticated machinery to drive pulsatile cytosolic Ca2+ changes. Voltage-gated Ca2+ channels, Ca2+ -activated K+ channels and Na+ /Ca2+ exchange all play important roles. The use of targeted Ca2+ probes has revealed that during each cytosolic Ca2+ pulse, uptake of Ca2+ by mitochondria, endoplasmic reticulum (ER), secretory granules and lysosomes fine-tune cytosolic Ca2+ dynamics and control organellar function. For example, changes in the expression of the Ca2+ -binding protein Sorcin appear to provide a link between ER Ca2+ levels and ER stress, affecting ß-cell function and survival. Across the islet, intercellular communication between highly interconnected "hubs," which act as pacemaker ß-cells, and subservient "followers," ensures efficient insulin secretion. Loss of connectivity is seen after the deletion of genes associated with type 2 diabetes (T2D) and follows metabolic and inflammatory insults that characterize this disease. Hubs, which typically comprise ~1%-10% of total ß-cells, are repurposed for their specialized role by expression of high glucokinase (Gck) but lower Pdx1 and Nkx6.1 levels. Single cell-omics are poised to provide a deeper understanding of the nature of these cells and of the networks through which they communicate. New insights into the control of both the intra- and intercellular Ca2+ dynamics may thus shed light on T2D pathology and provide novel opportunities for therapy.


Subject(s)
Calcium Signaling , Cell Communication , Islets of Langerhans/metabolism , Models, Biological , Animals , Cell Membrane/enzymology , Cell Membrane/metabolism , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Exocytosis , Gap Junctions/enzymology , Gap Junctions/metabolism , Gene Expression Regulation , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/enzymology , Mitochondria/enzymology , Mitochondria/metabolism , Secretory Pathway
12.
Am J Physiol Endocrinol Metab ; 311(2): E488-507, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27329800

ABSTRACT

Single nucleotide polymorphisms (SNPs) close to the VPS13C, C2CD4A and C2CD4B genes on chromosome 15q are associated with impaired fasting glucose and increased risk of type 2 diabetes. eQTL analysis revealed an association between possession of risk (C) alleles at a previously implicated causal SNP, rs7163757, and lowered VPS13C and C2CD4A levels in islets from female (n = 40, P < 0.041) but not from male subjects. Explored using promoter-reporter assays in ß-cells and other cell lines, the risk variant at rs7163757 lowered enhancer activity. Mice deleted for Vps13c selectively in the ß-cell were generated by crossing animals bearing a floxed allele at exon 1 to mice expressing Cre recombinase under Ins1 promoter control (Ins1Cre). Whereas Vps13c(fl/fl):Ins1Cre (ßVps13cKO) mice displayed normal weight gain compared with control littermates, deletion of Vps13c had little effect on glucose tolerance. Pancreatic histology revealed no significant change in ß-cell mass in KO mice vs. controls, and glucose-stimulated insulin secretion from isolated islets was not altered in vitro between control and ßVps13cKO mice. However, a tendency was observed in female null mice for lower insulin levels and ß-cell function (HOMA-B) in vivo. Furthermore, glucose-stimulated increases in intracellular free Ca(2+) were significantly increased in islets from female KO mice, suggesting impaired Ca(2+) sensitivity of the secretory machinery. The present data thus provide evidence for a limited role for changes in VPS13C expression in conferring altered disease risk at this locus, particularly in females, and suggest that C2CD4A may also be involved.


Subject(s)
Calcium-Binding Proteins/genetics , Glucose Intolerance/genetics , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/genetics , Proteins/genetics , Animals , Blotting, Western , Calcium/metabolism , Diabetes Mellitus, Type 2/genetics , Female , Glucagon-Secreting Cells/pathology , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout , Pancreas/pathology , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Sex Factors , Vesicular Transport Proteins
13.
Arch Biochem Biophys ; 611: 79-85, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27262257

ABSTRACT

Zn2+ ions are essential for the normal processing and storage of insulin and altered pancreatic insulin content is associated with all forms of diabetes mellitus. Work of the past decade has identified variants in the human SLC30A8 gene, encoding the zinc transporter ZnT8 which is expressed highly selectively on the secretory granule of pancreatic islet ß and α cells, as affecting the risk of Type 2 Diabetes. Here, we review the regulation and roles of Zn2+ ions in islet cells, the mechanisms through which SLC30A8 variants might affect glucose homeostasis and diabetes risk, and the novel technologies including recombinant targeted zinc probes and knockout mice which have been developed to explore these questions.


Subject(s)
Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Zinc/physiology , Animals , Cytoplasm/metabolism , Diabetes Mellitus, Type 2/genetics , Genome-Wide Association Study , Homeostasis , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Metallothionein/metabolism , Mice , Zinc Transporter 8
14.
Cells ; 12(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36980244

ABSTRACT

The rare SLC30A8 mutation encoding a truncating p.Arg138* variant (R138X) in zinc transporter 8 (ZnT8) is associated with a 65% reduced risk for type 2 diabetes. To determine whether ZnT8 is required for beta cell development and function, we derived human pluripotent stem cells carrying the R138X mutation and differentiated them into insulin-producing cells. We found that human pluripotent stem cells with homozygous or heterozygous R138X mutation and the null (KO) mutation have normal efficiency of differentiation towards insulin-producing cells, but these cells show diffuse granules that lack crystalline zinc-containing insulin granules. Insulin secretion is not compromised in vitro by KO or R138X mutations in human embryonic stem cell-derived beta cells (sc-beta cells). Likewise, the ability of sc-beta cells to secrete insulin and maintain glucose homeostasis after transplantation into mice was comparable across different genotypes. Interestingly, sc-beta cells with the SLC30A8 KO mutation showed increased cytoplasmic zinc, and cells with either KO or R138X mutation were resistant to apoptosis when extracellular zinc was limiting. These findings are consistent with a protective role of zinc in cell death and with the protective role of zinc in T2D.


Subject(s)
Cation Transport Proteins , Diabetes Mellitus, Type 2 , Human Embryonic Stem Cells , Zinc Transporter 8 , Zinc , Animals , Humans , Mice , Apoptosis/genetics , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/physiology , Insulin/metabolism , Loss of Function Mutation , Mutation/genetics , Zinc/metabolism , Zinc Transporter 8/genetics , Zinc Transporter 8/metabolism
15.
Life Sci ; 316: 121436, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36706832

ABSTRACT

AIMS: Spatially-organized increases in cytosolic Ca2+ within pancreatic beta cells in the pancreatic islet underlie the stimulation of insulin secretion by high glucose. Recent data have revealed the existence of subpopulations of beta cells including "leaders" which initiate Ca2+ waves. Whether leader cells possess unique molecular features, or localisation, is unknown. MAIN METHODS: High speed confocal Ca2+ imaging was used to identify leader cells and connectivity analysis, running under MATLAB and Python, to identify highly connected "hub" cells. To explore transcriptomic differences between beta cell sub-groups, individual leaders or followers were labelled by photo-activation of the cryptic fluorescent protein PA-mCherry and subjected to single cell RNA sequencing ("Flash-Seq"). KEY FINDINGS: Distinct Ca2+ wave types were identified in individual islets, with leader cells present in 73 % (28 of 38 islets imaged). Scale-free, power law-adherent behaviour was also observed in 29 % of islets, though "hub" cells in these islets did not overlap with leaders. Transcripts differentially expressed (295; padj < 0.05) between leader and follower cells included genes involved in cilium biogenesis and transcriptional regulation. Providing some support for these findings, ADCY6 immunoreactivity tended to be higher in leader than follower cells, whereas cilia number and length tended to be lower in the former. Finally, leader cells were located significantly closer to delta, but not alpha, cells in Euclidian space than were follower cells. SIGNIFICANCE: The existence of both a discrete transcriptome and unique localisation implies a role for these features in defining the specialized function of leaders. These data also raise the possibility that localised signalling between delta and leader cells contributes to the initiation and propagation of islet Ca2+ waves.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Insulin Secretion , Gene Expression Regulation , Cell Line , Insulin/metabolism , Glucose/metabolism
16.
bioRxiv ; 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38076935

ABSTRACT

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 focussing on the imprinted gene 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 eGFP under the control of the Nnat enhancer/promoter regions were generated for fluorescence-activated cell (FAC) sorting of beta cells and downstream analysis of CpG methylation by bisulphite and RNA sequencing, respectively. Animals deleted for the de novo methyltransferase, 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 and Cal-590. 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 data sets 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 sub-population specialised for insulin production, reminiscent of recently-described "ßHI" cells and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialization. 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 thus contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes.

17.
Diabetes ; 71(7): 1472-1489, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35472764

ABSTRACT

Mitochondrial glucose metabolism is essential for stimulated insulin release from pancreatic ß-cells. Whether mitofusin gene expression, and hence, mitochondrial network integrity, is important for glucose or incretin signaling has not previously been explored. Here, we generated mice with ß-cell-selective, adult-restricted deletion knock-out (dKO) of the mitofusin genes Mfn1 and Mfn2 (ßMfn1/2 dKO). ßMfn1/2-dKO mice displayed elevated fed and fasted glycemia and a more than fivefold decrease in plasma insulin. Mitochondrial length, glucose-induced polarization, ATP synthesis, and cytosolic and mitochondrial Ca2+ increases were all reduced in dKO islets. In contrast, oral glucose tolerance was more modestly affected in ßMfn1/2-dKO mice, and glucagon-like peptide 1 or glucose-dependent insulinotropic peptide receptor agonists largely corrected defective glucose-stimulated insulin secretion through enhanced EPAC-dependent signaling. Correspondingly, cAMP increases in the cytosol, as measured with an Epac-camps-based sensor, were exaggerated in dKO mice. Mitochondrial fusion and fission cycles are thus essential in the ß-cell to maintain normal glucose, but not incretin, sensing. These findings broaden our understanding of the roles of mitofusins in ß-cells, the potential contributions of altered mitochondrial dynamics to diabetes development, and the impact of incretins on this process.


Subject(s)
GTP Phosphohydrolases , Glucose , Incretins , Insulin-Secreting Cells , Animals , GTP Phosphohydrolases/genetics , Glucose/metabolism , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Incretins/metabolism , Incretins/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout
18.
Diabetes ; 71(7): 1525-1545, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35476777

ABSTRACT

Impaired pancreatic ß-cell function and insulin secretion are hallmarks of type 2 diabetes. miRNAs are short, noncoding RNAs that silence gene expression vital for the development and function of ß cells. We have previously shown that ß cell-specific deletion of the important energy sensor AMP-activated protein kinase (AMPK) results in increased miR-125b-5p levels. Nevertheless, the function of this miRNA in ß cells is unclear. We hypothesized that miR-125b-5p expression is regulated by glucose and that this miRNA mediates some of the deleterious effects of hyperglycemia in ß cells. Here, we show that islet miR-125b-5p expression is upregulated by glucose in an AMPK-dependent manner and that short-term miR-125b-5p overexpression impairs glucose-stimulated insulin secretion (GSIS) in the mouse insulinoma MIN6 cells and in human islets. An unbiased, high-throughput screen in MIN6 cells identified multiple miR-125b-5p targets, including the transporter of lysosomal hydrolases M6pr and the mitochondrial fission regulator Mtfp1. Inactivation of miR-125b-5p in the human ß-cell line EndoCß-H1 shortened mitochondria and enhanced GSIS, whereas mice overexpressing miR-125b-5p selectively in ß cells (MIR125B-Tg) were hyperglycemic and glucose intolerant. MIR125B-Tg ß cells contained enlarged lysosomal structures and had reduced insulin content and secretion. Collectively, we identify miR-125b as a glucose-controlled regulator of organelle dynamics that modulates insulin secretion.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , MicroRNAs , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Insulin-Secreting Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism
19.
Nat Commun ; 12(1): 5165, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34453049

ABSTRACT

Bariatric surgery improves both insulin sensitivity and secretion and can induce diabetes remission. However, the mechanisms and time courses of these changes, particularly the impact on ß cell function, are difficult to monitor directly. In this study, we investigated the effect of Vertical Sleeve Gastrectomy (VSG) on ß cell function in vivo by imaging Ca2+ dynamics in islets engrafted into the anterior eye chamber. Mirroring its clinical utility, VSG in mice results in significantly improved glucose tolerance, and enhanced insulin secretion. We reveal that these benefits are underpinned by augmented ß cell function and coordinated activity across the islet. These effects involve changes in circulating GLP-1 levels which may act both directly and indirectly on the ß cell, in the latter case through changes in body weight. Thus, bariatric surgery leads to time-dependent increases in ß cell function and intra-islet connectivity which are likely to contribute to diabetes remission.


Subject(s)
Calcium/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/surgery , Insulin-Secreting Cells/metabolism , Animals , Bariatric Surgery , Blood Glucose/metabolism , Diabetes Mellitus/diagnostic imaging , Female , Gastrectomy , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/metabolism , Intravital Microscopy , Male , Mice , Mice, Inbred C57BL , Stomach/surgery
20.
FEBS Lett ; 595(13): 1782-1796, 2021 07.
Article in English | MEDLINE | ID: mdl-33960419

ABSTRACT

Sorcin is a calcium-binding protein involved in maintaining endoplasmic reticulum (ER) Ca2+ stores. We have previously shown that overexpressing sorcin under the rat insulin promoter was protective against high-fat diet-induced pancreatic beta-cell dysfunction in vivo. Activating transcription factor 6 (ATF6) is a key mediator of the unfolded protein response (UPR) that provides cellular protection during the progression of ER stress. Here, using nonexcitable HEK293 cells, we show that sorcin overexpression increased ATF6 signalling, whereas sorcin knock out caused a reduction in ATF6 transcriptional activity and increased ER stress. Altogether, our data suggest that sorcin downregulation during lipotoxic stress may prevent full ATF6 activation and a normal UPR during the progression of obesity and insulin resistance.


Subject(s)
Activating Transcription Factor 6/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Gene Knockout Techniques/methods , Obesity/genetics , Palmitates/adverse effects , Animals , Calcium/metabolism , Cells, Cultured , Disease Progression , Down-Regulation , Endoplasmic Reticulum Stress/drug effects , Fibroblasts/cytology , Fibroblasts/metabolism , HEK293 Cells , Humans , Insulin Resistance , Mice , Obesity/metabolism , Signal Transduction , Transcriptional Activation/drug effects , Unfolded Protein Response/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL