Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Physiol Biochem ; 52(4): 879-892, 2019.
Article in English | MEDLINE | ID: mdl-30958662

ABSTRACT

BACKGROUND/AIMS: CXCL14, a secreted chemokine peptide that promotes obesity-induced insulin resistance, is expressed by islets, but its effects on islet function are unknown. The aim of this study was to determine the role of CXCL14 in ß-cells and investigate how it transduces these effects. METHODS: Cxcl14 and Cxc-receptor mRNA expression was quantified by qPCR and CXCL14 expression in the pancreas was determined by immunohistochemistry. The putative function of CXCL14 at CXCR4 and CXCR7 receptors was determined by ß-arrestin recruitment assays. The effects of CXCL14 on glucose-stimulated insulin secretion, cAMP production, glucose-6-phosphate accumulation, ATP generation, apoptosis and proliferation were determined using standard techniques. RESULTS: CXCL14 was present in mouse islets, where it was mainly localised to islet δ-cells. Cxc-receptor mRNA profiling indicated that Cxcr4 and Cxcr7 are the most abundant family members in islets, but CXCL14 did not promote ß-arrestin recruitment at CXCR4 or CXCR7 or antagonise CXCL12 activation of these receptors. CXCL14 induced a concentration-dependent inhibition of glucose-stimulated insulin secretion, which was not coupled to Gαi signalling. However, CXCL14 inhibited glucose-6-phosphate generation and ATP production in mouse islets. CONCLUSION: CXCL14 is expressed by islet δ-cells where it may have paracrine effects to inhibit insulin secretion in a CXCR4/CXCR7-independent manner through reductions in ß-cell ATP levels. These observations, together with the previously reported association of CXCL14 with obesity and impaired glucose homeostasis, suggest that inhibition of CXCL14 signalling could be explored to treat type 2 diabetes.


Subject(s)
Chemokines, CXC/metabolism , Cyclic AMP/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Chemokines, CXC/genetics , Cyclic AMP/genetics , Insulin/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Paracrine Communication , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Second Messenger Systems
2.
Cell Mol Life Sci ; 75(16): 3039-3050, 2018 08.
Article in English | MEDLINE | ID: mdl-29455414

ABSTRACT

INTRODUCTION: Islets synthesise and secrete numerous peptides, some of which are known to be important regulators of islet function and glucose homeostasis. In this study, we quantified mRNAs encoding all peptide ligands of islet G protein-coupled receptors (GPCRs) in isolated human and mouse islets and carried out in vitro islet hormone secretion studies to provide functional confirmation for the species-specific role of peptide YY (PYY) in mouse islets. MATERIALS AND METHODS: GPCR peptide ligand mRNAs in human and mouse islets were quantified by quantitative real-time PCR relative to the reference genes ACTB, GAPDH, PPIA, TBP and TFRC. The pathways connecting GPCR peptide ligands with their receptors were identified by manual searches in the PubMed, IUPHAR and Ingenuity databases. Distribution of PYY protein in mouse and human islets was determined by immunohistochemistry. Insulin, glucagon and somatostatin secretion from islets was measured by radioimmunoassay. RESULTS: We have quantified GPCR peptide ligand mRNA expression in human and mouse islets and created specific signalomes mapping the pathways by which islet peptide ligands regulate human and mouse GPCR signalling. We also identified species-specific islet expression of several GPCR ligands. In particular, PYY mRNA levels were ~ 40,000-fold higher in mouse than human islets, suggesting a more important role of locally secreted Pyy in mouse islets. This was confirmed by IHC and functional experiments measuring insulin, glucagon and somatostatin secretion. DISCUSSION: The detailed human and mouse islet GPCR peptide ligand atlases will allow accurate translation of mouse islet functional studies for the identification of GPCR/peptide signalling pathways relevant for human physiology, which may lead to novel treatment modalities of diabetes and metabolic disease.


Subject(s)
Islets of Langerhans/metabolism , Peptide YY/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Gene Expression , Humans , Immunohistochemistry , Ligands , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Peptide YY/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
Cell Physiol Biochem ; 45(2): 656-666, 2018.
Article in English | MEDLINE | ID: mdl-29408822

ABSTRACT

BACKGROUND/AIMS: CRISPR-Cas9, a RNA-guided targeted genome editing tool, has revolutionized genetic engineering by offering the ability to precisely modify DNA. GPRC5B is an orphan receptor belonging to the group C family of G protein-coupled receptors (GPCRs). In this study, we analysed the functional roles of the Gprc5b receptor in MIN6 ß-cells using CRISPR-Cas9 and transient over-expression of Gprc5b. METHODS: The optimal transfection reagent for use in MIN6 ß-cells was determined by analysing efficiency of GFP plasmid delivery by cell sorting. A MIN6 ß-cell line in which Gprc5b expression was knocked down (Gprc5b KD) was generated using CRISPR-Cas9 technology. Gprc5b receptor mRNA expression, proliferation, apoptosis, Cignal 45-Pathway Reporter Array signalling and western blot assays were carried out using Gpcr5b KD MIN6 ß-cells that had been transiently transfected with different concentrations of mouse Gprc5b plasmid to over-express Gprc5b. RESULTS: JetPRIME® was the best candidate for MIN6 ß-cell transfection, providing approximately 30% transfection efficiency. CRISPR-Cas9 technology targeting Gprc5b led to stable knock-down of this receptor in MIN6 ß-cells and its re-expression induced proliferation and potentiated cytokine- and palmitate-induced apoptosis. The Cignal 45 Reporter analysis indicated Gprc5b-dependent regulation of apoptotic and proliferative pathways, and western blotting confirmed activation of signalling via TGF-ß and IFNγ. CONCLUSION: This study provides evidence of CRISPR-Cas9 technology being used to down-regulate Gprc5b expression in MIN6 ß-cells. This strategy allowed us to identify signalling pathways linking GPRC5B receptor expression to ß-cell proliferation and apoptosis.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Interferon-gamma/metabolism , Mice , Neuropeptides/metabolism , Palmitic Acid/toxicity , Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
Cell Mol Life Sci ; 75(4): 715-726, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28921001

ABSTRACT

AIMS: Complement components 3 and 5 (C3 and C5) play essential roles in the complement system, generating C3a and C5a peptides that are best known as chemotactic and inflammatory factors. In this study we characterised islet expression of C3 and C5 complement components, and the impact of C3aR and C5aR1 activation on islet function and viability. MATERIALS AND METHODS: Human and mouse islet mRNAs encoding key elements of the complement system were quantified by qPCR and distribution of C3 and C5 proteins was determined by immunohistochemistry. Activation of C3aR and C5aR1 was determined using DiscoverX beta-arrestin assays. Insulin secretion from human and mouse islets was measured by radioimmunoassay, and intracellular calcium ([Ca2+]i), ATP generation and apoptosis were assessed by standard techniques. RESULTS: C3 and C5 proteins and C3aR and C5aR1 were expressed by human and mouse islets, and C3 and C5 were mainly localised to ß- and α-cells. Conditioned media from islets exposed for 1 h to 5.5 and 20 mM glucose stimulated C3aR and C5aR1-driven beta-arrestin recruitment. Activation of C3aR and C5aR1 potentiated glucose-induced insulin secretion from human and mouse islets, increased [Ca2+]i and ATP generation, and protected islets against apoptosis induced by a pro-apoptotic cytokine cocktail or palmitate. CONCLUSIONS: Our observations demonstrate a functional link between activation of components of the innate immune system and improved ß-cell function, suggesting that low-level chronic inflammation may improve glucose homeostasis through direct effects on ß-cells.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Complement C3/metabolism , Complement C5/metabolism , Cytokines/metabolism , Glucose/metabolism , Humans , Male , Mice , Mice, Inbred ICR , beta-Arrestins/metabolism
5.
Sci Rep ; 7: 46600, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28422162

ABSTRACT

G-protein coupled receptors (GPCRs) are essential for islet function, but most studies use rodent islets due to limited human islet availability. We have systematically compared the GPCR mRNA expression in human and mouse islets to determine to what extent mouse islets can be used as surrogates for human islets to study islet GPCR function, and we have identified species-specific expression of several GPCRs. The A3 receptor (ADORA3) was expressed only in mouse islets and the A3 agonist MRS 5698 inhibited glucose-induced insulin secretion from mouse islets, with no effect on human islets. Similarly, mRNAs encoding the galanin receptors GAL1 (GALR1), GAL2 (GALR2) and GAL3 GALR3) were abundantly expressed in mouse islets but present only at low levels in human islets, so that it reads (GALR3) and galanin inhibited insulin secretion only from mouse islets. Conversely, the sst1 receptor (SSTR1) was abundant only in human islets and its selective activation by CH 275 inhibited insulin secretion from human islets, with no effect on mouse islets. Our comprehensive human and mouse islet GPCR atlas has demonstrated that species differences do exist in islet GPCR expression and function, which are likely to impact on the translatability of mouse studies to the human context.


Subject(s)
Gene Expression Regulation , Insulin Secretion , Insulin/metabolism , Islets of Langerhans/metabolism , Receptor, Adenosine A3/metabolism , Receptors, Galanin/biosynthesis , Receptors, Somatostatin/biosynthesis , Animals , Humans , Islets of Langerhans/cytology , Male , Mice , Species Specificity
6.
Endocr J ; 64(3): 325-338, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28228611

ABSTRACT

Pancreatic islets express high levels of the orphan G-protein coupled receptor C5C (GPRC5C), the function of which remains to be established. Here we have examined the role of GPRC5C in the regulation of insulin secretion and ß-cell survival and proliferation using human and mouse pancreatic islets. The expression of GPRC5C was analysed by RNA-sequencing, qPCR, western blotting and confocal microscopy. Insulin secretion and cell viability were determined by RIA and MTS assays, respectively. GPRC5C mRNA expression and protein level were reduced in the islets from type-2 diabetic donors. RNA sequencing in human islets revealed GPRC5C expression correlated with the expression of genes controlling apoptosis, cell survival and proliferation. A reduction in Gprc5c mRNA and protein expression was observed in islets isolated from old mice (>46 weeks of age) compared to that in islets from newborn (<3 weeks) mice. Down-regulation of Gprc5c led to both moderately reduced glucose-stimulated insulin release and also reduced cAMP content in mouse islets. Potentiation of glucose-stimulated insulin secretion concomitant with enhanced islet cAMP level by all-trans retinoic acid (ATRA) was attenuated upon Gprc5c-KD. ATRA also increased [Ca+2]i in Huh7-cells. Gprc5c over expression in Huh7 cells was associated with increased ERK1/2 activity. Gprc5c-KD in clonal MIN6c4 cells reduced cell proliferation and in murine islets increased apoptosis and the sensitivity of primary islet cells to a cocktail of pro-apoptotic cytokines. Our results demonstrate that agents activating GPRC5C represent a novel modality for the treatment and/or prevention of diabetes by restoring and/or maintaining functional ß-cell mass.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, G-Protein-Coupled/agonists , Tretinoin/metabolism , Aging , Animals , Animals, Newborn , Apoptosis , Calcium Signaling , Cell Line , Diabetes Mellitus, Type 2/pathology , Female , Gene Expression Regulation , Genes, Reporter , Humans , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Tissue Culture Techniques
7.
Diabetes ; 65(1): 129-39, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26470781

ABSTRACT

We have previously demonstrated that coculture of islets with mesenchymal stromal cells (MSCs) enhanced islet insulin secretory capacity in vitro, correlating with improved graft function in vivo. To identify factors that contribute to MSC-mediated improvements in islet function, we have used an unbiased quantitative RT-PCR screening approach to identify MSC-derived peptide ligands of G-protein-coupled receptors that are expressed by islets cells. We demonstrated high expression of annexin A1 (ANXA1) mRNA by MSCs and confirmed expression at the protein level in lysates and MSC-conditioned media by Western blot analysis and ELISA. Preculturing islets with exogenous ANXA1 enhanced glucose-stimulated insulin secretion (GSIS), thereby mimicking the beneficial influence of MSC preculture in vitro. Small interfering RNA-mediated knockdown of ANXA1 in MSCs reduced their capacity to potentiate GSIS. MSCs derived from ANXA1(-/-) mice had no functional capacity to enhance GSIS, in contrast to wild-type controls. Preculturing islets with ANXA1 had modest effects on their capacity to regulate blood glucose in streptozotocin-induced diabetic mice, indicating that additional MSC-derived factors are required to fully mimic the beneficial effects of MSC preculture in vivo. These findings demonstrate the feasibility of harnessing the MSC secretome as a defined, noncellular strategy to improve the efficiency of clinical islet transplantation protocols.


Subject(s)
Annexin A1/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/genetics , Insulin/metabolism , Islets of Langerhans/metabolism , Mesenchymal Stem Cells/metabolism , RNA, Messenger/metabolism , Animals , Annexin A1/metabolism , Blotting, Western , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , In Vitro Techniques , Insulin Secretion , Mesenchymal Stem Cell Transplantation , Mice , RNA, Small Interfering , Real-Time Polymerase Chain Reaction
8.
Pharmacol Ther ; 146: 61-93, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25242198

ABSTRACT

G-protein coupled receptors (GPCRs) are involved in the regulation of adipose tissue function, but the total number of GPCRs expressed by human subcutaneous adipose tissue, as well as their function and interactions with drugs, is poorly understood. We have constructed an atlas of all GPCRs expressed by human subcutaneous adipose tissue: the 'adipose tissue GPCRome', to support the exploration of novel control nodes in metabolic and endocrine functions. This atlas describes how adipose tissue GPCRs regulate lipolysis, insulin resistance and adiponectin and leptin secretion. We also discuss how adipose tissue GPCRs interact with their endogenous ligands and with GPCR-targeting drugs, with a focus on how drug/receptor interactions may affect lipolysis, and present a model predicting how GPCRs with unknown effects on lipolysis might modulate cAMP-regulated lipolysis. Subcutaneous adipose tissue expresses 163 GPCRs, a majority of which have unknown effects on lipolysis, insulin resistance and adiponectin and leptin secretion. These GPCRs are activated by 180 different endogenous ligands, and are the targets of a large number of clinically used drugs. We identified 119 drugs, acting on 23 GPCRs, that are predicted to stimulate lipolysis and 173 drugs, acting on 25 GPCRs, that are predicted to inhibit lipolysis. This atlas highlights knowledge gaps in the current understanding of adipose tissue GPCR function, and identifies GPCR/ligand/drug interactions that might affect lipolysis, which is important for understanding and predicting metabolic side effects of drugs. This approach may aid in the design of new, safer therapeutic agents, with fewer undesired effects on lipid homeostasis.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Subcutaneous Fat/metabolism , Adiponectin/metabolism , Animals , Humans , Insulin Resistance , Leptin/metabolism , Ligands , Lipolysis
9.
Can Vet J ; 49(9): 889-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19043486

ABSTRACT

A mixed-breed beef cow was presented with swelling of the front and hind left quarters of the mammary gland and mild depression. Direct examination and culture of the serosanguinous-like milk samples collected from these quarters were consistent with Bacillus anthracis infection.


Subject(s)
Bacillus anthracis/isolation & purification , Mastitis, Bovine/microbiology , Animals , Bacillus anthracis/pathogenicity , Cattle , Disease Outbreaks/veterinary , Fatal Outcome , Female , Mastitis, Bovine/diagnosis , Mastitis, Bovine/epidemiology , Saskatchewan/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...