Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 569
Filter
Add more filters

Publication year range
1.
N Engl J Med ; 389(21): 1972-1978, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37991855

ABSTRACT

Mahvash disease is an exceedingly rare genetic disorder of glucagon signaling characterized by hyperglucagonemia, hyperaminoacidemia, and pancreatic α-cell hyperplasia. Although there is no known definitive treatment, octreotide has been used to decrease systemic glucagon levels. We describe a woman who presented to our medical center after three episodes of small-volume hematemesis. She was found to have hyperglucagonemia and pancreatic hypertrophy with genetically confirmed Mahvash disease and also had evidence of portal hypertension (recurrent portosystemic encephalopathy and variceal hemorrhage) in the absence of cirrhosis. These findings established a diagnosis of portosinusoidal vascular disease, a presinusoidal type of portal hypertension previously known as noncirrhotic portal hypertension. Liver transplantation was followed by normalization of serum glucagon and ammonia levels, reversal of pancreatic hypertrophy, and resolution of recurrent encephalopathy and bleeding varices.


Subject(s)
Genetic Diseases, Inborn , Glucagon , Hypertension, Portal , Liver Transplantation , Female , Humans , Esophageal and Gastric Varices/etiology , Esophageal and Gastric Varices/surgery , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/surgery , Glucagon/blood , Glucagon/genetics , Hypertension, Portal/blood , Hypertension, Portal/etiology , Hypertension, Portal/genetics , Hypertension, Portal/surgery , Hypertrophy/genetics , Liver Cirrhosis , Genetic Diseases, Inborn/blood , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/surgery , Pancreatic Diseases/genetics , Pancreatic Diseases/pathology , Pancreatic Diseases/surgery , Glucagon-Secreting Cells/pathology
2.
Development ; 150(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36897571

ABSTRACT

Hormone secretion from pancreatic islets is essential for glucose homeostasis, and loss or dysfunction of islet cells is a hallmark of type 2 diabetes. Maf transcription factors are crucial for establishing and maintaining adult endocrine cell function. However, during pancreas development, MafB is not only expressed in insulin- and glucagon-producing cells, but also in Neurog3+ endocrine progenitor cells, suggesting additional functions in cell differentiation and islet formation. Here, we report that MafB deficiency impairs ß cell clustering and islet formation, but also coincides with loss of neurotransmitter and axon guidance receptor gene expression. Moreover, the observed loss of nicotinic receptor gene expression in human and mouse ß cells implied that signaling through these receptors contributes to islet cell migration/formation. Inhibition of nicotinic receptor activity resulted in reduced ß cell migration towards autonomic nerves and impaired ß cell clustering. These findings highlight a novel function of MafB in controlling neuronal-directed signaling events required for islet formation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Adult , Animals , Humans , Glucagon/genetics , Glucagon/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Pancreas/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/metabolism
3.
BMC Genomics ; 24(1): 202, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37069576

ABSTRACT

BACKGROUND: High throughput sequencing has enabled the interrogation of the transcriptomic landscape of glucagon-secreting alpha cells, insulin-secreting beta cells, and somatostatin-secreting delta cells. These approaches have furthered our understanding of expression patterns that define healthy or diseased islet cell types and helped explicate some of the intricacies between major islet cell crosstalk and glucose regulation. All three endocrine cell types derive from a common pancreatic progenitor, yet alpha and beta cells have partially opposing functions, and delta cells modulate and control insulin and glucagon release. While gene expression signatures that define and maintain cellular identity have been widely explored, the underlying epigenetic components are incompletely characterized and understood. However, chromatin accessibility and remodeling is a dynamic attribute that plays a critical role to determine and maintain cellular identity. RESULTS: Here, we compare and contrast the chromatin landscape between mouse alpha, beta, and delta cells using ATAC-Seq to evaluate the significant differences in chromatin accessibility. The similarities and differences in chromatin accessibility between these related islet endocrine cells help define their fate in support of their distinct functional roles. We identify patterns that suggest that both alpha and delta cells are poised, but repressed, from becoming beta-like. We also identify patterns in differentially enriched chromatin that have transcription factor motifs preferentially associated with different regions of the genome. Finally, we not only confirm and visualize previously discovered common endocrine- and cell specific- enhancer regions across differentially enriched chromatin, but identify novel regions as well. We compiled our chromatin accessibility data in a freely accessible database of common endocrine- and cell specific-enhancer regions that can be navigated with minimal bioinformatics expertise. CONCLUSIONS: Both alpha and delta cells appear poised, but repressed, from becoming beta cells in murine pancreatic islets. These data broadly support earlier findings on the plasticity in identity of non-beta cells under certain circumstances. Furthermore, differential chromatin accessibility shows preferentially enriched distal-intergenic regions in beta cells, when compared to either alpha or delta cells.


Subject(s)
Chromatin , Enhancer Elements, Genetic , Islets of Langerhans , Somatostatin-Secreting Cells , Animals , Mice , Chromatin/genetics , Chromatin/metabolism , Glucagon/genetics , Glucagon/metabolism , Islets of Langerhans/metabolism , Somatostatin-Secreting Cells/metabolism
4.
Biochem Biophys Res Commun ; 643: 121-128, 2023 02 05.
Article in English | MEDLINE | ID: mdl-36596263

ABSTRACT

Glucagon receptor plays an important role in the regulation of glucose metabolism. Studies have revealed that glucagon receptor antagonism is a potential effective treatment for diabetes. However, the functions of GCGR have not been fully illustrated. Although two Gcgr truncation knockout mice models have been widely used for GCGR function studies, truncated gene may remain neomorphic and/or dominant-negative function. In this study, we took the advantages of Crispr-Cas9 technique and generated a novel allele of GCGR in the mouse that yields complete loss of GCGR protein. Our studies reveal that complete deletion of Gcgr results in hyperglucagonemia, α-cell hyperplasia, improvement of glucose tolerance. These results are similar to the Gcgr-truncated mutation in mice. Hence, we provide a novel strain of GCGR knockout mice for the GCGR function studies.


Subject(s)
CRISPR-Cas Systems , Receptors, Glucagon , Animals , Mice , Receptors, Glucagon/genetics , Hyperplasia/genetics , Glucagon/genetics , Glucagon/metabolism , Mice, Knockout
5.
Physiol Rev ; 95(2): 513-48, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25834231

ABSTRACT

The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.


Subject(s)
Blood Glucose/metabolism , Energy Metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon/metabolism , Signal Transduction , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Enteroendocrine Cells/metabolism , Gene Expression Regulation , Glucagon/genetics , Glucagon-Like Peptide 1/genetics , Glucagon-Secreting Cells/metabolism , Homeostasis , Humans
6.
Int J Mol Sci ; 23(23)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36499023

ABSTRACT

Hypoglycemia, as a complication of type 2 diabetes (T2D), causes increased morbidity and mortality but the physiological response underlying hypoglycemia has not been fully elucidated. Small noncoding microRNA (miRNA) have multiple downstream biological effects. This pilot exploratory study was undertaken to determine if induced miRNA changes would persist and contribute to effects seen 24 h post-hypoglycemia. A parallel, prospective study design was employed, involving T2D (n = 23) and control (n = 23) subjects. The subjects underwent insulin-induced hypoglycemia (2 mmol/L; 36 mg/dL); blood samples were drawn at baseline, upon the induction of hypoglycemia, and 4 h and 24 h post-hypoglycemia, with a quantitative polymerase chain reaction analysis of miRNA undertaken. The baseline miRNAs did not differ. In the controls, 15 miRNAs were downregulated and one was upregulated (FDR < 0.05) from the induction of hypoglycemia to 4 h later while, in T2D, only four miRNAs were altered (downregulated), and these were common to both cohorts (miR-191-5p; miR-143-3p; let-7b-5p; let-7g-5p), correlated with elevated glucagon levels, and all were associated with energy balance. From the induction of hypoglycemia to 24 h, 14 miRNAs were downregulated and 5 were upregulated (FDR < 0.05) in the controls; 7 miRNAs were downregulated and 7 upregulated (FDR < 0.05) in T2D; a total of 6 miRNAs were common between cohorts, 5 were downregulated (miR-93-5p, let-7b-5p, miR-191-5p, miR-185-5p, and miR-652-3p), and 1 was upregulated (miR-369-3p). An ingenuity pathway analysis indicated that many of the altered miRNAs were associated with metabolic and coagulation pathways; however, of the inflammatory proteins expressed, only miR-143-3p at 24 h correlated positively with tumor necrosis factor-α (TNFa; p < 0.05 and r = 0.46) and negatively with toll-like receptor-4 (TLR4; p < 0.05 and r = 0.43). The MiRNA levels altered by hypoglycemia reflected changes in counter-regulatory glucagon and differed between cohorts, and their expression at 24 h suggests miRNAs may potentiate and prolong the physiological response. Trial registration: ClinicalTrials.gov NCT03102801.


Subject(s)
Diabetes Mellitus, Type 2 , MicroRNAs , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Gene Expression Profiling , Glucagon/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Prospective Studies
7.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G617-G626, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33533304

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is an incretin secreted from enteroendocrine preproglucagon (PPG)-expressing cells (traditionally known as L cells) in response to luminal nutrients that potentiates insulin secretion. Augmentation of endogenous GLP-1 secretion might well represent a novel therapeutic target for diabetes treatment in addition to the incretin-associated drugs currently in use. In this study, we found that PPG cells substantially express carbonic anhydrase 8 (CAR8), which has been reported to inhibit inositol 1,4,5-trisphosphate (IP3) binding to the IP3 receptor and subsequent Ca2+ efflux from the endoplasmic reticulum in neuronal cells. In vitro experiments using STC-1 cells demonstrated that Car8 knockdown increases long-chain fatty acid (LCFA)-stimulated GLP-1 secretion. This effect was reduced in the presence of phospholipase C (PLC) inhibitor; in addition, Car8 knockdown increased the intracellular Ca2+ elevation caused by α-linolenic acid, indicating that CAR8 exerts its effect on GLP-1 secretion via the PLC/IP3/Ca2+ pathway. Car8wdl null mutant mice showed significant increase in GLP-1 response to oral corn oil administration compared with that in wild-type littermates, with no significant change in intestinal GLP-1 content. These results demonstrate that CAR8 negatively regulates GLP-1 secretion from PPG cells in response to LCFAs, suggesting the possibility of augmentation of postprandial GLP-1 secretion by CAR8 inhibition.NEW & NOTEWORTHY This study focused on the physiological significance of carbonic anhydrase 8 (CAR8) in GLP-1 secretion from enteroendocrine preproglucagon (PPG)-expressing cells. We found an inhibitory role of CAR8 in LCFA-induced GLP-1 secretion in vitro and in vivo, suggesting a novel therapeutic approach to diabetes and obesity through augmentation of postprandial GLP-1 secretion by CAR8 inhibition.


Subject(s)
Biomarkers, Tumor/metabolism , Corn Oil/pharmacology , Enteroendocrine Cells/drug effects , Fatty Acids/pharmacology , Glucagon-Like Peptide 1/metabolism , Nerve Tissue Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Calcium Signaling , Cell Line , Enteroendocrine Cells/enzymology , Glucagon/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Secretory Pathway , Type C Phospholipases/metabolism
9.
Circ Res ; 124(1): 38-51, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30582457

ABSTRACT

RATIONALE: Glucagon is a key hormone that regulates the adaptive metabolic responses to fasting. In addition to maintaining glucose homeostasis, glucagon participates in the regulation of cholesterol metabolism; however, the molecular pathways underlying this effect are incompletely understood. OBJECTIVE: We sought to determine the role of hepatic Gcgr (glucagon receptor) signaling in plasma cholesterol regulation and identify its underlying molecular mechanisms. METHODS AND RESULTS: We show that Gcgr signaling plays an essential role in LDL-C (low-density lipoprotein cholesterol) homeostasis through regulating the PCSK9 (proprotein convertase subtilisin/kexin type 9) levels. Silencing of hepatic Gcgr or inhibition of glucagon action increased hepatic and plasma PCSK9 and resulted in lower LDLR (LDL receptor) protein and increased plasma LDL-C. Conversely, treatment of wild-type (WT) mice with glucagon lowered LDL-C levels, whereas this response was abrogated in Pcsk9-/- and Ldlr-/- mice. Our gain- and loss-of-function studies identified Epac2 (exchange protein activated by cAMP-2) and Rap1 (Ras-related protein-1) as the downstream mediators of glucagon's action on PCSK9 homeostasis. Moreover, mechanistic studies revealed that glucagon affected the half-life of PCSK9 protein without changing the level of its mRNA, indicating that Gcgr signaling regulates PCSK9 degradation. CONCLUSIONS: These findings provide novel insights into the molecular interplay between hepatic glucagon signaling and lipid metabolism and describe a new posttranscriptional mechanism of PCSK9 regulation.


Subject(s)
Cholesterol, LDL/blood , Energy Metabolism , Glucagon/metabolism , Liver/metabolism , Proprotein Convertase 9/metabolism , Animals , Cell Line , Enzyme Stability , Glucagon/deficiency , Glucagon/genetics , Guanine Nucleotide Exchange Factors/metabolism , Half-Life , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 9/deficiency , Proprotein Convertase 9/genetics , Proteolysis , Receptors, Glucagon/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism
10.
Gen Comp Endocrinol ; 314: 113925, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34624309

ABSTRACT

In contrast to mammals, glucagon is reported as a much more potent blood glucose modulator in birds. Interestingly, we have found p.Thr16Ser mutation, a variation in the highly conserved glucagon hormone, in Galliformes as well as Strigiformes. To check the effect of this mutation on the receptor binding of glucagon, we predicted the ancestral glucagon receptor sequence of all available Galliformes and Strigiformes species. Subsequently, we analysed their binding to the mutated and wild type glucagon (ancestral) by molecular dynamics simulation. At first, we made a model of ancestral glucagon receptor and ancestral mutated, and wild type glucagon in the order Galliformes and Strigiformes. Then we performed molecular dynamics for each Galliformes and Strigiformes receptor as well as each glucagon peptide, respectively. The final structures were used for docking simulation of glucagon to their receptors. The results of the docking simulations showed a stronger binding affinity of mutated glucagon to glucagon receptors. Afterward, we obtained blood glucose concentrations of all available Galliformes members, as well as all available members of its only taxonomic neighbour (order Anseriformes) in superorder Galloanserae. Interestingly the p.Thr16Ser mutation could finely cluster these two orders into two groups: higher blood glucose concentration (order Galliformes, 17.64 ± 1.66 mMol/L) and lower blood glucose concentration (order Anseriformes, 11.34 ± 1.11 mMol/L). Strigiformes which carry the mutated glucagon peptide show also high blood glucose concentrations (17.40 ± 1.51 mMol/L). Therefore, the results suggest this mutation, which leads to stronger binding affinity of mutated glucagon to its receptor, may be a driving force for higher blood glucose homeostasis in the related birds.


Subject(s)
Galliformes , Glucagon , Strigiformes , Animals , Blood Glucose , Computer Simulation , Glucagon/genetics , Homeostasis , Insulin
11.
Proc Natl Acad Sci U S A ; 115(17): E4111-E4119, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29555772

ABSTRACT

Liver zonation characterizes the separation of metabolic pathways along the lobules and is required for optimal function. Wnt/ß-catenin signaling controls metabolic zonation by activating genes in the perivenous hepatocytes, while suppressing genes in the periportal counterparts. We now demonstrate that glucagon opposes the actions of Wnt/ß-catenin signaling on gene expression and metabolic zonation pattern. The effects were more pronounced in the periportal hepatocytes where 28% of all genes were activated by glucagon and inhibited by Wnt/ß-catenin. The glucagon and Wnt/ß-catenin receptors and their signaling pathways are uniformly distributed in periportal and perivenous hepatocytes and the expression is not regulated by the opposing signal. Collectively, our results show that glucagon controls gene expression and metabolic zonation in the liver through a counterplay with the Wnt/ß-catenin signaling pathway.


Subject(s)
Gene Expression Regulation/physiology , Glucagon/metabolism , Hepatocytes/metabolism , Liver/metabolism , Wnt Signaling Pathway/physiology , Animals , Glucagon/genetics , Mice , Mice, Knockout
12.
Biochem Biophys Res Commun ; 532(1): 47-53, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32826056

ABSTRACT

Although diabetic polyneuropathy (DPN) is the commonest diabetic complication, its pathology remains to be clarified. As previous papers have suggested the neuroprotective effects of glucagon-like peptide-1 in DPN, the current study investigated the physiological indispensability of glucagon gene-derived peptides (GCGDPs) including glucagon-like peptide-1 in the peripheral nervous system (PNS). Neurological functions and neuropathological changes of GCGDP deficient (gcg-/-) mice were examined. The gcg-/- mice showed tactile allodynia and thermal hyperalgesia at 12-18 weeks old, followed by tactile and thermal hypoalgesia at 36 weeks old. Nerve conduction studies revealed a decrease in sensory nerve conduction velocity at 36 weeks old. Pathological findings showed a decrease in intraepidermal nerve fiber densities. Electron microscopy revealed a decrease in circularity and an increase in g-ratio of myelinated fibers and a decrease of unmyelinated fibers in the sural nerves of the gcg-/- mice. Effects of glucagon on neurite outgrowth were examined using an ex vivo culture of dorsal root ganglia. A supraphysiological concentration of glucagon promoted neurite outgrowth. In conclusion, the mice with deficiency of GCGDPs developed peripheral neuropathy with age. Furthermore, glucagon might have neuroprotective effects on the PNS of mice. GCGDPs might be involved in the pathology of DPN.


Subject(s)
Diabetic Neuropathies/etiology , Glucagon-Like Peptides/deficiency , Animals , Diabetic Neuropathies/genetics , Diabetic Neuropathies/pathology , Disease Models, Animal , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Glucagon/deficiency , Glucagon/genetics , Glucagon/metabolism , Glucagon-Like Peptide 1/deficiency , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptides/genetics , Glucagon-Like Peptides/metabolism , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers, Myelinated/pathology , Neural Conduction , Neuronal Outgrowth , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
13.
Protein Expr Purif ; 170: 105590, 2020 06.
Article in English | MEDLINE | ID: mdl-32007557

ABSTRACT

N-terminal extensions ("tags") have proven valuable for producing peptides using high throughput recombinant expression technologies. However, the applicability is hampered by the limited options for specific and efficient proteases to release the fully native sequence without additional amino acids in the N-terminal. Here we describe the Escherichia coli (E. coli) expression, purification and characterization of engineered variants of Xaa-Pro dipeptidyl aminopeptidase (Xaa-Pro-DAP) derived from Lactococcus lactis for cleavage of Gly-Pro dipeptide extension in the N-terminal of glucagon and glucagon-like peptide 1 (GLP-1(7-37)). By single amino acid substitution in the Xaa-Pro-DAP protease, significantly higher product yields were achieved. The combination of HRV14 3C protease and engineered Xaa-Pro-DAP is suggested for obtaining native N-terminal of peptides.


Subject(s)
Bacterial Proteins/genetics , Dipeptidases/genetics , Glucagon-Like Peptide 1/genetics , Glucagon/genetics , Lactococcus lactis/enzymology , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cloning, Molecular , Dipeptidases/chemistry , Dipeptidases/metabolism , Enzyme Assays , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucagon/chemistry , Glucagon/metabolism , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Humans , Kinetics , Lactococcus lactis/genetics , Mutagenesis, Site-Directed , Protein Engineering/methods , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Biosci Biotechnol Biochem ; 84(5): 936-942, 2020 May.
Article in English | MEDLINE | ID: mdl-31916916

ABSTRACT

Endocrine cells in the gastrointestinal tract secrete multiple hormones to maintain homeostasis in the body. In the present study, we generated intestinal organoids from the duodenum, jejunum, and ileum of Neurogenin 3 (Ngn3)-EGFP mice and examined how enteroendocrine cells (EECs) within organoid cultures resemble native epithelial cells in the gut. Transcriptome analysis of EGFP-positive cells from Ngn3-EGFP organoids showed gene expression pattern comparable to EECs in vivo. We also compared mRNAs of five major hormones, namely, ghrelin (Ghrl), cholecystokinin (Cck), Gip, secretin (Sct), and glucagon (Gcg) in organoids and small intestine along the longitudinal axis and found that expression patterns of these hormones in organoids were similar to those in native tissues. These findings suggest that an intestinal organoid culture system can be utilized as a suitable model to study enteroendocrine cell functions in vitro.


Subject(s)
Duodenum/cytology , Enteroendocrine Cells/metabolism , Ileum/cytology , Jejunum/cytology , Organoids/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Cholecystokinin/genetics , Cholecystokinin/metabolism , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Ghrelin/genetics , Ghrelin/metabolism , Glucagon/genetics , Glucagon/metabolism , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Secretin/genetics , Secretin/metabolism , Signal Transduction , Transcriptome
15.
Proc Natl Acad Sci U S A ; 114(10): 2747-2752, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28143927

ABSTRACT

Genetic disruption or pharmacologic inhibition of glucagon signaling effectively lowers blood glucose but results in compensatory glucagon hypersecretion involving expansion of pancreatic α-cell mass. Ben-Zvi et al. recently reported that angiopoietin-like protein 4 (Angptl4) links glucagon receptor inhibition to hyperglucagonemia and α-cell proliferation [Ben-Zvi et al. (2015) Proc Natl Acad Sci USA 112:15498-15503]. Angptl4 is a secreted protein and inhibitor of lipoprotein lipase-mediated plasma triglyceride clearance. We report that Angptl4-/- mice treated with an anti-glucagon receptor monoclonal antibody undergo elevation of plasma glucagon levels and α-cell expansion similar to wild-type mice. Overexpression of Angptl4 in liver of mice caused a 8.6-fold elevation in plasma triglyceride levels, but did not alter plasma glucagon levels or α-cell mass. Furthermore, administration of glucagon receptor-blocking antibody to healthy individuals increased plasma glucagon and amino acid levels, but did not change circulating Angptl4 concentration. These data show that Angptl4 does not link glucagon receptor inhibition to compensatory hyperglucagonemia or expansion of α-cell mass, and that it cannot be given to induce such secretion and growth. The reduction of plasma triglyceride levels in Angptl4-/- mice and increase following Angptl4 overexpression suggest that changes in plasma triglyceride metabolism do not regulate α-cells in the pancreas. Our findings corroborate recent data showing that increased plasma amino acids and their transport into α-cells link glucagon receptor blockage to α-cell hyperplasia.


Subject(s)
Angiopoietin-Like Protein 4/genetics , Glucagon/metabolism , Hyperplasia/genetics , Lipid Metabolism/genetics , Animals , Blood Glucose/genetics , Cell Proliferation/genetics , Glucagon/genetics , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Humans , Hyperplasia/blood , Hyperplasia/pathology , Lipoproteins/blood , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Receptors, Glucagon/genetics , Signal Transduction/genetics , Triglycerides/blood
16.
Proc Natl Acad Sci U S A ; 114(10): 2753-2758, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28115707

ABSTRACT

Inactivating mutations in the insulin receptor results in extreme insulin resistance. The resulting hyperglycemia is very difficult to treat, and patients are at risk for early morbidity and mortality from complications of diabetes. We used the insulin receptor antagonist S961 to induce severe insulin resistance, hyperglycemia, and ketonemia in mice. Using this model, we show that glucagon receptor (GCGR) inhibition with a monoclonal antibody normalized blood glucose and ß-hydroxybutyrate levels. Insulin receptor antagonism increased pancreatic ß-cell mass threefold. Normalization of blood glucose levels with GCGR-blocking antibody unexpectedly doubled ß-cell mass relative to that observed with S961 alone and 5.8-fold over control. GCGR antibody blockage expanded α-cell mass 5.7-fold, and S961 had no additional effects. Collectively, these data show that GCGR antibody inhibition represents a potential therapeutic option for treatment of patients with extreme insulin-resistance syndromes.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Glucagon/metabolism , Insulin Resistance/genetics , Receptor, Insulin/genetics , Receptors, Glucagon/genetics , 3-Hydroxybutyric Acid/metabolism , Animals , Blood Glucose/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucagon/genetics , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/pathology , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Ketosis/genetics , Ketosis/metabolism , Ketosis/pathology , Mice , Mutation , Peptides/pharmacology , Receptors, Glucagon/antagonists & inhibitors
17.
Am J Physiol Endocrinol Metab ; 316(3): E397-E409, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30620636

ABSTRACT

The LIM-homeodomain (LIM-HD) transcription factor Islet-1 (Isl1) interacts with the LIM domain-binding protein 1 (Ldb1) coregulator to control expression of key pancreatic ß-cell genes. However, Ldb1 also has Isl1-independent effects, supporting that another LIM-HD factor interacts with Ldb1 to impact ß-cell development and/or function. LIM homeobox 1 (Lhx1) is an Isl1-related LIM-HD transcription factor that appears to be expressed in the developing mouse pancreas and in adult islets. However, roles for this factor in the pancreas are unknown. This study aimed to determine Lhx1 interactions and elucidate gene regulatory and physiological roles in the pancreas. Co-immunoprecipitation using ß-cell extracts demonstrated an interaction between Lhx1 and Isl1, and thus we hypothesized that Lhx1 and Isl1 regulate similar target genes. To test this, we employed siRNA-mediated Lhx1 knockdown in ß-cell lines and discovered reduced Glp1R mRNA. Chromatin immunoprecipitation revealed Lhx1 occupancy at a domain also known to be occupied by Isl1 and Ldb1. Through development of a pancreas-wide knockout mouse model ( Lhx1∆Panc), we demonstrate that aged Lhx1∆Panc mice have elevated fasting blood glucose levels, altered intraperitoneal and oral glucose tolerance, and significantly upregulated glucagon, somatostatin, pancreatic polypeptide, MafB, and Arx islet mRNAs. Additionally, Lhx1∆Panc mice exhibit significantly reduced Glp1R, an mRNA encoding the insulinotropic receptor for glucagon-like peptide 1 along with a concomitant dampened Glp1 response and mild glucose intolerance in mice challenged with oral glucose. These data are the first to reveal that the Lhx1 transcription factor contributes to normal glucose homeostasis and Glp1 responses.


Subject(s)
Blood Glucose/metabolism , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Glucagon/genetics , Glucagon-Like Peptide-1 Receptor/genetics , Homeodomain Proteins/genetics , Homeostasis , Insulin-Secreting Cells/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , MafB Transcription Factor/genetics , Mice , Mice, Knockout , Pancreatic Polypeptide/genetics , RNA, Messenger/metabolism , Somatostatin/genetics , Transcription Factors/genetics , Up-Regulation
18.
Am J Transplant ; 19(6): 1852-1858, 2019 06.
Article in English | MEDLINE | ID: mdl-30801971

ABSTRACT

Islet cell transplantation is curative therapy for patients with complicated autoimmune type 1 diabetes (T1D). We report the diagnostic potential of circulating transplant islet-specific exosomes to noninvasively distinguish pancreatic ß cell injury secondary to recurrent autoimmunity vs immunologic rejection. A T1D patient with hypoglycemic unawareness underwent islet transplantation and maintained normoglycemia until posttransplant day 1098 before requiring exogenous insulin. Plasma analysis showed decreased donor islet exosome quantities on day 1001, before hyperglycemia onset. This drop in islet exosome quantity signified islet injury, but did not distinguish injury type. However, analysis of purified transplant islet exosome cargoes showed decrease in insulin-containing exosomes, but not glucagon-containing exosomes, indicating selective destruction of transplanted ß cells secondary to recurrent T1D autoimmunity. Furthermore, donor islet exosome cargo analysis showed time-specific increase in islet autoantigen, glutamic acid decarboxylase 65 (GAD65), implicated in T1D autoimmunity. Time-matched analysis of plasma transplant islet exosomes in 3 control subjects undergoing islet cell transplantation failed to show changes in islet exosome quantities or intraexosomal cargo expression of insulin, glucagon, and GAD65. This is the first report of noninvasive diagnosis of recurrent autoimmunity after islet cell transplantation, suggesting that transplant tissue exosome platform may serve as a biomarker in islet transplant diagnostics.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Exosomes/genetics , Exosomes/metabolism , Glucagon/genetics , Glucagon/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recurrence , Retrospective Studies
19.
Biochem Biophys Res Commun ; 517(4): 629-635, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31387746

ABSTRACT

Type II diabetes is a complex, chronic, and progressive disease. Previously, we demonstrate that FXR inhibits GLP-1 secretion via interacting with CREB to inhibit the transcriptional activity of CREB, thus promoting the development of type II diabetes. Epigenetic modifications, such as DNA methylation, histone acetylation, and post-transcriptional RNA regulation, are essential mediators contributing to diabetes-associated morbidity and mortality. Thus, we attempted to investigate the epigenetic mechanisms of FXR modulating GLP-1 secretion. Firstly, the involvement of histone acetylation, DNA methylation, and post-transcriptional regulation in FXR inhibiting GLP-1 secretion was verified. As FXR overexpression significantly inhibited the activity of GCG 3'-UTR, we hypothesize that miRNA might participate in the mechanism. Two online tools and real-time PCR revealed that FXR promoted miR-33 expression. Moreover, miR-33 inhibited the expression of GCG and CREB1 through direct targeting in STC-1 cells. FXR overexpression in STC-1 cells significantly reduced the mRNA expression and protein levels of both GCG and CREB1, as well as the secretion of GLP-1; miR-33 inhibition exerted opposing effects. More importantly, the effects of FXR overexpression were significantly reversed by miR-33 inhibition, indicating that FXR inhibited GLP-1 secretion through promoting miR-33 expression, therefore inhibiting the expression of miR-33 targets, GCG and CREB1. In conclusion, we provide a novel epigenetic mechanism by which FXR inhibits the secretion of GLP-1 through miR-33 and its two downstream targets, GCG and CREB1. These findings might provide innovative strategies for improving type II diabetes, which needs further in vivo and clinical investigation.


Subject(s)
Epigenesis, Genetic , Glucagon-Like Peptide 1/metabolism , MicroRNAs/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Glucagon/genetics , Glucagon/metabolism , Mice , MicroRNAs/genetics , Transcription, Genetic
20.
Cell Tissue Res ; 375(2): 359-369, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30259122

ABSTRACT

We use a monoclonal antibody against the C-terminal of oxyntomodulin (OXM) to investigate enteroendocrine cells (EEC) in mouse, rat, human and pig. This antibody has cross-reactivity with the OXM precursor, glicentin (Gli) but does not recognise glucagon. The antibody stained EEC in the jejunum and colon of each species. We compared OXM/Gli immunoreactivity with that revealed by antibodies against structurally related peptides, GLP-1 and glucagon and against GIP and PYY that are predicted to be in some EEC that express OXM/Gli. We used super-resolution to locate immunoreactive vesicles. In the pancreas, OXM/Gli was in glucagon cells but was located in separate storage vesicles to glucagon. In jejunal EEC, OXM/Gli and GIP were in many of the same cells but often in separate vesicles, whereas PYY and OXM/Gli were commonly colocalised in the same storage vesicles of colonic EEC. When binding of anti-GLP-1 to the structurally related GIP was removed by absorption with GIP peptide, GLP-1 and OXM/Gli immunoreactivities were contained in the same population of EEC in the intestine. We conclude that anti-OXM/Gli is a more reliable marker than anti-GLP-1 for EEC expressing preproglucagon products. Storage vesicles that were immunoreactive for OXM/Gli were almost always immunoreactive for GLP-1. OXM concentrations, measured by ELISA, were highest in the distal ileum and colon. Lesser concentrations were found in more proximal parts of small intestine and pancreas. Very little was in the stomach. In EEC containing GIP and OXM/Gli, these hormones are packaged in different secretory vesicles. Separate packaging also occurred for OXM and glucagon, whereas OXM/Gli and PYY and OXM/Gli and GLP-1 were commonly contained together in secretory vesicles.


Subject(s)
Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Oxyntomodulin/metabolism , Amino Acid Sequence , Animals , Antibodies/metabolism , Colon/metabolism , Female , Glucagon/chemistry , Glucagon/genetics , Glucagon/metabolism , Humans , Jejunum/metabolism , Male , Mice, Inbred C57BL , Organ Specificity , Oxyntomodulin/chemistry , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Transport , Rats , Species Specificity , Subcellular Fractions , Swine
SELECTION OF CITATIONS
SEARCH DETAIL