101
|
El K, Capozzi ME, Campbell JE. Repositioning the Alpha Cell in Postprandial Metabolism. Endocrinology 2020; 161:5910252. [PMID: 32964214 PMCID: PMC7899437 DOI: 10.1210/endocr/bqaa169] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022]
Abstract
Glucose homeostasis is maintained in large part due to the actions of the pancreatic islet hormones insulin and glucagon, secreted from β- and α-cells, respectively. The historical narrative positions these hormones in opposition, with insulin primarily responsible for glucose-lowering and glucagon-driving elevations in glucose. Recent progress in this area has revealed a more complex relationship between insulin and glucagon, highlighted by data demonstrating that α-cell input is essential for β-cell function and glucose homeostasis. Moreover, the common perception that glucagon levels decrease following a nutrient challenge is largely shaped by the inhibitory effects of glucose administration alone on the α-cell. Largely overlooked is that a mixed nutrient challenge, which is more representative of typical human feeding, actually stimulates glucagon secretion. Thus, postprandial metabolism is associated with elevations, not decreases, in α-cell activity. This review discusses the recent advances in our understanding of how α-cells regulate metabolism, with a particular focus on the postprandial state. We highlight α- to β-cell communication, a term that describes how α-cell input into β-cells is a critical axis that regulates insulin secretion and glucose homeostasis. Finally, we discuss the open questions that have the potential to advance this field and continue to evolve our understanding of the role that α-cells play in postprandial metabolism.
Collapse
Affiliation(s)
- Kimberley El
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
- Department of Medicine, Division of Endocrinology, Duke University, Durham, North Carolina
- Correspondence: Jonathan E. Campbell, 300 N Duke Street, Durham, North Carolina 27701. E-mail:
| |
Collapse
|
102
|
Pedersen JS, Rygg MO, Kristiansen VB, Olsen BH, Serizawa RR, Holst JJ, Madsbad S, Gluud LL, Bendtsen F, Wewer Albrechtsen NJ. Nonalcoholic Fatty Liver Disease Impairs the Liver-Alpha Cell Axis Independent of Hepatic Inflammation and Fibrosis. Hepatol Commun 2020; 4:1610-1623. [PMID: 33163832 PMCID: PMC7603528 DOI: 10.1002/hep4.1562] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/22/2020] [Accepted: 05/31/2020] [Indexed: 01/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is associated with impaired hepatic actions of glucagon and insulin. Glucagon and amino acids are linked in an endocrine feedback circuit, the liver-alpha cell axis, that may be disrupted by NAFLD. We investigated how NAFLD severity affects glucagon and insulin resistance in individuals with obesity and whether bariatric surgery improves these parameters. Plasma and liver biopsies from 33 individuals with obesity (collectively, OBE) were obtained before and 12 months after bariatric surgery (Roux-en-Y gastric bypass [RYGB] or sleeve gastrectomy [SG]). Nine healthy control individuals (collectively, CON) undergoing cholecystectomy were used as a comparison group. The NAFLD activity score (NAS) was used to subdivide study participants into the following groups: OBE-no steatosis, OBE+steatosis, and nonalcoholic steatohepatitis (NASH) and/or grade 2 fibrosis (Fib) (OBE-NASH-Fib). Measurements of amino acids by targeted metabolomics and glucagon were performed. Glucagon, amino acids (P < 0.05), and the glucagon-alanine index, a validated surrogate marker of glucagon resistance, were increased in OBE by 60%, 56%, and 61%, respectively, when compared with CON but irrespective of NAFLD severity. In contrast, markers of hepatic insulin resistance increased concomitantly with NAS. Hyperglucagonemia resolved in OBE-no steatosis and OBE+steatosis but not in OBE-NASH-Fib (median, 7.0; interquartile range, 5.0-9.8 pmol/L), regardless of improvement in insulin resistance and NAS. The type of surgery that participants underwent had no effect on metabolic outcomes. Conclusion: Glucagon resistance to amino acid metabolism exists in individuals with NAFLD independent of NAS severity. Patients with NASH showed persistent hyperglucagonemia 12 months after bariatric surgery, indicating that a disrupted liver-alpha cell may remain in NAFLD despite major improvement in liver histology.
Collapse
Affiliation(s)
- Julie Steen Pedersen
- GastrounitMedical DivisionCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Marte Opseth Rygg
- GastrounitMedical DivisionCopenhagen University Hospital HvidovreHvidovreDenmark
| | | | - Beth Hærstedt Olsen
- Department of Nuclear Medicine and Functional Imaging, Ultrasound SectionCopenhagen University Hospital HvidovreHvidovreDenmark
| | | | - Jens Juul Holst
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Sten Madsbad
- Department of EndocrinologyCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Lise Lotte Gluud
- GastrounitMedical DivisionCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Flemming Bendtsen
- GastrounitMedical DivisionCopenhagen University Hospital HvidovreHvidovreDenmark
| | - Nicolai Jacob Wewer Albrechtsen
- Department of Biomedical SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Biochemistry, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
103
|
Smith DK, Kates L, Durinck S, Patel N, Stawiski EW, Kljavin N, Foreman O, Sipos B, Solloway MJ, Allan BB, Peterson AS. Elevated Serum Amino Acids Induce a Subpopulation of Alpha Cells to Initiate Pancreatic Neuroendocrine Tumor Formation. CELL REPORTS MEDICINE 2020; 1:100058. [PMID: 33205067 PMCID: PMC7659536 DOI: 10.1016/j.xcrm.2020.100058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/06/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022]
Abstract
The cellular origin of sporadic pancreatic neuroendocrine tumors (PNETs) is obscure. Hormone expression suggests that these tumors arise from glucagon-producing alpha cells or insulin-producing β cells, but instability in hormone expression prevents linage determination. We utilize loss of hepatic glucagon receptor (GCGR) signaling to drive alpha cell hyperproliferation and tumor formation to identify a cell of origin and dissect mechanisms that drive progression. Using a combination of genetically engineered Gcgr knockout mice and GCGR-inhibiting antibodies, we show that elevated plasma amino acids drive the appearance of a proliferative population of SLC38A5+ embryonic progenitor-like alpha cells in mice. Further, we characterize tumors from patients with rare bi-allelic germline GCGR loss-of-function variants and find prominent tumor-cell-associated expression of the SLC38A5 paralog SLC7A8 as well as markers of active mTOR signaling. Thus, progenitor cells arise from adult alpha cells in response to metabolic signals and, when inductive signals are chronically present, drive tumor initiation. GCGR inhibition induces an SLC38A5+ alpha cell population in aged mice An SLC38A5+ alpha cell subpopulation initiates pancreatic tumors in aged Gcgr−/− mice Tumors exhibit low mutational burden and response to mTOR inhibition by rapamycin Tumors in GCGR loss-of-function models lack immune cell infiltration
Collapse
Affiliation(s)
- Derek K Smith
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lance Kates
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Steffen Durinck
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nisha Patel
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Eric W Stawiski
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Noelyn Kljavin
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Oded Foreman
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bence Sipos
- University Hospital Tübingen, Internal Medicine VIII, Tübingen 72076, Germany
| | - Mark J Solloway
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Bernard B Allan
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | |
Collapse
|
104
|
Wang Z, Cui W. Two Sides of Electrospun Fiber in Promoting and Inhibiting Biomedical Processes. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Zhen Wang
- Shanghai Institute of Traumatology and Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| | - Wenguo Cui
- Shanghai Institute of Traumatology and Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases Ruijin Hospital Shanghai Jiao Tong University School of Medicine 197 Ruijin 2nd Road Shanghai 200025 P. R. China
| |
Collapse
|
105
|
Suppli MP, Bagger JI, Lund A, Demant M, van Hall G, Strandberg C, Kønig MJ, Rigbolt K, Langhoff JL, Wewer Albrechtsen NJ, Holst JJ, Vilsbøll T, Knop FK. Glucagon Resistance at the Level of Amino Acid Turnover in Obese Subjects With Hepatic Steatosis. Diabetes 2020; 69:1090-1099. [PMID: 31974144 DOI: 10.2337/db19-0715] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Glucagon secretion is regulated by circulating glucose, but it has turned out that amino acids also play an important role and that hepatic amino acid metabolism and glucagon are linked in a mutual feedback cycle, the liver-α-cell axis. On the basis of this knowledge, we hypothesized that hepatic steatosis might impair glucagon's action on hepatic amino acid metabolism and lead to hyperaminoacidemia and hyperglucagonemia. We subjected 15 healthy lean and 15 obese steatotic male participants to a pancreatic clamp with somatostatin and evaluated hepatic glucose and amino acid metabolism when glucagon was at basal levels and at high physiological levels. The degree of steatosis was evaluated from liver biopsy specimens. Total RNA sequencing of liver biopsy specimens from the obese steatotic individuals revealed perturbations in the expression of genes predominantly involved in amino acid metabolism. This group was characterized by fasting hyperglucagonemia, hyperaminoacidemia, and no lowering of amino acid levels in response to high levels of glucagon. Endogenous glucose production was similar between lean and obese individuals. Our results suggest that hepatic steatosis causes resistance to the effect of glucagon on amino acid metabolism. This results in increased amino acid concentrations and increased glucagon secretion, providing a likely explanation for fatty liver-associated hyperglucagonemia.
Collapse
Affiliation(s)
- Malte P Suppli
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Jonatan I Bagger
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Asger Lund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Mia Demant
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Gerrit van Hall
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Clinical Metabolomics Core Facility, Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Strandberg
- Department of Radiology, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Merete J Kønig
- Department of Radiology, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | | | - Jill L Langhoff
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
106
|
Tellez K, Hang Y, Gu X, Chang CA, Stein RW, Kim SK. In vivo studies of glucagon secretion by human islets transplanted in mice. Nat Metab 2020; 2:547-557. [PMID: 32694729 PMCID: PMC7739959 DOI: 10.1038/s42255-020-0213-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/24/2020] [Indexed: 02/06/2023]
Abstract
Little is known about regulated glucagon secretion by human islet α-cells compared to insulin secretion from β-cells, despite conclusive evidence of dysfunction in both cell types in diabetes mellitus. Distinct insulins in humans and mice permit in vivo studies of human β-cell regulation after human islet transplantation in immunocompromised mice, whereas identical glucagon sequences prevent analogous in vivo measures of glucagon output from human α-cells. Here, we use CRISPR-Cas9 editing to remove glucagon codons 2-29 in immunocompromised NSG mice, preserving the production of other proglucagon-derived hormones. Glucagon knockout NSG (GKO-NSG) mice have metabolic, liver and pancreatic phenotypes associated with glucagon-signalling deficits that revert after transplantation of human islets from non-diabetic donors. Glucagon hypersecretion by transplanted islets from donors with type 2 diabetes revealed islet-intrinsic defects. We suggest that GKO-NSG mice provide an unprecedented resource to investigate human α-cell regulation in vivo.
Collapse
Affiliation(s)
- Krissie Tellez
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Charles A Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Roland W Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine (Endocrinology Division), Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
107
|
Galsgaard KD, Jepsen SL, Kjeldsen SAS, Pedersen J, Wewer Albrechtsen NJ, Holst JJ. Alanine, arginine, cysteine, and proline, but not glutamine, are substrates for, and acute mediators of, the liver-α-cell axis in female mice. Am J Physiol Endocrinol Metab 2020; 318:E920-E929. [PMID: 32255678 DOI: 10.1152/ajpendo.00459.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this study was to identify the amino acids that stimulate glucagon secretion in mice and whose metabolism depends on glucagon receptor signaling. Pancreata of female C57BL/6JRj mice were perfused with 19 individual amino acids and pyruvate (at 10 mM), and secretion of glucagon was assessed using a specific glucagon radioimmunoassay. Separately, a glucagon receptor antagonist (GRA; 25-2648, 100 mg/kg) or vehicle was administered to female C57BL/6JRj mice 3 h before an intraperitoneal injection of four different isomolar amino acid mixtures (in total 7 µmol/g body wt) as follows: mixture 1 contained alanine, arginine, cysteine, and proline; mixture 2 contained aspartate, glutamate, histidine, and lysine; mixture 3 contained citrulline, methionine, serine, and threonine; and mixture 4 contained glutamine, leucine, isoleucine, and valine. Blood glucose, plasma glucagon, amino acid, and insulin concentrations were measured using well-characterized methodologies. Alanine (P = 0.03), arginine (P < 0.0001), cysteine (P = 0.01), glycine (P = 0.02), lysine (P = 0.02), and proline (P = 0.03), but not glutamine (P = 0.9), stimulated glucagon secretion from the perfused mouse pancreas. However, when the four isomolar amino acid mixtures were administered in vivo, the four mixtures elicited similar glucagon responses (P > 0.5). Plasma concentrations of total amino acids in vivo were higher after administration of GRA when mixture 1 (P = 0.004) or mixture 3 (P = 0.04) were injected. Our data suggest that alanine, arginine, cysteine, and proline, but not glutamine, are involved in the acute regulation of the liver-α-cell axis in female mice, as they all increased glucagon secretion and their disappearance rate was altered by GRA.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara L Jepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sasha A S Kjeldsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Nephrology and Endocrinology, Nordsjaellands Hospital Hilleroed, University of Copenhagen, Hilleroed, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
108
|
Amino acid and lipid metabolism in post-gestational diabetes and progression to type 2 diabetes: A metabolic profiling study. PLoS Med 2020; 17:e1003112. [PMID: 32433647 PMCID: PMC7239388 DOI: 10.1371/journal.pmed.1003112] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/20/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Women with a history of gestational diabetes mellitus (GDM) have a 7-fold higher risk of developing type 2 diabetes (T2D) during midlife and an elevated risk of developing hypertension and cardiovascular disease. Glucose tolerance reclassification after delivery is recommended, but fewer than 40% of women with GDM are tested. Thus, improved risk stratification methods are needed, as is a deeper understanding of the pathology underlying the transition from GDM to T2D. We hypothesize that metabolites during the early postpartum period accurately distinguish risk of progression from GDM to T2D and that metabolite changes signify underlying pathophysiology for future disease development. METHODS AND FINDINGS The study utilized fasting plasma samples collected from a well-characterized prospective research study of 1,035 women diagnosed with GDM. The cohort included racially/ethnically diverse pregnant women (aged 20-45 years-33% primiparous, 37% biparous, 30% multiparous) who delivered at Kaiser Permanente Northern California hospitals from 2008 to 2011. Participants attended in-person research visits including 2-hour 75-g oral glucose tolerance tests (OGTTs) at study baseline (6-9 weeks postpartum) and annually thereafter for 2 years, and we retrieved diabetes diagnoses from electronic medical records for 8 years. In a nested case-control study design, we collected fasting plasma samples among women without diabetes at baseline (n = 1,010) to measure metabolites among those who later progressed to incident T2D or did not develop T2D (non-T2D). We studied 173 incident T2D cases and 485 controls (pair-matched on BMI, age, and race/ethnicity) to discover metabolites associated with new onset of T2D. Up to 2 years post-baseline, we analyzed samples from 98 T2D cases with 239 controls to reveal T2D-associated metabolic changes. The longitudinal analysis tracked metabolic changes within individuals from baseline to 2 years of follow-up as the trajectory of T2D progression. By building prediction models, we discovered a distinct metabolic signature in the early postpartum period that predicted future T2D with a median discriminating power area under the receiver operating characteristic curve of 0.883 (95% CI 0.820-0.945, p < 0.001). At baseline, the most striking finding was an overall increase in amino acids (AAs) as well as diacyl-glycerophospholipids and a decrease in sphingolipids and acyl-alkyl-glycerophospholipids among women with incident T2D. Pathway analysis revealed up-regulated AA metabolism, arginine/proline metabolism, and branched-chain AA (BCAA) metabolism at baseline. At follow-up after the onset of T2D, up-regulation of AAs and down-regulation of sphingolipids and acyl-alkyl-glycerophospholipids were sustained or strengthened. Notably, longitudinal analyses revealed only 10 metabolites associated with progression to T2D, implicating AA and phospholipid metabolism. A study limitation is that all of the analyses were performed with the same cohort. It would be ideal to validate our findings in an independent longitudinal cohort of women with GDM who had glucose tolerance tested during the early postpartum period. CONCLUSIONS In this study, we discovered a metabolic signature predicting the transition from GDM to T2D in the early postpartum period that was superior to clinical parameters (fasting plasma glucose, 2-hour plasma glucose). The findings suggest that metabolic dysregulation, particularly AA dysmetabolism, is present years prior to diabetes onset, and is revealed during the early postpartum period, preceding progression to T2D, among women with GDM. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01967030.
Collapse
|
109
|
Galsgaard KD, Pedersen J, Kjeldsen SAS, Winther-Sørensen M, Stojanovska E, Vilstrup H, Ørskov C, Wewer Albrechtsen NJ, Holst JJ. Glucagon receptor signaling is not required for N-carbamoyl glutamate- and l-citrulline-induced ureagenesis in mice. Am J Physiol Gastrointest Liver Physiol 2020; 318:G912-G927. [PMID: 32174131 DOI: 10.1152/ajpgi.00294.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon regulates the hepatic amino acid metabolism and increases ureagenesis. Ureagenesis is activated by N-acetylglutamate (NAG), formed via activation of N-acetylglutamate synthase (NAGS). With the aim to identify the steps whereby glucagon both acutely and chronically regulates ureagenesis, we investigated whether glucagon receptor-mediated activation of ureagenesis is required in a situation where NAGS activity and/or NAG levels are sufficient to activate the first step of the urea cycle in vivo. Female C57BL/6JRj mice treated with a glucagon receptor antagonist (GRA), glucagon receptor knockout (Gcgr-/-) mice, and wild-type (Gcgr+/+) littermates received an intraperitoneal injection of N-carbamoyl glutamate (Car; a stable variant of NAG), l-citrulline (Cit), Car and Cit (Car + Cit), or PBS. In separate experiments, Gcgr-/- and Gcgr+/+ mice were administered N-carbamoyl glutamate and l-citrulline (wCar + wCit) in the drinking water for 8 wk. Car, Cit, and Car + Cit significantly (P < 0.05) increased plasma urea concentrations, independently of pharmacological and genetic disruption of glucagon receptor signaling (P = 0.9). Car increased blood glucose concentrations equally in GRA- and vehicle-treated mice (P = 0.9), whereas the increase upon Car + Cit was impaired in GRA-treated mice (P = 0.008). Blood glucose concentrations remained unchanged in Gcgr-/- mice upon Car (P = 0.2) and Car + Cit (P = 0.9). Eight weeks administration of wCar + wCit did not change blood glucose (P > 0.2), plasma amino acid (P > 0.4), and urea concentrations (P > 0.3) or the area of glucagon-positive cells (P > 0.3) in Gcgr-/- and Gcgr+/+ mice. Our data suggest that glucagon-mediated activation of ureagenesis is not required when NAGS activity and/or NAG levels are sufficient to activate the first step of the urea cycle.NEW & NOTEWORTHY Hepatic ureagenesis is essential in amino acid metabolism and is importantly regulated by glucagon, but the exact mechanism is unclear. With the aim to identify the steps whereby glucagon both acutely and chronically regulates ureagenesis, we here show, contrary to our hypothesis, that glucagon receptor-mediated activation of ureagenesis is not required when N-acetylglutamate synthase activity and/or N-acetylglutamate levels are sufficient to activate the first step of the urea cycle in vivo.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology and Nephrology, Nordsjaellands Hospital Hilleroed, Hilleroed, Denmark
| | - Sasha A S Kjeldsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Winther-Sørensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Elena Stojanovska
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hendrik Vilstrup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Cathrine Ørskov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
110
|
Abstract
Glucagon and its partner insulin are dually linked in both their secretion from islet cells and their action in the liver. Glucagon signaling increases hepatic glucose output, and hyperglucagonemia is partly responsible for the hyperglycemia in diabetes, making glucagon an attractive target for therapeutic intervention. Interrupting glucagon signaling lowers blood glucose but also results in hyperglucagonemia and α-cell hyperplasia. Investigation of the mechanism for α-cell proliferation led to the description of a conserved liver-α-cell axis where glucagon is a critical regulator of amino acid homeostasis. In return, amino acids regulate α-cell function and proliferation. New evidence suggests that dysfunction of the axis in humans may result in the hyperglucagonemia observed in diabetes. This discussion outlines important but often overlooked roles for glucagon that extend beyond glycemia and supports a new role for α-cells as amino acid sensors.
Collapse
Affiliation(s)
- E Danielle Dean
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
111
|
Finan B, Capozzi ME, Campbell JE. Repositioning Glucagon Action in the Physiology and Pharmacology of Diabetes. Diabetes 2020; 69:532-541. [PMID: 31178432 PMCID: PMC7085250 DOI: 10.2337/dbi19-0004] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/30/2019] [Indexed: 01/03/2023]
Abstract
Glucagon is historically described as the counterregulatory hormone to insulin, induced by fasting/hypoglycemia to raise blood glucose through action mediated in the liver. However, it is becoming clear that the biology of glucagon is much more complex and extends beyond hepatic actions to exert control on glucose metabolism. We discuss the inconsistencies with the canonical view that glucagon is primarily a hyperglycemic agent driven by fasting/hypoglycemia and highlight the recent advances that have reshaped the metabolic role of glucagon. These concepts are placed within the context of both normal physiology and the pathophysiology of disease and then extended to discuss emerging strategies that incorporate glucagon agonism in the pharmacology of treating diabetes.
Collapse
Affiliation(s)
- Brian Finan
- Novo Nordisk Research Center, Indianapolis, IN
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, NC
- Division of Endocrinology, Department of Medicine, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| |
Collapse
|
112
|
Hartig SM, Cox AR. Paracrine signaling in islet function and survival. J Mol Med (Berl) 2020; 98:451-467. [PMID: 32067063 DOI: 10.1007/s00109-020-01887-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/05/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023]
Abstract
The pancreatic islet is a dense cellular network comprised of several cell types with endocrine function vital in the control of glucose homeostasis, metabolism, and feeding behavior. Within the islet, endocrine hormones also form an intricate paracrine network with supportive cells (endothelial, neuronal, immune) and secondary signaling molecules regulating cellular function and survival. Modulation of these signals has potential consequences for diabetes development, progression, and therapeutic intervention. Beta cell loss, reduced endogenous insulin secretion, and dysregulated glucagon secretion are hallmark features of both type 1 and 2 diabetes that not only impact systemic regulation of glucose, but also contribute to the function and survival of cells within the islet. Advancing research and technology have revealed new islet biology (cellular identity and transcriptomes) and identified previously unrecognized paracrine signals and mechanisms (somatostatin and ghrelin paracrine actions), while shifting prior views of intraislet communication. This review will summarize the paracrine signals regulating islet endocrine function and survival, the disruption and dysfunction that occur in diabetes, and potential therapeutic targets to preserve beta cell mass and function.
Collapse
Affiliation(s)
- Sean M Hartig
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Aaron R Cox
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
113
|
Global Transcriptomic Analysis of Zebrafish Glucagon Receptor Mutant Reveals Its Regulated Metabolic Network. Int J Mol Sci 2020; 21:ijms21030724. [PMID: 31979106 PMCID: PMC7037442 DOI: 10.3390/ijms21030724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/23/2019] [Accepted: 01/20/2020] [Indexed: 12/18/2022] Open
Abstract
The glucagon receptor (GCGR) is a G-protein-coupled receptor (GPCR) that mediates the activity of glucagon. Disruption of GCGR results in many metabolic alterations, including increased glucose tolerance, decreased adiposity, hypoglycemia, and pancreatic α-cell hyperplasia. To better understand the global transcriptomic changes resulting from GCGR deficiency, we performed whole-organism RNA sequencing analysis in wild type and gcgr-deficient zebrafish. We found that the expression of 1645 genes changes more than two-fold among mutants. Most of these genes are related to metabolism of carbohydrates, lipids, and amino acids. Genes related to fatty acid β-oxidation, amino acid catabolism, and ureagenesis are often downregulated. Among gcrgr-deficient zebrafish, we experimentally confirmed increases in lipid accumulation in the liver and whole-body glucose uptake, as well as a modest decrease in total amino acid content. These results provide new information about the global metabolic network that GCGR signaling regulates in addition to a better understanding of the receptor’s physiological functions.
Collapse
|
114
|
Grubelnik V, Markovič R, Lipovšek S, Leitinger G, Gosak M, Dolenšek J, Valladolid-Acebes I, Berggren PO, Stožer A, Perc M, Marhl M. Modelling of dysregulated glucagon secretion in type 2 diabetes by considering mitochondrial alterations in pancreatic α-cells. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191171. [PMID: 32218947 PMCID: PMC7029933 DOI: 10.1098/rsos.191171] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/16/2019] [Indexed: 05/15/2023]
Abstract
Type 2 diabetes mellitus (T2DM) has been associated with insulin resistance and the failure of β-cells to produce and secrete enough insulin as the disease progresses. However, clinical treatments based solely on insulin secretion and action have had limited success. The focus is therefore shifting towards α-cells, in particular to the dysregulated secretion of glucagon. Our qualitative electron-microscopy-based observations gave an indication that mitochondria in α-cells are altered in Western-diet-induced T2DM. In particular, α-cells extracted from mouse pancreatic tissue showed a lower density of mitochondria, a less expressed matrix and a lower number of cristae. These deformities in mitochondrial ultrastructure imply a decreased efficiency in mitochondrial ATP production, which prompted us to theoretically explore and clarify one of the most challenging problems associated with T2DM, namely the lack of glucagon secretion in hypoglycaemia and its oversecretion at high blood glucose concentrations. To this purpose, we constructed a novel computational model that links α-cell metabolism with their electrical activity and glucagon secretion. Our results show that defective mitochondrial metabolism in α-cells can account for dysregulated glucagon secretion in T2DM, thus improving our understanding of T2DM pathophysiology and indicating possibilities for new clinical treatments.
Collapse
Affiliation(s)
- Vladimir Grubelnik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia
| | - Rene Markovič
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
| | - Saška Lipovšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Marko Gosak
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, 171 76 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, 171 76 Stockholm, Sweden
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Complexity Science Hub Vienna, 1080 Vienna, Austria
- Authors for correspondence: Matjač Perc e-mail:
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Education, University of Maribor, 2000 Maribor, Slovenia
- Authors for correspondence: Marko Marhl e-mail:
| |
Collapse
|
115
|
Godoy-Matos AF, Silva Júnior WS, Valerio CM. NAFLD as a continuum: from obesity to metabolic syndrome and diabetes. Diabetol Metab Syndr 2020; 12:60. [PMID: 32684985 PMCID: PMC7359287 DOI: 10.1186/s13098-020-00570-y] [Citation(s) in RCA: 325] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The prevalence of non-alcoholic fatty liver disease (NAFLD) has been increasing rapidly. It is nowadays recognized as the most frequent liver disease, affecting a quarter of global population and regularly coexisting with metabolic disorders such as type 2 diabetes, hypertension, obesity, and cardiovascular disease. In a more simplistic view, NAFLD could be defined as an increase in liver fat content, in the absence of secondary cause of steatosis. In fact, the clinical onset of the disease is a much more complex process, closely related to insulin resistance, limited expandability and dysfunctionality of adipose tissue. A fatty liver is a main driver for a new recognized liver-pancreatic α-cell axis and increased glucagon, contributing to diabetes pathophysiology. MAIN TEXT This review will focus on the clinical and pathophysiological connections between NAFLD, insulin resistance and type 2 diabetes. We reviewed non-invasive methods and several scoring systems for estimative of steatosis and fibrosis, proposing a multistep process for NAFLD evaluation. We will also discuss treatment options with a more comprehensive view, focusing on the current available therapies for obesity and/or type 2 diabetes that impact each stage of NAFLD. CONCLUSION The proper understanding of NAFLD spectrum-as a continuum from obesity to metabolic syndrome and diabetes-may contribute to the early identification and for establishment of targeted treatment.
Collapse
Affiliation(s)
- Amélio F. Godoy-Matos
- Metabolism Department, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Pontifical Catholic University of Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ CEP 20211-340 Brazil
| | - Wellington S. Silva Júnior
- Endocrinology Discipline, Faculty of Medicine, Center of Natural, Human, Health, and Technology Sciences, Federal University of Maranhão (UFMA), Pinheiro, MA CEP 65200-000 Brazil
| | - Cynthia M. Valerio
- Metabolism Department, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Pontifical Catholic University of Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ CEP 20211-340 Brazil
| |
Collapse
|
116
|
Pettus JH, D'Alessio D, Frias JP, Vajda EG, Pipkin JD, Rosenstock J, Williamson G, Zangmeister MA, Zhi L, Marschke KB. Efficacy and Safety of the Glucagon Receptor Antagonist RVT-1502 in Type 2 Diabetes Uncontrolled on Metformin Monotherapy: A 12-Week Dose-Ranging Study. Diabetes Care 2020; 43:161-168. [PMID: 31694861 DOI: 10.2337/dc19-1328] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/15/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Evaluate the safety and efficacy of RVT-1502, a novel oral glucagon receptor antagonist, in subjects with type 2 diabetes inadequately controlled on metformin. RESEARCH DESIGN AND METHODS In a phase 2, double-blind, randomized, placebo-controlled study, subjects with type 2 diabetes (n = 166) on a stable dose of metformin were randomized (1:1:1:1) to placebo or RVT-1502 5, 10, or 15 mg once daily for 12 weeks. The primary end point was change from baseline in HbA1c for each dose of RVT-1502 compared with placebo. Secondary end points included change from baseline in fasting plasma glucose (FPG) and safety assessments. RESULTS Over 12 weeks, RVT-1502 significantly reduced HbA1c relative to placebo by 0.74%, 0.76%, and 1.05% in the 5-, 10-, and 15-mg groups (P < 0.001), respectively, and FPG decreased by 2.1, 2.2, and 2.6 mmol/L (P < 0.001). The proportions of subjects achieving an HbA1c <7.0% were 19.5%, 39.5%, 39.5%, and 45.0% with placebo and RVT-1502 5, 10, and 15 mg (P ≤ 0.02 vs. placebo). The frequency of hypoglycemia was low, and no episodes were severe. Mild increases in mean aminotransferase levels remaining below the upper limit of normal were observed with RVT-1502 but were reversible and did not appear to be dose related, with no other liver parameter changes. Weight and lipid changes were similar between RVT-1502 and placebo. RVT-1502-associated mild increases in blood pressure were not dose related or consistent across time. CONCLUSIONS Glucagon receptor antagonism with RVT-1502 significantly lowers HbA1c and FPG, with a safety profile that supports further clinical development with longer-duration studies (NCT02851849).
Collapse
Affiliation(s)
| | | | | | - Eric G Vajda
- Ligand Pharmaceuticals Incorporated, San Diego, CA
| | | | | | | | | | - Lin Zhi
- Ligand Pharmaceuticals Incorporated, San Diego, CA
| | | |
Collapse
|
117
|
Grubelnik V, Markovič R, Lipovšek S, Leitinger G, Gosak M, Dolenšek J, Valladolid-Acebes I, Berggren PO, Stožer A, Perc M, Marhl M. Modelling of dysregulated glucagon secretion in type 2 diabetes by considering mitochondrial alterations in pancreatic α-cells. ROYAL SOCIETY OPEN SCIENCE 2020. [PMID: 32218947 DOI: 10.5061/dryad.9n2k1vk] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) has been associated with insulin resistance and the failure of β-cells to produce and secrete enough insulin as the disease progresses. However, clinical treatments based solely on insulin secretion and action have had limited success. The focus is therefore shifting towards α-cells, in particular to the dysregulated secretion of glucagon. Our qualitative electron-microscopy-based observations gave an indication that mitochondria in α-cells are altered in Western-diet-induced T2DM. In particular, α-cells extracted from mouse pancreatic tissue showed a lower density of mitochondria, a less expressed matrix and a lower number of cristae. These deformities in mitochondrial ultrastructure imply a decreased efficiency in mitochondrial ATP production, which prompted us to theoretically explore and clarify one of the most challenging problems associated with T2DM, namely the lack of glucagon secretion in hypoglycaemia and its oversecretion at high blood glucose concentrations. To this purpose, we constructed a novel computational model that links α-cell metabolism with their electrical activity and glucagon secretion. Our results show that defective mitochondrial metabolism in α-cells can account for dysregulated glucagon secretion in T2DM, thus improving our understanding of T2DM pathophysiology and indicating possibilities for new clinical treatments.
Collapse
Affiliation(s)
- Vladimir Grubelnik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia
| | - Rene Markovič
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
| | - Saška Lipovšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, 2000 Maribor, Slovenia
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Marko Gosak
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Ismael Valladolid-Acebes
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, 171 76 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, 171 76 Stockholm, Sweden
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Complexity Science Hub Vienna, 1080 Vienna, Austria
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Education, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
118
|
Zhao L, Wang L, Aierken R, Wang W, Wang X, Li M. Characterization of Insulin and Glucagon Genes and Their Producing Endocrine Cells From Pygmy Sperm Whale ( Kogia breviceps). Front Endocrinol (Lausanne) 2020; 11:174. [PMID: 32296396 PMCID: PMC7137828 DOI: 10.3389/fendo.2020.00174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/11/2020] [Indexed: 02/05/2023] Open
Abstract
Insulin and glucagon are hormones secreted by pancreatic β and α cells, respectively, which together regulate glucose homeostasis. Dysregulation of insulin or glucagon can result in loss of blood glucose control, characterized by hyperglycemia or hypoglycemia. To better understand the endocrine physiology of cetaceans, we cloned and characterized the insulin and glucagon genes from pygmy sperm whale (Kogia breviceps). We obtained the complete coding sequences of the preproinsulin and preproglucagon genes, which encodes the preproinsulin protein of 110 amino acid (aa) residues and encodes the preproglucagon protein of 179 aa residues, respectively. Sequence comparison and phylogenetic analyses demonstrate that protein structures were similar to other mammalian orthologs. Immunohistochemistry and immunofluorescence staining using insulin, glucagon, and somatostatin antibodies allowed analysis of pygmy sperm whale islet distribution, architecture, and composition. Our results showed the pygmy sperm whale islet was irregularly shaped and randomly distributed throughout the pancreas. The architecture of α, β, and δ cells of the pygmy sperm whale was similar to that of artiodactyls species. This is the first report about insulin and glucagon genes in cetaceans, which provides new information about the structural conservation of the insulin and glucagon genes. Furthermore, offers novel information on the properties of endocrine cells in cetacean for further studies.
Collapse
Affiliation(s)
- Liyuan Zhao
- Laboratory of Marine Biology and Ecology, Fujian Provincial Key Laboratory of Marine Ecological Conservation and Restoration, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Likun Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Reyilamu Aierken
- Laboratory of Marine Biology and Ecology, Fujian Provincial Key Laboratory of Marine Ecological Conservation and Restoration, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Wei Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xianyan Wang
- Laboratory of Marine Biology and Ecology, Fujian Provincial Key Laboratory of Marine Ecological Conservation and Restoration, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
- *Correspondence: Xianyan Wang
| | - Mingyu Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
- Mingyu Li
| |
Collapse
|
119
|
Noguchi GM, Huising MO. Integrating the inputs that shape pancreatic islet hormone release. Nat Metab 2019; 1:1189-1201. [PMID: 32694675 PMCID: PMC7378277 DOI: 10.1038/s42255-019-0148-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
The pancreatic islet is a complex mini organ composed of a variety of endocrine cells and their support cells, which together tightly control blood glucose homeostasis. Changes in glucose concentration are commonly regarded as the chief signal controlling insulin-secreting beta cells, glucagon-secreting alpha cells and somatostatin-secreting delta cells. However, each of these cell types is highly responsive to a multitude of endocrine, paracrine, nutritional and neural inputs, which collectively shape the final endocrine output of the islet. Here, we review the principal inputs for each islet-cell type and the physiological circumstances in which these signals arise, through the prism of the insights generated by the transcriptomes of each of the major endocrine-cell types. A comprehensive integration of the factors that influence blood glucose homeostasis is essential to successfully improve therapeutic strategies for better diabetes management.
Collapse
Affiliation(s)
- Glyn M Noguchi
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA.
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
120
|
Douros JD, Tong J, D’Alessio DA. The Effects of Bariatric Surgery on Islet Function, Insulin Secretion, and Glucose Control. Endocr Rev 2019; 40:1394-1423. [PMID: 31241742 PMCID: PMC6749890 DOI: 10.1210/er.2018-00183] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 04/23/2019] [Indexed: 01/19/2023]
Abstract
Although bariatric surgery was developed primarily to treat morbid obesity, evidence from the earliest clinical observations to the most recent clinical trials consistently demonstrates that these procedures have substantial effects on glucose metabolism. A large base of research indicates that bariatric surgeries such as Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), and biliopancreatic diversion (BPD) improve diabetes in most patients, with effects frequently evident prior to substantial weight reduction. There is now unequivocal evidence from randomized controlled trials that the efficacy of surgery is superior to intensive life-style/medical management. Despite advances in the clinical understanding and application of bariatric surgery, there remains only limited knowledge of the mechanisms by which these procedures confer such large changes to metabolic physiology. The improvement of insulin sensitivity that occurs with weight loss (e.g., the result of diet, illness, physical training) also accompanies bariatric surgery. However, there is evidence to support specific effects of surgery on insulin clearance, hepatic glucose production, and islet function. Understanding the mechanisms by which surgery affects these parameters of glucose regulation has the potential to identify new targets for therapeutic discovery. Studies to distinguish among bariatric surgeries on key parameters of glucose metabolism are limited but would be of considerable value to assist clinicians in selecting specific procedures and investigators in delineating the resulting physiology. This review is based on literature related to factors governing glucose metabolism and insulin secretion after the commonly used RYGB and VSG, and the less frequently used BPD and adjustable gastric banding.
Collapse
Affiliation(s)
- Jonathan D Douros
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Jenny Tong
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - David A D’Alessio
- Division of Endocrinology, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| |
Collapse
|
121
|
Wewer Albrechtsen NJ, Pedersen J, Galsgaard KD, Winther-Sørensen M, Suppli MP, Janah L, Gromada J, Vilstrup H, Knop FK, Holst JJ. The Liver-α-Cell Axis and Type 2 Diabetes. Endocr Rev 2019; 40:1353-1366. [PMID: 30920583 DOI: 10.1210/er.2018-00251] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/19/2019] [Indexed: 02/08/2023]
Abstract
Both type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD) strongly associate with increasing body mass index, and together these metabolic diseases affect millions of individuals. In patients with T2D, increased secretion of glucagon (hyperglucagonemia) contributes to diabetic hyperglycemia as proven by the significant lowering of fasting plasma glucose levels following glucagon receptor antagonist administration. Emerging data now indicate that the elevated plasma concentrations of glucagon may also be associated with hepatic steatosis and not necessarily with the presence or absence of T2D. Thus, fatty liver disease, most often secondary to overeating, may result in impaired amino acid turnover, leading to increased plasma concentrations of certain glucagonotropic amino acids (e.g., alanine). This, in turn, causes increased glucagon secretion that may help to restore amino acid turnover and ureagenesis, but it may eventually also lead to increased hepatic glucose production, a hallmark of T2D. Early experimental findings support the hypothesis that hepatic steatosis impairs glucagon's actions on amino acid turnover and ureagenesis. Hepatic steatosis also impairs hepatic insulin sensitivity and clearance that, together with hyperglycemia and hyperaminoacidemia, lead to peripheral hyperinsulinemia; systemic hyperinsulinemia may itself contribute to worsen peripheral insulin resistance. Additionally, obesity is accompanied by an impaired incretin effect, causing meal-related glucose intolerance. Lipid-induced impairment of hepatic sensitivity, not only to insulin but potentially also to glucagon, resulting in both hyperinsulinemia and hyperglucagonemia, may therefore contribute to the development of T2D at least in a subset of individuals with NAFLD.
Collapse
Affiliation(s)
- Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Nephrology and Endocrinology, Nordsjællands Hospital Hillerød, University of Copenhagen, Hillerød, Denmark
| | - Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie Winther-Sørensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Malte P Suppli
- Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Lina Janah
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Hendrik Vilstrup
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
122
|
Abstract
Amino acids perform a variety of functions in cells and organisms, particularly in the synthesis of proteins, as energy metabolites, neurotransmitters, and precursors for many other molecules. Amino acid transport plays a key role in all these functions. Inhibition of amino acid transport is pursued as a therapeutic strategy in several areas, such as diabetes and related metabolic disorders, neurological disorders, cancer, and stem cell biology. The role of amino acid transporters in these disorders and processes is well established, but the implementation of amino acid transporters as drug targets is still in its infancy. This is at least in part due to the underdeveloped pharmacology of this group of membrane proteins. Recent advances in structural biology, membrane protein expression, and inhibitor screening methodology will see an increased number of improved and selective inhibitors of amino acid transporters that can serve as tool compounds for further studies.
Collapse
Affiliation(s)
- Stefan Bröer
- 1 Research School of Biology, College of Science, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
123
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a brief summary of recent advances in our understanding of liver metabolism. The critical role of the liver in controlling whole-body energy homeostasis makes such understanding crucial to efficiently design new treatments for metabolic syndrome diseases, including type 2 diabetes (T2D). RECENT FINDINGS Significant advances have been made regarding our understanding of the direct and indirect effects of insulin on hepatic metabolism and the communication between the liver and other tissues. Moreover, the catabolic functions of glucagon, as well as the importance of hepatic redox status for the regulation of glucose production, are emerging as potential targets to reduce hyperglycemia. A resolution to the long-standing question "insulin suppression of hepatic glucose production, direct or indirect effect?" is starting to emerge. New advances in our understanding of important fasting-induced hepatic metabolic fluxes may help design better therapies for T2D.
Collapse
Affiliation(s)
- Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA.
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, 450 Brookline Av. LC-6219H, Boston, MA, 02215, USA.
- Dana-Farber Cancer Institute, 450 Brookline Av. LC-6213, Boston, MA, 02215, USA.
| |
Collapse
|
124
|
Karanth S, Chaurasia B, Bowman FM, Tippetts TS, Holland WL, Summers SA, Schlegel A. FOXN3 controls liver glucose metabolism by regulating gluconeogenic substrate selection. Physiol Rep 2019; 7:e14238. [PMID: 31552709 PMCID: PMC6759504 DOI: 10.14814/phy2.14238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/23/2019] [Accepted: 08/25/2019] [Indexed: 01/08/2023] Open
Abstract
The FOXN3 gene locus is associated with fasting blood glucose levels in non-diabetic human population genetic studies. The blood glucose-modifying variation within this gene regulates the abundance of both FOXN3 protein and transcript in primary human hepatocytes, with the hyperglycemia risk allele causing increases in both FOXN3 protein and transcript. Using transgenic and knock-out zebrafish models, we showed previously that FOXN3 is a transcriptional repressor that regulates fasting blood glucose by altering liver gene expression of MYC, a master transcriptional regulator of glucose utilization, and by modulating pancreatic α cell mass and function through an unknown mechanism. Since homozygous Foxn3 null mice die perinatally, and heterozygous carries of the null allele are smaller than wild-type siblings, we examine the metabolic effects of decreasing mouse liver Foxn3 expression in adult life, performing dynamic endocrine tests not feasible in adult zebrafish. Fasting glucose, glucagon, and insulin; and dynamic responses to glucose, insulin, pyruvate, glutamine, and glucagon were measured. Gluconeogenic and amino acid catabolic gene expression was examined in livers, as well. Knocking down liver Foxn3 expression via transduction with adeno-associated virus serotype 8 particles encoding a short hairpin RNA targeting Fonx3 decreases fasting glucose and increases Myc expression, without altering fasting glucagon or fasting insulin. Liver Foxn3 knock-down confers increases glucose tolerance, has no effect on insulin tolerance or response to glucagon challenge, blunts pyruvate and glutamine tolerance, and modulates expression of amino acid transporters and catabolic enzymes. We conclude that liver Foxn3 regulates substrate selection for gluconeogenesis.
Collapse
Affiliation(s)
- Santhosh Karanth
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
| | - Bhagirath Chaurasia
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
| | - Faith M. Bowman
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of BiochemistryUniversity of Utah School of MedicineSalt Lake CityUtah
| | - Trevor S. Tippetts
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
| | - William L. Holland
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
- Department of BiochemistryUniversity of Utah School of MedicineSalt Lake CityUtah
| | - Scott A. Summers
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
- Department of BiochemistryUniversity of Utah School of MedicineSalt Lake CityUtah
| | - Amnon Schlegel
- University of Utah Molecular Medicine ProgramSalt Lake CityUtah
- University of Utah Diabetes and Metabolism Research CenterSalt Lake CityUtah
- Department of Nutrition and Integrative PhysiologyCollege of HealthUniversity of UtahSalt Lake CityUtah
- Department of BiochemistryUniversity of Utah School of MedicineSalt Lake CityUtah
- Division of Endocrinology, Metabolism and DiabetesDepartment of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtah
| |
Collapse
|
125
|
Rose AJ. Role of Peptide Hormones in the Adaptation to Altered Dietary Protein Intake. Nutrients 2019; 11:E1990. [PMID: 31443582 PMCID: PMC6770041 DOI: 10.3390/nu11091990] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 12/25/2022] Open
Abstract
Dietary protein profoundly influences organismal traits ultimately affecting healthspan. While intracellular signalling downstream of altered amino acid supply is undoubtedly important, peptide hormones have emerged as critical factors determining systemic responses to variations in protein intake. Here the regulation and role of certain peptides hormones in such responses to altered dietary protein intake is reviewed.
Collapse
Affiliation(s)
- Adam J Rose
- Nutrient Metabolism & Signalling Laboratory, Department of Biochemistry and Molecular Biology, Metabolism, Diabetes and Obesity Program, Biomedicine Discovery Institute, Monash University, Clayton 3800, Australia.
| |
Collapse
|
126
|
Himuro M, Miyatsuka T, Suzuki L, Miura M, Katahira T, Goto H, Nishida Y, Sasaki S, Koike M, Shiota C, Gittes GK, Fujitani Y, Watada H. Cellular Autophagy in α Cells Plays a Role in the Maintenance of Islet Architecture. J Endocr Soc 2019; 3:1979-1992. [PMID: 31620668 PMCID: PMC6786006 DOI: 10.1210/js.2019-00075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/22/2019] [Indexed: 01/31/2023] Open
Abstract
Autophagy is known to play a pivotal role in intracellular quality control through the degradation of subcellular damaged organelles and components. Whereas autophagy is essential for maintaining β-cell function in pancreatic islets, it remains unclear as to how the cellular autophagy affects the homeostasis and function of glucagon-secreting α cells. To investigate the role of autophagy in α cells, we generated a mutant mouse model lacking Atg7, a key molecule for autophagosome formation, specifically in α cells. Histological analysis demonstrated more glucagon-positive cells, with a multilayered structure, in the islets under Atg7 deficiency, although metabolic profiles, such as body weight, blood glucose, and plasma glucagon levels were comparable between Atg7-deficient mice and control littermates. Consistent with our previous findings that Atg7 deficiency suppressed β-cell proliferation, cellular proliferation was suppressed in Atg7-deficient α cells. These findings suggest that α-cell autophagy plays a role in maintaining α-cell area and normal islet architecture but appears to be dispensable for metabolic homeostasis.
Collapse
Affiliation(s)
- Miwa Himuro
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takeshi Miyatsuka
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Luka Suzuki
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaki Miura
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Katahira
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiromasa Goto
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuya Nishida
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shugo Sasaki
- Department of Surgery, The University of British Columbia, Vancouver BC, Canada
| | - Masato Koike
- Departments of Cell Biology and Neurosciences, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chiyo Shiota
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George K Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular & Cellular Regulation, Gunma University, Maebashi, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
127
|
Hayashi Y. Glutaminostatin: Another facet of glucagon as a regulator of plasma amino acid concentrations. J Diabetes Investig 2019; 10:1391-1393. [PMID: 31254453 PMCID: PMC6825951 DOI: 10.1111/jdi.13110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/27/2022] Open
Abstract
Glucagon plays an essential role in robust feedback regulation between the liver and α‐cells, and exerts suppressive or static effects on the plasma concentration of amino acids, especially glutamine. Thereby, “glutaminostatin” might be an alternative name in recognition of another facet of glucagon as a suppressor of plasma glutamine levels.![]()
Collapse
Affiliation(s)
- Yoshitaka Hayashi
- Department of Endocrinology, Division of Stress Adaptation and Recognition, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
128
|
Janah L, Kjeldsen S, Galsgaard KD, Winther-Sørensen M, Stojanovska E, Pedersen J, Knop FK, Holst JJ, Wewer Albrechtsen NJ. Glucagon Receptor Signaling and Glucagon Resistance. Int J Mol Sci 2019; 20:E3314. [PMID: 31284506 PMCID: PMC6651628 DOI: 10.3390/ijms20133314] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023] Open
Abstract
Hundred years after the discovery of glucagon, its biology remains enigmatic. Accurate measurement of glucagon has been essential for uncovering its pathological hypersecretion that underlies various metabolic diseases including not only diabetes and liver diseases but also cancers (glucagonomas). The suggested key role of glucagon in the development of diabetes has been termed the bihormonal hypothesis. However, studying tissue-specific knockout of the glucagon receptor has revealed that the physiological role of glucagon may extend beyond blood-glucose regulation. Decades ago, animal and human studies reported an important role of glucagon in amino acid metabolism through ureagenesis. Using modern technologies such as metabolomic profiling, knowledge about the effects of glucagon on amino acid metabolism has been expanded and the mechanisms involved further delineated. Glucagon receptor antagonists have indirectly put focus on glucagon's potential role in lipid metabolism, as individuals treated with these antagonists showed dyslipidemia and increased hepatic fat. One emerging field in glucagon biology now seems to include the concept of hepatic glucagon resistance. Here, we discuss the roles of glucagon in glucose homeostasis, amino acid metabolism, and lipid metabolism and present speculations on the molecular pathways causing and associating with postulated hepatic glucagon resistance.
Collapse
Affiliation(s)
- Lina Janah
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sasha Kjeldsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Marie Winther-Sørensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Elena Stojanovska
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Cardiology, Nephrology and Endocrinology, Nordsjællands Hospital Hillerød, University of Copenhagen, 3400 Hillerød, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, 2820 Gentofte, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Department of Clinical Biochemistry, Rigshospitalet, 2100 Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark.
| |
Collapse
|
129
|
Sharma AX, Quittner-Strom EB, Lee Y, Johnson JA, Martin SA, Yu X, Li J, Lu J, Cai Z, Chen S, Wang MY, Zhang Y, Pearson MJ, Dorn AC, McDonald JG, Gordillo R, Yan H, Thai D, Wang ZV, Unger RH, Holland WL. Glucagon Receptor Antagonism Improves Glucose Metabolism and Cardiac Function by Promoting AMP-Mediated Protein Kinase in Diabetic Mice. Cell Rep 2019; 22:1760-1773. [PMID: 29444429 PMCID: PMC5978750 DOI: 10.1016/j.celrep.2018.01.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/30/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
The antidiabetic potential of glucagon receptor antagonism presents an opportunity for use in an insulin-centric clinical environment. To investigate the metabolic effects of glucagon receptor antagonism in type 2 diabetes, we treated Leprdb/db and Lepob/ob mice with REMD 2.59, a human monoclonal antibody and competitive antagonist of the glucagon receptor. As expected, REMD 2.59 suppresses hepatic glucose production and improves glycemia. Surprisingly, it also enhances insulin action in both liver and skeletal muscle, coinciding with an increase in AMP-activated protein kinase (AMPK)-mediated lipid oxidation. Furthermore, weekly REMD 2.59 treatment over a period of months protects against diabetic cardiomyopathy. These functional improvements are not derived simply from correcting the systemic milieu; nondiabetic mice with cardiac-specific overexpression of lipoprotein lipase also show improvements in contractile function after REMD 2.59 treatment. These observations suggest that hyperglucagonemia enables lipotoxic conditions, allowing the development of insulin resistance and cardiac dysfunction during disease progression.
Collapse
Affiliation(s)
- Ankit X Sharma
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ezekiel B Quittner-Strom
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Young Lee
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Joshua A Johnson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Sarah A Martin
- Department of Molecular Genetics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Xinxin Yu
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Jianping Li
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - John Lu
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | | | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - May-Yun Wang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - Yiyi Zhang
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Mackenzie J Pearson
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Andie C Dorn
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Center for Human Nutrition, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Hai Yan
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | - Dung Thai
- REMD Biotherapeutics Inc., Camarillo, CA 93012, USA
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA
| | - Roger H Unger
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA; Medical Service, Veteran's Administration North Texas Health Care System, Dallas, TX 75216, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390-8549, USA.
| |
Collapse
|
130
|
Mezza T, Cinti F, Cefalo CMA, Pontecorvi A, Kulkarni RN, Giaccari A. β-Cell Fate in Human Insulin Resistance and Type 2 Diabetes: A Perspective on Islet Plasticity. Diabetes 2019; 68:1121-1129. [PMID: 31109941 PMCID: PMC6905483 DOI: 10.2337/db18-0856] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/17/2019] [Indexed: 12/23/2022]
Abstract
Although it is well established that type 2 diabetes (T2D) is generally due to the progressive loss of β-cell insulin secretion against a background of insulin resistance, the actual correlation of reduced β-cell mass to its defective function continues to be debated. There is evidence that a compensatory increase in β-cell mass, and the consequent insulin secretion, can effectively cope with states of insulin resistance, until hyperglycemia supervenes. Recent data strongly indicate that the mechanisms by which islets are able to compensate in response to insulin resistance in peripheral tissues is secondary to hyperplasia, as well as the activation of multiple cellular machineries with diverse functions. Importantly, islet cells exhibit plasticity in altering their endocrine commitment; for example, by switching from secretion of glucagon to secretion of insulin and back (transdifferentiation) or from an active secretory state to a nonsecretory quiescent state (dedifferentiation) and back. Lineage tracing (a method used to track each cell though its differentiation process) has demonstrated these potentials in murine models. A limitation to drawing conclusions from human islet research is that most studies are derived from human autopsy and/or organ donor samples, which lack in vivo functional and metabolic profiling. In this review, we specifically focus on evidence of islet plasticity in humans-from the normal state, progressing to insulin resistance to overt T2D-to explain the seemingly contradictory results from different cross-sectional studies in the literature. We hope the discussion on this intriguing scenario will provide a forum for the scientific community to better understand the disease and in the long term pave the way for personalized therapies.
Collapse
Affiliation(s)
- Teresa Mezza
- U.O.C. Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italia
- Istituto di Patologia Speciale Medica e Semeiotica Clinica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Francesca Cinti
- U.O.C. Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italia
- Istituto di Patologia Speciale Medica e Semeiotica Clinica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Chiara Maria Assunta Cefalo
- U.O.C. Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italia
- Istituto di Patologia Speciale Medica e Semeiotica Clinica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Alfredo Pontecorvi
- U.O.C. Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italia
- Istituto di Patologia Speciale Medica e Semeiotica Clinica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Rohit N Kulkarni
- Islet Cell & Regenerative Biology, Joslin Diabetes Center and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Andrea Giaccari
- U.O.C. Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italia
- Istituto di Patologia Speciale Medica e Semeiotica Clinica, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
131
|
Lam CJ, Rankin MM, King KB, Wang MC, Shook BC, Kushner JA. Glucagon Receptor Antagonist-Stimulated α-Cell Proliferation Is Severely Restricted With Advanced Age. Diabetes 2019; 68:963-974. [PMID: 30833466 PMCID: PMC6477910 DOI: 10.2337/db18-1293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/18/2019] [Indexed: 01/26/2023]
Abstract
Glucagon-containing α-cells potently regulate glucose homeostasis, but the developmental biology of α-cells in adults remains poorly understood. Although glucagon receptor antagonists (GRAs) have great potential as antidiabetic therapies, murine and human studies have raised concerns that GRAs might cause uncontrolled α-cell growth. Surprisingly, previous rodent GRA studies were only performed in young mice, implying that the potential impact of GRAs to drive α-cell expansion in adult patients is unclear. We assessed adaptive α-cell turnover and adaptive proliferation, administering a novel GRA (JNJ-46207382) to both young and aged mice. Basal α-cell proliferation rapidly declined soon after birth and continued to drop to very low levels in aged mice. GRA drove a 2.4-fold increase in α-cell proliferation in young mice. In contrast, GRA-induced α-cell proliferation was severely reduced in aged mice, although still present at 3.2-fold the very low basal rate of aged controls. To interrogate the lineage of GRA-induced α-cells, we sequentially administered thymidine analogs and quantified their incorporation into α-cells. Similar to previous studies of β-cells, α-cells only divided once in both basal and stimulated conditions. Lack of contribution from highly proliferative "transit-amplifying" cells supports a model whereby α-cells expand by self-renewal and not via specialized progenitors.
Collapse
Affiliation(s)
- Carol J Lam
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Matthew M Rankin
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
- Janssen Research and Development, Johnson & Johnson, Springhouse, PA
| | - Kourtney B King
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Melinda C Wang
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
| | - Brian C Shook
- Janssen Research and Development, Johnson & Johnson, Springhouse, PA
| | - Jake A Kushner
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, TX
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
132
|
Dean ED, Vuguin PM, Charron MJ. Glucagon: The Name Says It All, or Not! Endocrinology 2019; 160:1359-1361. [PMID: 31144717 PMCID: PMC6482032 DOI: 10.1210/en.2019-00251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 11/19/2022]
Affiliation(s)
- E Danielle Dean
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Patricia M Vuguin
- Department of Pediatrics, Pediatric Endocrinology, Columbia University, Vagelos College of Physicians & Surgeons, New York, New York
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Obstetrics and Gynecology and Women’s Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
133
|
Galsgaard KD, Pedersen J, Knop FK, Holst JJ, Wewer Albrechtsen NJ. Glucagon Receptor Signaling and Lipid Metabolism. Front Physiol 2019; 10:413. [PMID: 31068828 PMCID: PMC6491692 DOI: 10.3389/fphys.2019.00413] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 03/26/2019] [Indexed: 01/04/2023] Open
Abstract
Glucagon is secreted from the pancreatic alpha cells upon hypoglycemia and stimulates hepatic glucose production. Type 2 diabetes is associated with dysregulated glucagon secretion, and increased glucagon concentrations contribute to the diabetic hyperglycemia. Antagonists of the glucagon receptor have been considered as glucose-lowering therapy in type 2 diabetes patients, but their clinical applicability has been questioned because of reports of therapy-induced increments in liver fat content and increased plasma concentrations of low-density lipoprotein. Conversely, in animal models, increased glucagon receptor signaling has been linked to improved lipid metabolism. Glucagon acts primarily on the liver and by regulating hepatic lipid metabolism glucagon may reduce hepatic lipid accumulation and decrease hepatic lipid secretion. Regarding whole-body lipid metabolism, it is controversial to what extent glucagon influences lipolysis in adipose tissue, particularly in humans. Glucagon receptor agonists combined with glucagon-like peptide 1 receptor agonists (dual agonists) improve dyslipidemia and reduce hepatic steatosis. Collectively, emerging data support an essential role of glucagon for lipid metabolism.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Nephrology and Endocrinology, Nordsjællands Hospital Hillerød, University of Copenhagen, Hillerød, Denmark
| | - Filip K Knop
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
134
|
Galsgaard KD, Winther-Sørensen M, Pedersen J, Kjeldsen SAS, Rosenkilde MM, Wewer Albrechtsen NJ, Holst JJ. Glucose and amino acid metabolism in mice depend mutually on glucagon and insulin receptor signaling. Am J Physiol Endocrinol Metab 2019; 316:E660-E673. [PMID: 30807215 DOI: 10.1152/ajpendo.00410.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucagon and insulin are important regulators of blood glucose. The importance of insulin receptor signaling for alpha-cell secretion and of glucagon receptor signaling for beta-cell secretion is widely discussed and of clinical interest. Amino acids are powerful secretagogues for both hormones, and glucagon controls amino acid metabolism through ureagenesis. The role of insulin in amino acid metabolism is less clear. Female C57BL/6JRj mice received an insulin receptor antagonist (IRA) (S961; 30 nmol/kg), a glucagon receptor antagonist (GRA) (25-2648; 100 mg/kg), or both GRA and IRA (GRA + IRA) 3 h before intravenous administration of similar volumes of saline, glucose (0.5 g/kg), or amino acids (1 µmol/g) while anesthetized with isoflurane. IRA caused basal hyperglycemia, hyperinsulinemia, and hyperglucagonemia. Unexpectedly, IRA lowered basal plasma concentrations of amino acids, whereas GRA increased amino acids, lowered glycemia, and increased glucagon but did not influence insulin concentrations. After administration of GRA + IRA, insulin secretion was significantly reduced compared with IRA administration alone. Blood glucose responses to a glucose and amino acid challenge were similar after vehicle and GRA + IRA administration but greater after IRA and lower after GRA. Anesthesia may have influenced the results, which otherwise strongly suggest that both hormones are essential for the maintenance of glucose homeostasis and that the secretion of both is regulated by powerful negative feedback mechanisms. In addition, insulin limits glucagon secretion, while endogenous glucagon stimulates insulin secretion, revealed during lack of insulin autocrine feedback. Finally, glucagon receptor signaling seems to be of greater importance for amino acid metabolism than insulin receptor signaling.
Collapse
Affiliation(s)
- Katrine D Galsgaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Cardiology, Nephrology and Endocrinology, Nordsjaellands Hospital Hilleroed, University of Copenhagen, Hilleroed, Denmark
| | - Sasha A S Kjeldsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen , Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
135
|
Gao C, Ren SV, Yu J, Baal U, Thai D, Lu J, Zeng C, Yan H, Wang Y. Glucagon Receptor Antagonism Ameliorates Progression of Heart Failure. JACC Basic Transl Sci 2019; 4:161-172. [PMID: 31061918 PMCID: PMC6488764 DOI: 10.1016/j.jacbts.2018.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 02/08/2023]
Abstract
Mice were treated with a fully human monoclonal glucagon receptor antagonistic antibody REMD2.59 following myocardial infarction or pressure overload. REMD2.59 treatment blunted cardiac hypertrophy and fibrotic remodeling, and attenuated contractile dysfunction at 4 weeks after myocardial infarction. In addition, REMD2.59 treatment at the onset of pressure overload significantly suppressed cardiac hypertrophy and chamber dilation with marked preservation of cardiac systolic and diastolic function. Initiation of REMD2.59 treatment 2 weeks after pressure overload significantly blunted the progression of cardiac pathology. These results provide the first in vivo proof-of-concept evidence that glucagon receptor antagonism is a potentially efficacious therapy to ameliorate both onset and progression of heart failure.
Collapse
Affiliation(s)
- Chen Gao
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| | - Shuxun Vincent Ren
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Junyi Yu
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Ulysis Baal
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Dung Thai
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - John Lu
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
| | - Hai Yan
- REMD Biotherapeutics, Camarillo, California
- Beijing Cosci-REMD Biotherapeutics, Beijing, China
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Address for correspondence: Dr. Yibin Wang or Dr. Chen Gao, Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, 650 Charles E. Young Drive, Room CHS 37-200J, Los Angeles, California 90095.
| |
Collapse
|
136
|
Amino acid transporters in the regulation of insulin secretion and signalling. Biochem Soc Trans 2019; 47:571-590. [PMID: 30936244 DOI: 10.1042/bst20180250] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 01/02/2023]
Abstract
Amino acids are increasingly recognised as modulators of nutrient disposal, including their role in regulating blood glucose through interactions with insulin signalling. More recently, cellular membrane transporters of amino acids have been shown to form a pivotal part of this regulation as they are primarily responsible for controlling cellular and circulating amino acid concentrations. The availability of amino acids regulated by transporters can amplify insulin secretion and modulate insulin signalling in various tissues. In addition, insulin itself can regulate the expression of numerous amino acid transporters. This review focuses on amino acid transporters linked to the regulation of insulin secretion and signalling with a focus on those of the small intestine, pancreatic β-islet cells and insulin-responsive tissues, liver and skeletal muscle. We summarise the role of the amino acid transporter B0AT1 (SLC6A19) and peptide transporter PEPT1 (SLC15A1) in the modulation of global insulin signalling via the liver-secreted hormone fibroblast growth factor 21 (FGF21). The role of vesicular vGLUT (SLC17) and mitochondrial SLC25 transporters in providing glutamate for the potentiation of insulin secretion is covered. We also survey the roles SNAT (SLC38) family and LAT1 (SLC7A5) amino acid transporters play in the regulation of and by insulin in numerous affective tissues. We hypothesise the small intestine amino acid transporter B0AT1 represents a crucial nexus between insulin, FGF21 and incretin hormone signalling pathways. The aim is to give an integrated overview of the important role amino acid transporters have been found to play in insulin-regulated nutrient signalling.
Collapse
|
137
|
Gu L, Cui X, Lang S, Wang H, Hong T, Wei R. Glucagon receptor antagonism increases mouse pancreatic δ-cell mass through cell proliferation and duct-derived neogenesis. Biochem Biophys Res Commun 2019; 512:864-870. [PMID: 30929915 DOI: 10.1016/j.bbrc.2019.03.148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 03/22/2019] [Indexed: 01/05/2023]
Abstract
Pancreatic δ-cells, which produce somatostatin, play an indispensable role in glucose homeostasis by inhibiting glucagon and insulin secretion in a paracrine manner. Recent studies have shown that δ-cells are couple with β-cells to suppress α-cell activity. Under certain circumstances, δ-cells could also be trans-differentiated into insulin-producing β-cells. Thus, pancreatic islet may benefit from δ-cell hyperplasia. However, an effective way to increase δ-cell mass has been rarely reported. Here, we found that REMD 2.59, a human monoclonal antibody and competitive antagonist of the glucagon receptor, massively boosted δ-cell number and increased plasma somatostatin level in both normoglycemic and type 1 diabetic (T1D) mice. The increased δ-cells were due to both δ-cell proliferation and derivation of duct lining cells. Notably, the enlarged δ-cell mass could reduce β-cell burdens by inducing FoxO1 nuclear translocation in normoglycemic mice. Moreover, some somatostatin-positive cells were co-localized with C-peptide in T1D mice, suggesting that δ-cells might be a source of the newborn β-cells. Collectively, these observations suggest that treatment with the glucagon receptor monoclonal antibody can increase pancreatic δ-cell mass by promoting self-replication and inducing duct-derived neogenesis both in normoglycemia and diabetic mice.
Collapse
Affiliation(s)
- Liangbiao Gu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Shan Lang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Haining Wang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
138
|
Capozzi ME, Svendsen B, Encisco SE, Lewandowski SL, Martin MD, Lin H, Jaffe JL, Coch RW, Haldeman JM, MacDonald PE, Merrins MJ, D'Alessio DA, Campbell JE. β Cell tone is defined by proglucagon peptides through cAMP signaling. JCI Insight 2019; 4:126742. [PMID: 30720465 DOI: 10.1172/jci.insight.126742] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/29/2019] [Indexed: 01/05/2023] Open
Abstract
Paracrine interactions between pancreatic islet cells have been proposed as a mechanism to regulate hormone secretion and glucose homeostasis. Here, we demonstrate the importance of proglucagon-derived peptides (PGDPs) for α to β cell communication and control of insulin secretion. Signaling through this system occurs through both the glucagon-like peptide receptor (Glp1r) and glucagon receptor (Gcgr). Loss of PGDPs, or blockade of their receptors, decreases insulin secretion in response to both metabolic and nonmetabolic stimulation of mouse and human islets. This effect is due to reduced β cell cAMP and affects the quantity but not dynamics of insulin release, indicating that PGDPs dictate the magnitude of insulin output in an isolated islet. In healthy mice, additional factors that stimulate cAMP can compensate for loss of PGDP signaling; however, input from α cells is essential to maintain glucose tolerance during the metabolic stress induced by high-fat feeding. These findings demonstrate an essential role for α cell regulation of β cells, raising the possibility that abnormal paracrine signaling contributes to impaired insulin secretion in diabetes. Moreover, these findings support reconsideration of the role for α cells in postprandial glucose control.
Collapse
Affiliation(s)
- Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Berit Svendsen
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Sara E Encisco
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Sophie L Lewandowski
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mackenzie D Martin
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Haopeng Lin
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Alberta, Canada
| | - Justin L Jaffe
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Reilly W Coch
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.,Department of Medicine and
| | - Jonathan M Haldeman
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Alberta, Canada
| | - Matthew J Merrins
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David A D'Alessio
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.,Department of Medicine and
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA.,Department of Medicine and.,Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
139
|
Sakurai K, Eguchi A, Watanabe M, Yamamoto M, Ishikawa K, Mori C. Exploration of predictive metabolic factors for gestational diabetes mellitus in Japanese women using metabolomic analysis. J Diabetes Investig 2019; 10:513-520. [PMID: 29956893 PMCID: PMC6400174 DOI: 10.1111/jdi.12887] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
AIMS/INTRODUCTION We aimed to explore novel predictive markers for gestational diabetes mellitus using metabolomic analysis in pregnant Japanese women. MATERIALS AND METHODS We carried out a case-control study with a cohort of participants enrolled during the first or early second trimester in the Center of Chiba Unit of the Japan Environment and Children's Study. Participants were classified as either gestational diabetes mellitus cases or matched controls based on age, body mass index and parity. Metabolite levels of their serum and urine obtained randomly before the diagnosis of gestational diabetes mellitus were analyzed using hydrophilic interaction chromatography tandem mass spectrometry. Orthogonal projections to latent structures discriminant analysis was carried out to investigate metabolome profiles for the different groups. Metabolites with a variable importance in projection value of >1.5 were identified as potential markers. RESULTS In total, 242 participants were enrolled in the study, of which 121 were cases. The R2X, R2Y and Q2 parameters for the discrimination ability of the resulting models were 0.388, 0.492 and 0.45 for serum, and 0.454, 0.674 and 0.483 for urine, respectively. We finally identified three metabolites in serum and 20 in urine as potential biomarkers. Glutamine in serum and ethanolamine and 1,3-diphosphoglycerate in urine showed >0.8 area under the receiver operating characteristic curves. CONCLUSIONS The present study identified serum and urine metabolites that are possible predictive markers of subsequent gestational diabetes mellitus in Japanese women. Further studies are required to elucidate their efficacy.
Collapse
Affiliation(s)
- Kenichi Sakurai
- Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Akifumi Eguchi
- Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | | | - Midori Yamamoto
- Center for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Ko Ishikawa
- Department of Clinical Cell Biology and MedicineGraduate School of MedicineChiba UniversityChibaJapan
| | - Chisato Mori
- Center for Preventive Medical SciencesChiba UniversityChibaJapan
- Department of Bioenvironmental MedicineGraduate School of MedicineChiba UniversityChibaJapan
| |
Collapse
|
140
|
Hart NJ, Powers AC. Use of human islets to understand islet biology and diabetes: progress, challenges and suggestions. Diabetologia 2019; 62:212-222. [PMID: 30547228 PMCID: PMC6325002 DOI: 10.1007/s00125-018-4772-2] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
Abstract
Over the last two decades, improved access to human islets and the development of human islet distribution networks have enabled the use of millions of human islets in hundreds of scientific research projects, leading to a dramatic increase in our understanding of human islet biology. Here we discuss recent scientific advances as well as methodological and experimental challenges that impact human islet quality, experimental outcomes and the reporting of human islets used in scientific publications. In a survey of over 200 scientific publications with human islet experimentation, we found that the reporting of critical information was quite variable, sometimes obscure, and often failed to adequately outline the experiments and results using human islets. As the complexity of human islet research grows, we propose that members of the human islet research ecosystem work together to develop procedures and approaches for accessible and transparent collecting and reporting of crucial human islet characteristics and, through this, enhance collaboration, reproducibility and rigour, leading to further advances in our understanding of human islet biology.
Collapse
Affiliation(s)
- Nathaniel J Hart
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, 7465 Medical Research Bldg IV, Vanderbilt University Medical Center, 2215 Garland Avenue, Nashville, TN, 37232-0475, USA
- Institute for Cellular Transplantation, College of Medicine, Department of Surgery, Arizona Health Sciences Center, Tucson, AZ, USA
| | - Alvin C Powers
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, 7465 Medical Research Bldg IV, Vanderbilt University Medical Center, 2215 Garland Avenue, Nashville, TN, 37232-0475, USA.
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.
- VA Tennessee Valley Healthcare, Nashville, TN, USA.
| |
Collapse
|
141
|
Abstract
Findings from the past 10 years have placed the glucagon-secreting pancreatic α-cell centre stage in the development of diabetes mellitus, a disease affecting almost one in every ten adults worldwide. Glucagon secretion is reduced in patients with type 1 diabetes mellitus, increasing the risk of insulin-induced hypoglycaemia, but is enhanced in type 2 diabetes mellitus, exacerbating the effects of diminished insulin release and action on blood levels of glucose. A better understanding of the mechanisms underlying these changes is therefore an important goal. RNA sequencing reveals that, despite their opposing roles in the control of blood levels of glucose, α-cells and β-cells have remarkably similar patterns of gene expression. This similarity might explain the fairly facile interconversion between these cells and the ability of the α-cell compartment to serve as a source of new β-cells in models of extreme β-cell loss that mimic type 1 diabetes mellitus. Emerging data suggest that GABA might facilitate this interconversion, whereas the amino acid glutamine serves as a liver-derived factor to promote α-cell replication and maintenance of α-cell mass. Here, we survey these developments and their therapeutic implications for patients with diabetes mellitus.
Collapse
Affiliation(s)
| | - Pauline Chabosseau
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK.
| |
Collapse
|
142
|
Rivero-Gutierrez B, Haller A, Holland J, Yates E, Khrisna R, Habegger K, Dimarchi R, D'Alessio D, Perez-Tilve D. Deletion of the glucagon receptor gene before and after experimental diabetes reveals differential protection from hyperglycemia. Mol Metab 2018; 17:28-38. [PMID: 30170980 PMCID: PMC6197675 DOI: 10.1016/j.molmet.2018.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Mice with congenital loss of the glucagon receptor gene (Gcgr-/- mice) remain normoglycemic in insulinopenic conditions, suggesting that unopposed glucagon action is the driving force for hyperglycemia in Type-1 Diabetes Mellitus (T1DM). However, chronic loss of GCGR results in a neomorphic phenotype that includes hormonal signals with hypoglycemic activity. We combined temporally-controlled GCGR deletion with pharmacological treatments to dissect the direct contribution of GCGR signaling to glucose control in a common mouse model of T1DM. METHODS We induced experimental T1DM by injecting the beta-cell cytotoxin streptozotocin (STZ) in mice with congenital or temporally-controlled Gcgr loss-of-function using tamoxifen (TMX). RESULTS Disruption of Gcgr expression, using either an inducible approach in adult mice or animals with congenital knockout, abolished the response to a long-acting Gcgr agonist. Mice with either developmental Gcgr disruption or inducible deletion several weeks before STZ treatment maintained normoglycemia. However, mice with inducible knockout of the Gcgr one week after the onset of STZ diabetes had only partial correction of hyperglycemia, an effect that was reversed by GLP-1 receptor blockade. Mice with Gcgr deletion for either 2 or 6 weeks had similar patterns of gene expression, although the changes were generally larger with longer GCGR knockout. CONCLUSIONS These findings demonstrate that the effects of glucagon to mitigate diabetic hyperglycemia are not through acute signaling but require compensations that take weeks to develop.
Collapse
Affiliation(s)
- Belen Rivero-Gutierrez
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - April Haller
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Jenna Holland
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Emily Yates
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA
| | - Radha Khrisna
- Department of Medicine, Duke University School of Medicine, NC, USA
| | - Kirk Habegger
- Comprehensive Diabetes Center and Department of Medicine - Endocrinology, Diabetes & and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard Dimarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - David D'Alessio
- Department of Medicine, Duke University School of Medicine, NC, USA
| | - Diego Perez-Tilve
- Department of Internal Medicine, University of Cincinnati, 2180 E. Galbraith Rd, Cincinnati, OH, USA.
| |
Collapse
|
143
|
Demant M, Bagger JI, Suppli MP, Lund A, Gyldenløve M, Hansen KB, Hare KJ, Christensen M, Sonne DP, Holst JJ, Vilsbøll T, Knop FK. Determinants of Fasting Hyperglucagonemia in Patients with Type 2 Diabetes and Nondiabetic Control Subjects. Metab Syndr Relat Disord 2018; 16:530-536. [PMID: 30325692 DOI: 10.1089/met.2018.0066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Fasting hyperglucagonemia can be detrimental to glucose metabolism in patients with type 2 diabetes (T2D) and may contribute to metabolic disturbances in obese and/or prediabetic subjects. However, the mechanisms underlying fasting hyperglucagonemia remain elusive. METHODS We evaluated the interrelationship between fasting hyperglucagonemia and demographic and biochemical parameters in 106 patients with T2D (31% female, age: 57 ± 9 years [mean ± standard deviation; body mass index (BMI): 30.1 ± 4.4 kg/m2; fasting plasma glucose (FPG): 9.61 ± 2.39 mM; hemoglobin A1c (HbA1c): 57.1 ± 13.1 mmol/mol] and 163 nondiabetic control subjects (29% female; age: 45 ± 17 years; BMI: 25.8 ± 4.1 kg/m2; FPG: 5.2 ± 0.4 mM; and HbA1c: 35.4 ± 3.8 mmol/mol). Multiple linear regression analysis was applied using a stepwise approach with fasting plasma glucagon as dependent parameter and BMI, waist-to-hip ratio (WHR), blood pressure, hemoglobin A1c, FPG, and insulin concentrations as independent parameters. RESULTS Fasting plasma glucagon concentrations were significantly higher among patients with T2D (13.5 ± 6.3 vs. 8.5 ± 3.8 mM, P < 0.001) together with HbA1c (P < 0.001), FPG (P < 0.001), and insulin (84.9 ± 56.4 vs. 57.7 ± 35.3 mM, P < 0.001). When adjusted for T2D, HbA1c and insulin were significantly positive determinants for fasting plasma glucagon concentrations. Furthermore, WHR comprised a significant positive determinant. CONCLUSIONS We confirm that fasting plasma glucagon concentrations are abnormally high in patients with T2D, and show that fasting plasma glucagon concentrations are influenced by WHR (in addition to glycemic control and fasting plasma insulin concentrations), which may point to visceral fat deposition as an important determinant of increased fasting plasma glucagon concentrations.
Collapse
Affiliation(s)
- Mia Demant
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Jonatan I Bagger
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Malte P Suppli
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Asger Lund
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Mette Gyldenløve
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Katrine B Hansen
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Kristine J Hare
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Mikkel Christensen
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - David P Sonne
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark
| | - Jens J Holst
- 2 Department of Biomedical Sciences and Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Tina Vilsbøll
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark .,3 Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen, Denmark
| | - Filip K Knop
- 1 Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, Hellerup, Denmark .,3 Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen, Denmark .,4 Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen, Denmark
| |
Collapse
|
144
|
The first pediatric case of glucagon receptor defect due to biallelic mutations in GCGR is identified by newborn screening of elevated arginine. Mol Genet Metab Rep 2018; 17:46-52. [PMID: 30294546 PMCID: PMC6171159 DOI: 10.1016/j.ymgmr.2018.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022] Open
Abstract
Glucagon receptor (GCGR) defect (Mahvash disease) is an autosomal recessive hereditary pancreatic neuroendocrine tumor (PNET) syndrome that has only been reported in adults with pancreatic α cell hyperplasia and PNETs. We describe a 7-year-old girl with persistent hyperaminoacidemia, notable for elevations of glutamine (normal ammonia), alanine (normal lactate), dibasic amino acids (arginine, lysine and ornithine), threonine and serine. She initially was brought to medical attention by an elevated arginine on newborn screening (NBS) and treated for presumed arginase deficiency with a low protein diet, essential amino acids formula and an ammonia scavenger drug. This treatment normalized plasma amino acids. She had intermittent emesis and anorexia, but was intellectually normal. Arginase enzyme assay and ARG1 sequencing and deletion/duplication analysis were normal. Treatments were stopped, but similar pattern of hyperaminoacidemia recurred. She also had hypercholesterolemia type IIa, with only elevated LDL cholesterol, despite an extremely lean body habitus. Exome sequencing was initially non-diagnostic. Through a literature search, we recognized the pattern of hyperaminoacidemia was strikingly similar to that reported in the Gcgr−/− knockout mice. Subsequently the patient was found to have an extremely elevated plasma glucagon and a novel, homozygous c.958_960del (p.Phe320del) variant in GCGR. Functional studies confirmed the pathogenicity of this variant. This case expands the clinical phenotype of GCGR defect in children and emphasizes the clinical utility of plasma amino acids in screening, diagnosis and monitoring glucagon signaling interruption. Early identification of a GCGR defect may provide an opportunity for potential beneficial treatment for an adult onset tumor predisposition disease. Describe the first case of glucagon receptor defect uniquely identified by abnormal newborn screening for elevated arginine. Characterize the pattern of hyperaminoacidemia in GCGR defect. Expand the clinical spectrum of GCGR defect from adult to childhood with a unique gastrointestinal manifestation.
Collapse
|
145
|
Hashim M, Yokoi N, Takahashi H, Gheni G, Okechi OS, Hayami T, Murao N, Hidaka S, Minami K, Mizoguchi A, Seino S. Inhibition of SNAT5 Induces Incretin-Responsive State From Incretin-Unresponsive State in Pancreatic β-Cells: Study of β-Cell Spheroid Clusters as a Model. Diabetes 2018; 67:1795-1806. [PMID: 29954738 DOI: 10.2337/db17-1486] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/11/2018] [Indexed: 11/13/2022]
Abstract
β-Cell-β-cell interactions are required for normal regulation of insulin secretion. We previously found that formation of spheroid clusters (called K20-SC) from MIN6-K20 clonal β-cells lacking incretin-induced insulin secretion (IIIS) under monolayer culture (called K20-MC) drastically induced incretin responsiveness. Here we investigated the mechanism by which an incretin-unresponsive state transforms to an incretin-responsive state using K20-SC as a model. Glutamate production by glucose through the malate-aspartate shuttle and cAMP signaling, both of which are critical for IIIS, were enhanced in K20-SC. SC formed from β-cells deficient for aspartate aminotransferase 1, a critical enzyme in the malate-aspartate shuttle, exhibited reduced IIIS. Expression of the sodium-coupled neutral amino acid transporter 5 (SNAT5), which is involved in glutamine transport, was downregulated in K20-SC and pancreatic islets of normal mice but was upregulated in K20-MC and islets of rodent models of obesity and diabetes, both of which exhibit impaired IIIS. Inhibition of SNAT5 significantly increased cellular glutamate content and improved IIIS in islets of these models and in K20-MC. These results suggest that suppression of SNAT5 activity, which results in increased glutamate production, and enhancement of cAMP signaling endows incretin-unresponsive β-cells with incretin responsiveness.
Collapse
MESH Headings
- Amino Acid Transport Systems, Neutral/agonists
- Amino Acid Transport Systems, Neutral/antagonists & inhibitors
- Amino Acid Transport Systems, Neutral/genetics
- Amino Acid Transport Systems, Neutral/metabolism
- Animals
- Anti-Obesity Agents/pharmacology
- Cell Communication/drug effects
- Cell Line
- Cells, Cultured
- Clone Cells
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Drug Resistance/drug effects
- Gene Expression Regulation/drug effects
- Hypoglycemic Agents/pharmacology
- Incretins/pharmacology
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Insulin-Secreting Cells/ultrastructure
- Islets of Langerhans/drug effects
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Islets of Langerhans/ultrastructure
- Male
- Membrane Transport Modulators/pharmacology
- Mice, Inbred Strains
- Microscopy, Electron, Transmission
- Models, Biological
- Obesity/drug therapy
- Obesity/metabolism
- Obesity/pathology
- RNA Interference
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Spheroids, Cellular/ultrastructure
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Mahira Hashim
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Norihide Yokoi
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Kansai Electric Power Medical Research Institute, Kobe, Japan
| | - Harumi Takahashi
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Kansai Electric Power Medical Research Institute, Kobe, Japan
| | - Ghupurjan Gheni
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Oduori S Okechi
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomohide Hayami
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Kansai Electric Power Medical Research Institute, Kobe, Japan
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Naoya Murao
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shihomi Hidaka
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kohtaro Minami
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Mizoguchi
- Department of Neural Regeneration and Cell Communication, Mie University Graduate School of Medicine, Tsu, Japan
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Kansai Electric Power Medical Research Institute, Kobe, Japan
| |
Collapse
|
146
|
Kim T, Nason S, Holleman C, Pepin M, Wilson L, Berryhill TF, Wende AR, Steele C, Young ME, Barnes S, Drucker DJ, Finan B, DiMarchi R, Perez-Tilve D, Tschöp M, Habegger KM. Glucagon Receptor Signaling Regulates Energy Metabolism via Hepatic Farnesoid X Receptor and Fibroblast Growth Factor 21. Diabetes 2018; 67:1773-1782. [PMID: 29925501 PMCID: PMC6110317 DOI: 10.2337/db17-1502] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Glucagon, an essential regulator of glucose and lipid metabolism, also promotes weight loss, in part through potentiation of fibroblast growth factor 21 (FGF21) secretion. However, FGF21 is only a partial mediator of metabolic actions ensuing from glucagon receptor (GCGR) activation, prompting us to search for additional pathways. Intriguingly, chronic GCGR agonism increases plasma bile acid levels. We hypothesized that GCGR agonism regulates energy metabolism, at least in part, through farnesoid X receptor (FXR). To test this hypothesis, we studied whole-body and liver-specific FXR-knockout (Fxr∆liver) mice. Chronic GCGR agonist (IUB288) administration in diet-induced obese (DIO) Gcgr, Fgf21, and Fxr whole-body or liver-specific knockout (∆liver) mice failed to reduce body weight when compared with wild-type (WT) mice. IUB288 increased energy expenditure and respiration in DIO WT mice, but not Fxr∆liver mice. GCGR agonism increased [14C]palmitate oxidation in hepatocytes isolated from WT mice in a dose-dependent manner, an effect blunted in hepatocytes from Fxr∆liver mice. Our data clearly demonstrate that control of whole-body energy expenditure by GCGR agonism requires intact FXR signaling in the liver. This heretofore-unappreciated aspect of glucagon biology has implications for the use of GCGR agonism in the therapy of metabolic disorders.
Collapse
MESH Headings
- Adiposity/drug effects
- Animals
- Anti-Obesity Agents/therapeutic use
- Calorimetry, Indirect
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Energy Metabolism/drug effects
- Fibroblast Growth Factors/genetics
- Fibroblast Growth Factors/metabolism
- Gene Expression Regulation/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/enzymology
- Mitochondria, Liver/metabolism
- Obesity/drug therapy
- Obesity/etiology
- Obesity/metabolism
- Obesity/pathology
- Organ Specificity
- Oxidative Phosphorylation/drug effects
- Peptides/therapeutic use
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Glucagon/agonists
- Receptors, Glucagon/genetics
- Receptors, Glucagon/metabolism
- Signal Transduction/drug effects
- Weight Gain/drug effects
Collapse
Affiliation(s)
- Teayoun Kim
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Shelly Nason
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Cassie Holleman
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Mark Pepin
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Landon Wilson
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Taylor F Berryhill
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Adam R Wende
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, AL
| | - Chad Steele
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Martin E Young
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Stephen Barnes
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, Ontario, Canada
| | - Brian Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
| | - Richard DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Diego Perez-Tilve
- Division of Endocrinology, Diabetes and Metabolism, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, OH
| | - Matthias Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, München, Germany
| | - Kirk M Habegger
- Comprehensive Diabetes Center and Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
147
|
Role of mTOR in Glucose and Lipid Metabolism. Int J Mol Sci 2018; 19:ijms19072043. [PMID: 30011848 PMCID: PMC6073766 DOI: 10.3390/ijms19072043] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin, mTOR is the master regulator of a cell’s growth and metabolic state in response to nutrients, growth factors and many extracellular cues. Its dysregulation leads to a number of metabolic pathological conditions, including obesity and type 2 diabetes. Here, we review recent findings on the role of mTOR in major metabolic organs, such as adipose tissues, liver, muscle, pancreas and brain. And their potentials as the mTOR related pharmacological targets will be also discussed.
Collapse
|
148
|
Karanth S, Adams JD, Serrano MDLA, Quittner-Strom EB, Simcox J, Villanueva CJ, Ozcan L, Holland WL, Yost HJ, Vella A, Schlegel A. A Hepatocyte FOXN3-α Cell Glucagon Axis Regulates Fasting Glucose. Cell Rep 2018; 24:312-319. [PMID: 29996093 DOI: 10.1016/j.celrep.2018.06.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/07/2018] [Accepted: 06/08/2018] [Indexed: 01/26/2023] Open
Abstract
The common genetic variation at rs8004664 in the FOXN3 gene is independently and significantly associated with fasting blood glucose, but not insulin, in non-diabetic humans. Recently, we reported that primary hepatocytes from rs8004664 hyperglycemia risk allele carriers have increased FOXN3 transcript and protein levels and liver-limited overexpression of human FOXN3, a transcriptional repressor that had not been implicated in metabolic regulation previously, increases fasting blood glucose in zebrafish. Here, we find that injection of glucagon into mice and adult zebrafish decreases liver Foxn3 protein and transcript levels. Zebrafish foxn3 loss-of-function mutants have decreased fasting blood glucose, blood glucagon, liver gluconeogenic gene expression, and α cell mass. Conversely, liver-limited overexpression of foxn3 increases α cell mass. Supporting these genetic findings in model organisms, non-diabetic rs8004664 risk allele carriers have decreased suppression of glucagon during oral glucose tolerance testing. By reciprocally regulating each other, liver FOXN3 and glucagon control fasting glucose.
Collapse
Affiliation(s)
- Santhosh Karanth
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - J D Adams
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Maria de Los Angeles Serrano
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Ezekiel B Quittner-Strom
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA
| | - Judith Simcox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Claudio J Villanueva
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Lale Ozcan
- Department of Medicine, Division of Molecular Medicine, Columbia University Medical Center, New York, NY, USA
| | - William L Holland
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - H Joseph Yost
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Adrian Vella
- Department of Internal Medicine, Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Amnon Schlegel
- University of Utah Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Internal Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT, USA; Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA.
| |
Collapse
|
149
|
|
150
|
Abstract
The lobules are the functional units of the liver. They consist of 15–25 layers of hepatocytes with specialized metabolic functions and gene expression patterns relative to their position along the lobule, a phenomenon referred to as metabolic zonation. The Wnt/β-catenin pathway regulates hepatocyte function but how the zonation is controlled to meet the metabolic demands of the liver is unclear. Glucagon regulates hepatic function. We now demonstrate that glucagon contributes to liver zonation by interacting and opposing the actions of the Wnt/β-catenin pathway. Liver zonation characterizes the separation of metabolic pathways along the lobules and is required for optimal function. Wnt/β-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/β-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/β-catenin. The glucagon and Wnt/β-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/β-catenin signaling pathway.
Collapse
|