1
|
Folli F, Finzi G, Manfrini R, Galli A, Casiraghi F, Centofanti L, Berra C, Fiorina P, Davalli A, La Rosa S, Perego C, Higgins PB. Mechanisms of action of incretin receptor based dual- and tri-agonists in pancreatic islets. Am J Physiol Endocrinol Metab 2023; 325:E595-E609. [PMID: 37729025 PMCID: PMC10874655 DOI: 10.1152/ajpendo.00236.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023]
Abstract
Simultaneous activation of the incretin G-protein-coupled receptors (GPCRs) via unimolecular dual-receptor agonists (UDRA) has emerged as a new therapeutic approach for type 2 diabetes. Recent studies also advocate triple agonism with molecules also capable of binding the glucagon receptor. In this scoping review, we discuss the cellular mechanisms of action (MOA) underlying the actions of these novel and therapeutically important classes of peptide receptor agonists. Clinical efficacy studies of several UDRAs have demonstrated favorable results both as monotherapies and when combined with approved hypoglycemics. Although the additive insulinotropic effects of dual glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic peptide receptor (GIPR) agonism were anticipated based on the known actions of either glucagon-like peptide-1 (GLP-1) or glucose-dependent insulinotropic peptide (GIP) alone, the additional benefits from GCGR were largely unexpected. Whether additional synergistic or antagonistic interactions among these G-protein receptor signaling pathways arise from simultaneous stimulation is not known. The signaling pathways affected by dual- and tri-agonism require more trenchant investigation before a comprehensive understanding of the cellular MOA. This knowledge will be essential for understanding the chronic efficacy and safety of these treatments.
Collapse
Affiliation(s)
- Franco Folli
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
- Unit of Diabetes, Endocrinology and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Giovanna Finzi
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
| | - Roberto Manfrini
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
- Unit of Diabetes, Endocrinology and Metabolism, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Alessandra Galli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Francesca Casiraghi
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Lucia Centofanti
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| | - Cesare Berra
- IRCCS MultiMedica, Sesto San Giovanni, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC, Università di Milano, Milan, Italy
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Carla Perego
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Paul B Higgins
- Department of Life & Physical Sciences, Atlantic Technological University, Letterkenny, Ireland
| |
Collapse
|
2
|
Fiorentino TV, Succurro E, Sciacqua A, Andreozzi F, Perticone F, Sesti G. Non-alcoholic fatty liver disease is associated with cardiovascular disease in subjects with different glucose tolerance. Diabetes Metab Res Rev 2020; 36:e3333. [PMID: 32356922 DOI: 10.1002/dmrr.3333] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is associated with cardiovascular disease (CVD) in patients with type 2 diabetes; nonetheless, it is unknown whether the relationship between NAFLD and CVD occurs also in subjects with prediabetes. Herein, we evaluated whether NAFLD is associated with prevalent CVD in subjects with different glucose tolerance states independently of cardiovascular risk factors. MATERIALS AND METHODS Presence of NALFD, defined by liver ultrasound, and its association with prevalent composite and individual CVD, including coronary artery disease (CAD) and cerebrovascular disease, was assessed in a cohort of 1254 Caucasian subjects classified as having normal glucose tolerance (NGT, n = 517), prediabetes (n = 397) or type 2 diabetes (n = 340). RESULTS Prevalence of NAFLD in the study population was 47.9%. Presence of NAFLD was linked to an augmented prevalence of composite CVD and individual CAD in all the three glucose tolerance groups. In a logistic regression model adjusted for several cardio-metabolic risk factors, subjects with NGT and NAFLD exhibited a 3.2- and 3.4-fold increased risk of having CVD or CAD, respectively, as compared with those without NAFLD. Similarly, subjects with prediabetes and NAFLD showed an increased risk of having CVD or CAD by 2.3- and 2.0-fold, respectively, in comparison to their counterpart without NAFLD. Within the group with type 2 diabetes, subjects having NAFLD displayed a 2.3- and 2.0-fold higher risk of having CVD or CAD, respectively, in comparison to those without NAFLD. CONCLUSION Ultrasonography-defined NAFLD is independently associated with an increased risk of having CVD in individuals with different glucose tolerance.
Collapse
Affiliation(s)
- Teresa Vanessa Fiorentino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Elena Succurro
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Francesco Perticone
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, University of Rome-Sapienza, Rome, Italy
| |
Collapse
|
3
|
Are Cape Peninsula baboons raiding their way to obesity and type II diabetes? - a comparative study. Comp Biochem Physiol A Mol Integr Physiol 2020; 250:110794. [PMID: 32827764 DOI: 10.1016/j.cbpa.2020.110794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/16/2020] [Accepted: 08/16/2020] [Indexed: 01/20/2023]
Abstract
Researchers, managers and conservationists in the Cape Peninsula, South Africa, have reported cases of individual baboons (Papio ursinus) appearing overweight, lethargic and having poor teeth. Despite an intensive baboon management programme, there are certain individual baboons and troops that continue to raid human food sources. These food sources often are high in processed carbohydrates and saturated fats. As this diet is highly associated with obesity, insulin resistance and type II diabetes, the present study aimed to establish if these baboons may be at risk of developing insulin resistance. Post mortem muscle samples from 17 Cape Peninsula and 7 control adult male baboons were rapidly frozen in liquid nitrogen and analysed for insulin receptor substrate-1 (IRS-1), glucose transporter 4 (GLUT4), oxidative and glycolytic markers of metabolism (citrate synthase, 3-hydroxyacyl-CoA-dehydrogenase, lactate dehydrogenase and creatine kinase activities), and muscle fibre morphology. The sampled Peninsula baboons were heavier (33 ± 2 vs. 29 ± 2 kg, P < 0.05) and had a higher frequency of poor teeth compared to control baboons. Muscle fibre type, fibre size, GLUT4 content, oxidative and glycolytic metabolism were not different between the two groups. However, IRS-1 content, a marker of insulin sensitivity, was significantly lower (by 43%, P < 0.001) in the Peninsula baboons compared to the controls. This study provides the first indirect evidence that some Peninsula baboons with a history of raiding human food sources, may be at risk of developing insulin resistance in the wild, with long term implications for population health.
Collapse
|
4
|
Fiorentino TV, Casiraghi F, Davalli AM, Finzi G, La Rosa S, Higgins PB, Abrahamian GA, Marando A, Sessa F, Perego C, Guardado-Mendoza R, Kamath S, Ricotti A, Fiorina P, Daniele G, Paez AM, Andreozzi F, Bastarrachea RA, Comuzzie AG, Gastaldelli A, Chavez AO, Di Cairano ES, Frost P, Luzi L, Dick EJ, Halff GA, DeFronzo RA, Folli F. Exenatide regulates pancreatic islet integrity and insulin sensitivity in the nonhuman primate baboon Papio hamadryas. JCI Insight 2019; 4:93091. [PMID: 31536476 DOI: 10.1172/jci.insight.93091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022] Open
Abstract
The glucagon-like peptide-1 receptor agonist exenatide improves glycemic control by several and not completely understood mechanisms. Herein, we examined the effects of chronic intravenous exenatide infusion on insulin sensitivity, β cell and α cell function and relative volumes, and islet cell apoptosis and replication in nondiabetic nonhuman primates (baboons). At baseline, baboons received a 2-step hyperglycemic clamp followed by an l-arginine bolus (HC/A). After HC/A, baboons underwent a partial pancreatectomy (tail removal) and received a continuous exenatide (n = 12) or saline (n = 12) infusion for 13 weeks. At the end of treatment, HC/A was repeated, and the remnant pancreas (head-body) was harvested. Insulin sensitivity increased dramatically after exenatide treatment and was accompanied by a decrease in insulin and C-peptide secretion, while the insulin secretion/insulin resistance (disposition) index increased by about 2-fold. β, α, and δ cell relative volumes in exenatide-treated baboons were significantly increased compared with saline-treated controls, primarily as the result of increased islet cell replication. Features of cellular stress and secretory dysfunction were present in islets of saline-treated baboons and absent in islets of exenatide-treated baboons. In conclusion, chronic administration of exenatide exerts proliferative and cytoprotective effects on β, α, and δ cells and produces a robust increase in insulin sensitivity in nonhuman primates.
Collapse
Affiliation(s)
- Teresa Vanessa Fiorentino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Francesca Casiraghi
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.,Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Alberto M Davalli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.,Department of Medicine, Endocrinology Unit, Ospedale San Raffaele, Milan, Italy
| | - Giovanna Finzi
- Unit of Pathology, Ospedale di Circolo and Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Stefano La Rosa
- Service of Clinical Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul B Higgins
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Gregory A Abrahamian
- Department of Surgery, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Alessandro Marando
- Unit of Pathology, Ospedale di Circolo and Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Unit of Pathology, Ospedale di Circolo and Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Carla Perego
- Department of Pharmacology and Biomolecular Science, University of Milan, Milan, Italy
| | - Rodolfo Guardado-Mendoza
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Subhash Kamath
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Andrea Ricotti
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Paolo Fiorina
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, Division of Health Science, Harvard University, Boston, Massachusetts, USA
| | - Giuseppe Daniele
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Ana M Paez
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy.,Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Raul A Bastarrachea
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Anthony G Comuzzie
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Amalia Gastaldelli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.,Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Alberto O Chavez
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Eliana S Di Cairano
- Department of Pharmacology and Biomolecular Science, University of Milan, Milan, Italy
| | - Patrice Frost
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Livio Luzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy.,Metabolism Research Centre, IRCCS Policlinico San Donato, Milan, Italy
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Glenn A Halff
- Department of Surgery, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Ralph A DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA.,Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA.,Department of Health Science, University of Milan, Milan, Italy
| |
Collapse
|
5
|
Higgins PB, Folli F, Andrade MCR, Foster J, Mattern V, Paroni R, Schlabritz-Loutsevitch N, Voruganti VS, Kumar S, Guardado-Mendoza R, Bulfamante G, Fiorina P, Pontiroli AE, Hubbard GB, Owston M, Dick EJ, Comuzzie AG. Duodenal adipose tissue is associated with obesity in baboons (Papio sp): a novel site of ectopic fat deposition in non-human primates. Acta Diabetol 2019; 56:227-236. [PMID: 30673859 PMCID: PMC6691506 DOI: 10.1007/s00592-019-01286-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022]
Abstract
AIMS Ectopic fat is a recognized contributor to insulin resistance and metabolic dysfunction, while the role of fat deposition inside intestinal wall tissue remains understudied. We undertook this study to directly quantify and localize intramural fat deposition in duodenal tissue and determine its association with adiposity. METHODS Duodenal tissues were collected from aged (21.2 ± 1.3 years, 19.5 ± 3.1 kg, n = 39) female baboons (Papio sp.). Fasted blood was collected for metabolic profiling and abdominal circumference (AC) measurements were taken. Primary tissue samples were collected at the major duodenal papilla at necropsy: one full cross section was processed for hematoxylin and eosin staining and evaluated; a second full cross section was processed for direct chemical lipid analysis on which percentage duodenal fat content was calculated. RESULTS Duodenal fat content obtained by direct tissue quantification showed considerable variability (11.95 ± 6.93%) and was correlated with AC (r = 0.60, p < 0.001), weight (r = 0.38, p = 0.02), leptin (r = 0.63, p < 0.001), adiponectin (r = - 0.32, p < 0.05), and triglyceride (r = 0.41, p = 0.01). The relationship between duodenal fat content and leptin remained after adjusting for body weight and abdominal circumference. Intramural adipocytes were found in duodenal sections from all animals and were localized to the submucosa. Consistent with the variation in tissue fat content, the submucosal adipocytes were non-uniformly distributed in clusters of varying size. Duodenal adipocytes were larger in obese vs. lean animals (106.9 vs. 66.7 µm2, p = 0.02). CONCLUSIONS Fat accumulation inside the duodenal wall is strongly associated with adiposity and adiposity related circulating biomarkers in baboons. Duodenal tissue fat represents a novel and potentially metabolically active site of ectopic fat deposition.
Collapse
Affiliation(s)
- Paul B Higgins
- Department of Genetics, Texas Biomedical Research Institute, PO Box 760549, San Antonio, TX, 78245-0549, USA.
| | - Franco Folli
- Endocrinology and Metabolism, Department of Health Science, University of Milan, Via A. di Rudini, 8, 20142, Milan, Italy.
- UOSD of Diabetes and Metabolic Disorders, ASST Santi Paolo e Carlo, Milan, Italy.
| | - Marcia C R Andrade
- Center for Laboratory Animal Breeding, Oswaldo Cruz Foundation, Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Jaydee Foster
- Department of Genetics, Texas Biomedical Research Institute, PO Box 760549, San Antonio, TX, 78245-0549, USA
| | - Vicki Mattern
- Department of Genetics, Texas Biomedical Research Institute, PO Box 760549, San Antonio, TX, 78245-0549, USA
| | - Rita Paroni
- Laboratory of Clinical Biochemistry and Mass Spectrometry, Department of Health Science, University of Milan, Milan, Italy
| | - Natalia Schlabritz-Loutsevitch
- Department of Obstetrics and Gynecology, School of Medicine, Texas Tech University Health Sciences Center at the Permian Basin, Odessa, TX, USA
| | - V Saroja Voruganti
- Nutrition Research Institute, Department of Nutrition, University of North Carolina at Chapel Hill, Kannapolis, NC, USA
| | - Shyamesh Kumar
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Gaetano Bulfamante
- Pathological Anatomy, Department of Health Science, University of Milano, Via A. di Rudini' 8, 20142, Milan, Italy
- ASST Santi Paolo e Carlo, Milan, Italy
| | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences, "L. Sacco", University of Milan, Milan, Italy
| | | | - Gene B Hubbard
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Michael Owston
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | |
Collapse
|
6
|
Blanco CL, Gastaldelli A, Anzueto DG, Winter LA, Seidner SR, McCurnin DC, Liang H, Javors MA, DeFronzo RA, Musi N. Effects of intravenous AICAR (5-aminoimidazole-4-carboximide riboside) administration on insulin signaling and resistance in premature baboons, Papio sp. PLoS One 2018; 13:e0208757. [PMID: 30540820 PMCID: PMC6291136 DOI: 10.1371/journal.pone.0208757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022] Open
Abstract
Premature baboons exhibit peripheral insulin resistance and impaired insulin signaling. 5' AMP-activated protein kinase (AMPK) activation improves insulin sensitivity by enhancing glucose uptake (via increased glucose transporter type 4 [GLUT4] translocation and activation of the extracellular signal-regulated kinase [ERK]/ atypical protein kinase C [aPKC] pathway), and increasing fatty acid oxidation (via inhibition of acetyl-CoA carboxylase 1 [ACC]), while downregulating gluconeogenesis (via induction of small heterodimer partner [SHP] and subsequent downregulation of the gluconeogenic enzymes: phosphoenolpyruvate carboxykinase [PEPCK], glucose 6-phosphatase [G6PASE], fructose- 1,6-bisphosphatase 1 [FBP1], and forkhead box protein 1 [FOXO1]). The purpose of this study was to investigate whether pharmacologic activation of AMPK with AICAR (5-aminoimidazole-4-carboximide riboside) administration improves peripheral insulin sensitivity in preterm baboons. 11 baboons were delivered prematurely at 125±2 days (67%) gestation. 5 animals were randomized to receive 5 days of continuous AICAR infusion at a dose of 0.5 mg·g-1·day-1. 6 animals were in the placebo group. Euglycemic hyperinsulinemic clamps were performed at 5±2 and 14±2 days of life. Key molecules potentially altered by AICAR (AMPK, GLUT4, ACC, PEPCK, G6PASE, FBP1, and FOXO1), and the insulin signaling molecules: insulin receptor (INSR), insulin receptor substrate 1 (IRS-1), protein kinase B (AKT), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) were measured using RT-PCR and western blotting. AICAR infusion did not improve whole body insulin-stimulated glucose disposal in preterm baboons (12.8±2.4 vs 12.4±2.0 mg/(kg·min), p = 0.8, placebo vs AICAR). One animal developed complications during treatment. In skeletal muscle, AICAR infusion did not increase phosphorylation of ACC, AKT, or AMPK whereas it increased mRNA expression of ACACA (ACC), AKT, and PPARGC1A (PGC1α). In the liver, INSR, IRS1, G6PC3, AKT, PCK1, FOXO1, and FBP1 were unchanged, whereas PPARGC1A mRNA expression increased after AICAR infusion. This study provides evidence that AICAR does not improve insulin sensitivity in premature euglycemic baboons, and may have adverse effects.
Collapse
Affiliation(s)
- Cynthia L. Blanco
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Amalia Gastaldelli
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Institute of Clinical Physiology Consiglio Nazionale delle Ricerche, Pisa Italy
| | - Diana G. Anzueto
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Lauryn A. Winter
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Steven R. Seidner
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Donald C. McCurnin
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Hanyu Liang
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Martin A. Javors
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Ralph A. DeFronzo
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Texas Diabetes Institute, San Antonio, TX, United States of America
| | - Nicolas Musi
- Department of Medicine, Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Texas Diabetes Institute, San Antonio, TX, United States of America
- Sam and Ann Barshop Institute for Longevity and Aging Studies, San Antonio, TX, United States of America
| |
Collapse
|
7
|
Bifari F, Manfrini R, Dei Cas M, Berra C, Siano M, Zuin M, Paroni R, Folli F. Multiple target tissue effects of GLP-1 analogues on non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Pharmacol Res 2018; 137:219-229. [PMID: 30359962 DOI: 10.1016/j.phrs.2018.09.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/11/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022]
Abstract
Accumulating experimental and clinical evidences over the last decade indicate that GLP-1 analogues have a series of central nervous system and peripheral target tissues actions which are able to significantly influence the liver metabolism. GLP-1 analogues pleiotropic effects proved to be efficacious in T2DM subjects not only reducing liver steatosis and ameliorating NAFLD and NASH, but also in lowering plasma glucose and liver inflammation, improving cardiac function and protecting from kidney dysfunction. While the experimental and clinical data are robust, the precise mechanisms of action potentially involved in these protective multi-target effects need further investigation. Here we present a systematic review of the most recent literature data on the multi-target effects of GLP-1 analogues on the liver, on adipose and muscular tissue and on the nervous system, all capable of influencing significant aspects of the fatty liver disease physiopathology. From this analysis, we can conclude that the multi-target beneficial action of the GLP-1 analogues could explain the positive effects observed in animal and human models on progression of NAFLD to NASH.
Collapse
Affiliation(s)
- Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Roberto Manfrini
- Department of Internal Medicine ASST Santi Paolo e Carlo, Milan, Italy
| | - Michele Dei Cas
- Laboratory of Clinical Biochemistry and Mass Spectrometry, Department of Health Science, University of Milan, Milan, Italy
| | - Cesare Berra
- Metabolic Disease and Diabetes, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Matteo Siano
- Department of Internal Medicine ASST Santi Paolo e Carlo, Milan, Italy
| | - Massimo Zuin
- Unit of Medicine, Gastroenterology and Hepatology, Milan, Italy
| | - Rita Paroni
- Laboratory of Clinical Biochemistry and Mass Spectrometry, Department of Health Science, University of Milan, Milan, Italy
| | - Franco Folli
- Unit of Endocrinology and Metabolism ASST Santi Paolo e Carlo, Department of Health Science, University of Milan, Milan, Italy.
| |
Collapse
|
8
|
Folli F, La Rosa S, Finzi G, Davalli AM, Galli A, Dick EJ, Perego C, Mendoza RG. Pancreatic islet of Langerhans' cytoarchitecture and ultrastructure in normal glucose tolerance and in type 2 diabetes mellitus. Diabetes Obes Metab 2018; 20 Suppl 2:137-144. [PMID: 30230173 DOI: 10.1111/dom.13380] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/20/2018] [Accepted: 05/23/2018] [Indexed: 01/19/2023]
Abstract
While a number of structural and cellular abnormalities occur in the islet of Langerhans in diabetes, and in particular in type 2 diabetes, the focus has been mostly on the insulin producing β-cells and only more recently on glucagon producing α- and δ-cells. There is ample evidence that in type 2 diabetes mellitus (T2DM), in addition to a progressive decline in β-cell function and associated insulin resistance in a number of insulin-sensitive tissues, alterations in glucagon secretion are also present and may play an important role in the pathogenesis of hyperglycemia both in the fasting and in the postprandial state. Recently, a number of studies have showed that there are also functional and structural alterations in glucagon-producing α-cells and somatostatin-producing δ-cells. Thus, it is becoming increasingly clear that multiple cellular alterations of multiple cell types occur, which adds even more complexity to our understanding of the pathophysiology of this common and severe disease. We believe that persistent efforts to increase the understanding of the pathophysiology of hormone secretion in the islets of Langerhans will also improve our capability to better prevent and treat diabetes mellitus.
Collapse
Affiliation(s)
- Franco Folli
- Endocrinology and Metabolism, Department of Health Science, University of Milano, Milan, Italy
- Department of Medicine, Endocrinology and Metabolism, Azienda Socio-Sanitaria Santi Paolo e Carlo, Milan, Italy
| | - Stefano La Rosa
- Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Giovanna Finzi
- Anatomical Pathology, Ospedale di Circolo Varese, Varese, Italy
| | - Alberto M Davalli
- Department of Medicine, Endocrinology and Metabolism, H.S Raffaele, Milan, Italy
| | - Alessandra Galli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Carla Perego
- Anatomical Pathology, Ospedale di Circolo Varese, Varese, Italy
| | - Rodolfo Guardado Mendoza
- Division of Health Sciences, Department of Medicine and Nutrition, University of Guanajuato, Guanajuato, Mexico
- Departamento de Investigación, Hospital Regional de Alta Especialidad del Bajío, Guanajuato, Mexico
| |
Collapse
|
9
|
Helminth infection in mice improves insulin sensitivity via modulation of gut microbiota and fatty acid metabolism. Pharmacol Res 2018; 132:33-46. [PMID: 29653264 DOI: 10.1016/j.phrs.2018.04.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/14/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022]
Abstract
Intestinal helminths are prevalent in individuals who live in rural areas of developing countries, where obesity, type 2 diabetes, and metabolic syndrome are rare. In the present study, we analyzed the modulation of the gut microbiota in mice infected with the helminth Strongyloides venezuelensis, and fed either a standard rodent chow diet or high-fat diet (HFD). To investigate the effects of the microbiota modulation on the metabolism, we analyzed the expression of tight-junction proteins present in the gut epithelium, inflammatory markers in the serum and tissue and quantified glucose tolerance and insulin sensitivity and resistance. Additionally, the levels of lipids related to inflammation were evaluated in the feces and serum. Our results show that infection with Strongyloides venezuelensis results in a modification of the gut microbiota, most notably by increasing Lactobacillus spp. These modifications in the microbiota alter the host metabolism by increasing the levels of anti-inflammatory cytokines, switching macrophages from a M1 to M2 pattern in the adipose tissue, increasing the expression of tight junction proteins in the intestinal cells (thereby reducing the permeability) and decreasing LPS in the serum. Taken together, these changes correlate with improved insulin signaling and sensitivity, which could also be achieved with HFD mice treated with probiotics. Additionally, helminth infected mice produce higher levels of oleic acid, which participates in anti-inflammatory pathways. These results suggest that modulation of the microbiota by helminth infection or probiotic treatment causes a reduction in subclinical inflammation, which has a positive effect on the glucose metabolism of the host.
Collapse
|
10
|
Andrew MS, Huffman DM, Rodriguez-Ayala E, Williams NN, Peterson RM, Bastarrachea RA. Mesenteric visceral lipectomy using tissue liquefaction technology reverses insulin resistance and causes weight loss in baboons. Surg Obes Relat Dis 2018; 14:833-841. [PMID: 29631983 DOI: 10.1016/j.soard.2018.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/04/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Visceral obesity is associated with diabetogenic and atherogenic abnormalities, including insulin resistance and increased risk for cardiometabolic diseases and mortality. Rodent lipectomy studies have demonstrated a causal link between visceral fat and insulin resistance, yet human omentectomy studies have failed to replicate this metabolic benefit, perhaps owing to the inability to target the mesentery. OBJECTIVES We aimed to demonstrate that safe and effective removal of mesenteric fat could be achieved in obese insulin-resistant baboons using tissue liquefaction technology. SETTING Southwest National Primate Research Center, San Antonio, Texas. METHODS Tissue liquefaction technology has been developed to enable mesenteric visceral lipectomy (MVL) to be safely performed without disturbing the integrity of surrounding nerves and vessels in the mesentary. After an initial MVL optimization study (n = 3), we then performed MVL (n = 4) or sham surgery (n = 2) in a cohort of insulin-resistant baboons, and the metabolic phenotype was assessed via hyperinsulinemic-euglycemic clamps at baseline and 6 weeks later. RESULTS MVL led to a 75% improvement in glucose disposal at 6-weeks follow-up (P = .01). Moreover, despite removing only an average of 430 g of mesenteric fat (~1% of total body mass), MVL led to a 14.4% reduction in total weight (P = .001). Thus, these data demonstrate that mesenteric fat can be safely targeted for removal by tissue liquefaction technology in a nonhuman primate, leading to substantial metabolic improvements, including reversal of insulin resistance and weight loss. CONCLUSIONS These data provide the first demonstration of successful adipose tissue removal from the mesentery in a mammal. Importantly, we have demonstrated that when MVL is performed in obese, insulin-resistant baboons, insulin resistance is reversed, and significant weight loss occurs. Therefore, trials performing MVL in humans with abdominal obesity and related metabolic sequelae should be explored as a potential clinical tool to ameliorate insulin resistance and treat type 2 diabetes.
Collapse
Affiliation(s)
| | - Derek M Huffman
- Departments of Molecular Pharmacology and Medicine, The Albert Einstein College of Medicine, Bronx, New York
| | - Ernesto Rodriguez-Ayala
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac Norte, México
| | - Noel N Williams
- Department of Metabolic and Bariatric Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard M Peterson
- Department of Bariatric Surgery, University of Texas Health Sciences Center, San Antonio, Texas
| | - Raul A Bastarrachea
- Department of Genetics and Southwest National Primate Research Center (SNPRC), Texas Biomedical Research Institute, San Antonio, Texas.
| |
Collapse
|
11
|
Havel PJ, Kievit P, Comuzzie AG, Bremer AA. Use and Importance of Nonhuman Primates in Metabolic Disease Research: Current State of the Field. ILAR J 2017; 58:251-268. [PMID: 29216341 PMCID: PMC6074797 DOI: 10.1093/ilar/ilx031] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 10/13/2017] [Accepted: 10/22/2017] [Indexed: 12/16/2022] Open
Abstract
Obesity and its multiple metabolic sequelae, including type 2 diabetes, cardiovascular disease, and fatty liver disease, are becoming increasingly widespread in both the developed and developing world. There is an urgent need to identify new approaches for the prevention and treatment of these costly and prevalent metabolic conditions. Accomplishing this will require the use of appropriate animal models for preclinical and translational investigations in metabolic disease research. Although studies in rodent models are often useful for target/pathway identification and testing hypotheses, there are important differences in metabolic physiology between rodents and primates, and experimental findings in rodent models have often failed to be successfully translated into new, clinically useful therapeutic modalities in humans. Nonhuman primates represent a valuable and physiologically relevant model that serve as a critical translational bridge between basic studies performed in rodent models and clinical studies in humans. The purpose of this review is to evaluate the evidence, including a number of specific examples, in support of the use of nonhuman primate models in metabolic disease research, as well as some of the disadvantages and limitations involved in the use of nonhuman primates. The evidence taken as a whole indicates that nonhuman primates are and will remain an indispensable resource for evaluating the efficacy and safety of novel therapeutic strategies targeting clinically important metabolic diseases, including dyslipidemia and atherosclerosis, type 2 diabetes, hepatic steatosis, steatohepatitis, and hepatic fibrosis, and potentially the cognitive decline and dementia associated with metabolic dysfunction, prior to taking these therapies into clinical trials in humans.
Collapse
Affiliation(s)
- Peter J Havel
- Peter J. Havel, DVM, PhD, is a professor in the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, California National Primate Research Center, University of California, Davis, California. Paul Kievit, PhD, is an assistant professor at Oregon Health & Sciences University, Portland, Oregon and Director of the Obese NHP Resource at the Oregon National Primate Research Center, Beaverton, Oregon. Anthony G. Comuzzie, PhD, is a senior scientist at the Southwest National Primate Research Center and the Department of Genetics at the Texas Biomedical Research Institute, San Antonio, Texas and currently the Executive Director of The Obesity Society, Silver Springs, Maryland. Andrew A. Bremer, MD, PhD, is Scientific Program Director in the Division of Diabetes, Endocrinology and Metabolic Diseases at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Paul Kievit
- Peter J. Havel, DVM, PhD, is a professor in the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, California National Primate Research Center, University of California, Davis, California. Paul Kievit, PhD, is an assistant professor at Oregon Health & Sciences University, Portland, Oregon and Director of the Obese NHP Resource at the Oregon National Primate Research Center, Beaverton, Oregon. Anthony G. Comuzzie, PhD, is a senior scientist at the Southwest National Primate Research Center and the Department of Genetics at the Texas Biomedical Research Institute, San Antonio, Texas and currently the Executive Director of The Obesity Society, Silver Springs, Maryland. Andrew A. Bremer, MD, PhD, is Scientific Program Director in the Division of Diabetes, Endocrinology and Metabolic Diseases at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Anthony G Comuzzie
- Peter J. Havel, DVM, PhD, is a professor in the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, California National Primate Research Center, University of California, Davis, California. Paul Kievit, PhD, is an assistant professor at Oregon Health & Sciences University, Portland, Oregon and Director of the Obese NHP Resource at the Oregon National Primate Research Center, Beaverton, Oregon. Anthony G. Comuzzie, PhD, is a senior scientist at the Southwest National Primate Research Center and the Department of Genetics at the Texas Biomedical Research Institute, San Antonio, Texas and currently the Executive Director of The Obesity Society, Silver Springs, Maryland. Andrew A. Bremer, MD, PhD, is Scientific Program Director in the Division of Diabetes, Endocrinology and Metabolic Diseases at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Andrew A Bremer
- Peter J. Havel, DVM, PhD, is a professor in the Department of Molecular Biosciences, School of Veterinary Medicine and Department of Nutrition, California National Primate Research Center, University of California, Davis, California. Paul Kievit, PhD, is an assistant professor at Oregon Health & Sciences University, Portland, Oregon and Director of the Obese NHP Resource at the Oregon National Primate Research Center, Beaverton, Oregon. Anthony G. Comuzzie, PhD, is a senior scientist at the Southwest National Primate Research Center and the Department of Genetics at the Texas Biomedical Research Institute, San Antonio, Texas and currently the Executive Director of The Obesity Society, Silver Springs, Maryland. Andrew A. Bremer, MD, PhD, is Scientific Program Director in the Division of Diabetes, Endocrinology and Metabolic Diseases at the National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
12
|
Garbossa SG, Folli F. Vitamin D, sub-inflammation and insulin resistance. A window on a potential role for the interaction between bone and glucose metabolism. Rev Endocr Metab Disord 2017; 18:243-258. [PMID: 28409320 DOI: 10.1007/s11154-017-9423-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vitamin D is a key hormone involved in the regulation of calcium/phosphorous balance and recently it has been implicated in the pathogenesis of sub-inflammation, insulin resistance and obesity. The two main forms of vitamin D are cholecalciferol (Vitamin D3) and ergocalciferol (Vitamin D2): the active form (1,25-dihydroxyvitamin D) is the result of two hydroxylations that take place in liver, kidney, pancreas and immune cells. Vitamin D increases the production of some anti-inflammatory cytokines and reduces the release of some pro-inflammatory cytokines. Low levels of Vitamin D are also associated with an up-regulation of TLRs expression and a pro-inflammatory state. Regardless of the effect on inflammation, Vitamin D seems to directly increase insulin sensitivity and secretion, through different mechanisms. Considering the importance of low grade chronic inflammation in metabolic syndrome, obesity and diabetes, many authors hypothesized the involvement of this nutrient/hormone in the pathogenesis of these diseases. Vitamin D status could alter the balance between pro and anti-inflammatory cytokines and thus affect insulin action, lipid metabolism and adipose tissue function and structure. Numerous studies have shown that Vitamin D concentrations are inversely associated with pro-inflammatory markers, insulin resistance, glucose intolerance and obesity. Interestingly, some longitudinal trials suggested also an inverse association between vitamin D status and incident type 2 diabetes mellitus. However, vitamin D supplementation in humans showed controversial effects: with some studies demonstrating improvements in insulin sensitivity, glucose and lipid metabolism while others showing no beneficial effect on glycemic control and on inflammation. In conclusion, although the evidences of a significant role of Vitamin D on inflammation, insulin resistance and insulin secretion in the pathogenesis of obesity, metabolic syndrome and type 2 diabetes, its potential function in treatment and prevention of type 2 diabetes mellitus is unclear. Encouraging results have emerged from Vitamin D supplementation trials on patients at risk of developing diabetes and further studies are needed to fully explore and understand its clinical applications.
Collapse
Affiliation(s)
- Stefania Giuliana Garbossa
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy
- Program in Nutritional Science, Universitá degli Studi di Milano, Milan, Italy
| | - Franco Folli
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy.
- Endocrinology and Metabolism, Department of Health Science, Universitá degli Studi di Milano, San Paolo Hospital, Room #737, Via A. Di Rudiní 8, 20142, Milan, Italy.
- Department of Medicine, Diabetes Division, University of Texas Health System, 7703 Floyd Curl Drive, San Antonio, 78229, TX, USA.
| |
Collapse
|
13
|
Kochunov P, Wey HY, Fox PT, Lancaster JL, Davis MD, Wang DJJ, Lin AL, Bastarrachea RA, Andrade MCR, Mattern V, Frost P, Higgins PB, Comuzzie AG, Voruganti VS. Changes in Cerebral Blood Flow during an Alteration in Glycemic State in a Large Non-human Primate ( Papio hamadryas sp.). Front Neurosci 2017; 11:49. [PMID: 28261040 PMCID: PMC5306336 DOI: 10.3389/fnins.2017.00049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/23/2017] [Indexed: 11/24/2022] Open
Abstract
Changes in cerebral blood flow (CBF) during a hyperglycemic challenge were mapped, using perfusion-weighted MRI, in a group of non-human primates. Seven female baboons were fasted for 16 h prior to 1-h imaging experiment, performed under general anesthesia, that consisted of a 20-min baseline, followed by a bolus infusion of glucose (500 mg/kg). CBF maps were collected every 7 s and blood glucose and insulin levels were sampled at regular intervals. Blood glucose levels rose from 51.3 ± 10.9 to 203.9 ± 38.9 mg/dL and declined to 133.4 ± 22.0 mg/dL, at the end of the experiment. Regional CBF changes consisted of four clusters: cerebral cortex, thalamus, hypothalamus, and mesencephalon. Increases in the hypothalamic blood flow occurred concurrently with the regulatory response to systemic glucose change, whereas CBF declined for other clusters. The return to baseline of hypothalamic blood flow was observed while CBF was still increasing in other brain regions. The spatial pattern of extra-hypothalamic CBF changes was correlated with the patterns of several cerebral networks including the default mode network. These findings suggest that hypothalamic blood flow response to systemic glucose levels can potentially be explained by regulatory activity. The response of extra-hypothalamic clusters followed a different time course and its spatial pattern resembled that of the default-mode network.
Collapse
Affiliation(s)
- Peter Kochunov
- Maryland Psychiatric Research Center, University of Maryland School of MedicineBaltimore, MA, USA; Research Imaging Institute, University of Texas Health Science Center at San AntonioSan Antonio, TX, USA; Southwest National Primate Research CenterSan Antonio, TX, USA
| | - Hsiao-Ying Wey
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan Antonio, TX, USA; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolCharlestown, MA, USA
| | - Peter T Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Jack L Lancaster
- Research Imaging Institute, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Michael D Davis
- Research Imaging Institute, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Danny J J Wang
- Ahmanson-Lovelace Brain Mapping Center, University of California at Los AngelesLos Angeles, CA, USA; Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos Angeles, CA, USA
| | - Ai-Ling Lin
- Research Imaging Institute, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Raul A Bastarrachea
- Southwest National Primate Research CenterSan Antonio, TX, USA; Department of Genetics, Texas Biomedical Research InstituteSan Antonio, TX, USA
| | - Marcia C R Andrade
- Department of Genetics, Texas Biomedical Research InstituteSan Antonio, TX, USA; Center for Laboratory Animal Breeding, Oswaldo Cruz FoundationRio de Janeiro, Brazil
| | - Vicki Mattern
- Department of Genetics, Texas Biomedical Research Institute San Antonio, TX, USA
| | - Patrice Frost
- Southwest National Primate Research Center San Antonio, TX, USA
| | - Paul B Higgins
- Department of Genetics, Texas Biomedical Research Institute San Antonio, TX, USA
| | - Anthony G Comuzzie
- Southwest National Primate Research CenterSan Antonio, TX, USA; Department of Genetics, Texas Biomedical Research InstituteSan Antonio, TX, USA
| | - Venkata S Voruganti
- Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill Kannapolis, NC, USA
| |
Collapse
|
14
|
Pepe GJ, Maniu A, Aberdeen G, Lynch TJ, Kim SO, Nadler J, Albrecht ED. Insulin resistance elicited in postpubertal primate offspring deprived of estrogen in utero. Endocrine 2016; 54:788-797. [PMID: 27770396 PMCID: PMC6038696 DOI: 10.1007/s12020-016-1145-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/05/2016] [Indexed: 12/25/2022]
Abstract
We recently demonstrated that offspring delivered to baboons deprived of estrogen during the second half of gestation exhibited insulin resistance prior to onset of puberty. Because gonadal hormones have a profound effect on insulin action and secretion in adults, we determined whether insulin resistance is retained after initiation of gonadal secretion of testosterone and estradiol. Glucose tolerance tests were performed in postpubertal baboon offspring of untreated and letrozole-treated animals (serum estradiol reduced >95 %). Basal fasting levels of insulin (P < 0.05) and peak 1 min and 1 + 3 + 5 min levels of glucose after glucose tolerance tests challenge (P < 0.03) were greater in offspring delivered to letrozole-treated, estrogen-deprived baboons than untreated animals. Moreover, the value for the HOMA-IR, an accepted index of insulin resistance, was 2-fold greater (P < 0.05) in offspring delivered to baboons treated with letrozole than in untreated animals. Collectively these results support the proposal that estrogen normally has an important role in programming mechanisms in utero within the developing fetus that lead to insulin sensitivity after birth.
Collapse
Affiliation(s)
- Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| | - Adina Maniu
- Department of Obstetrics/Gynecology/Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Graham Aberdeen
- Department of Obstetrics/Gynecology/Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Terrie J Lynch
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Jerry Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Eugene D Albrecht
- Departments of Obstetrics/Gynecology/Reproductive Sciences and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Higgins PB, Shade RE, Rodríguez-Sánchez IP, Garcia-Forey M, Tejero ME, Voruganti VS, Cole SA, Comuzzie AG, Folli F. Central GIP signaling stimulates peripheral GIP release and promotes insulin and pancreatic polypeptide secretion in nonhuman primates. Am J Physiol Endocrinol Metab 2016; 311:E661-E670. [PMID: 27530231 PMCID: PMC5241561 DOI: 10.1152/ajpendo.00166.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/10/2016] [Indexed: 01/07/2023]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) has important actions on whole body metabolic function. GIP and its receptor are also present in the central nervous system and have been linked to neurotrophic actions. Metabolic effects of central nervous system GIP signaling have not been reported. We investigated whether centrally administered GIP could increase peripheral plasma GIP concentrations and influence the metabolic response to a mixed macronutrient meal in nonhuman primates. An infusion and sampling system was developed to enable continuous intracerebroventricular (ICV) infusions with serial venous sampling in conscious nonhuman primates. Male baboons (Papio sp.) that were healthy and had normal body weights (28.9 ± 2.1 kg) were studied (n = 3). Animals were randomized to receive continuous ICV infusions of GIP (20 pmol·kg-1·h-1) or vehicle before and over the course of a 300-min mixed meal test (15 kcal/kg, 1.5g glucose/kg) on two occasions. A significant increase in plasma GIP concentration was observed under ICV GIP infusion (66.5 ± 8.0 vs. 680.6 ± 412.8 pg/ml, P = 0.04) before administration of the mixed meal. Increases in postprandial, but not fasted, insulin (P = 0.01) and pancreatic polypeptide (P = 0.04) were also observed under ICV GIP. Effects of ICV GIP on fasted or postprandial glucagon, glucose, triglyceride, and free fatty acids were not observed. Our data demonstrate that central GIP signaling can promote increased plasma GIP concentrations independent of nutrient stimulation and increase insulin and pancreatic polypeptide responses to a mixed meal.
Collapse
Affiliation(s)
- Paul B Higgins
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas; Southwest National Primate Research Center, San Antonio, Texas;
| | - Robert E Shade
- Southwest National Primate Research Center, San Antonio, Texas
| | - Irám P Rodríguez-Sánchez
- Department of Genetics, School of Medicine, Autonomous University of Nuevo León (Universidad Autonoma de Nuevo León), Monterrey, Nuevo León, Mexico
| | | | - M Elizabeth Tejero
- Laboratory of Nutrigenetics and Nutrigenomics, National Institute of Genomic Medicine (Instituto Nacional de Medicina Genómica), Mexico City, Mexico
| | - V Saroja Voruganti
- Department of Nutrition and UNC Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina
| | - Shelley A Cole
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas; Southwest National Primate Research Center, San Antonio, Texas
| | - Franco Folli
- Southwest National Primate Research Center, San Antonio, Texas; Diabetes Division, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas; Department of Medicine, Obesity and Comorbidities Research Center, University of Campinas, Campinas, São Paulo, Brazil; and Dipartimento di Scienze della Salute, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
16
|
Maniu A, Aberdeen GW, Lynch TJ, Nadler JL, Kim SOK, Quon MJ, Pepe GJ, Albrecht ED. Estrogen deprivation in primate pregnancy leads to insulin resistance in offspring. J Endocrinol 2016; 230:171-83. [PMID: 27207093 PMCID: PMC4946970 DOI: 10.1530/joe-15-0530] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 12/29/2022]
Abstract
This study tested the hypothesis that estrogen programs mechanisms within the primate fetus that promote insulin sensitivity and glucose homeostasis in offspring. Glucose tolerance tests were performed longitudinally in prepubertal offspring of baboons untreated or treated on days 100 to 165/175 of gestation (term is 184 days) with the aromatase inhibitor letrozole, which decreased fetal estradiol levels by 95%. Basal plasma insulin levels were over two-fold greater in offspring delivered to letrozole-treated than untreated animals. Moreover, the peak 1min, average of the 1, 3, and 5min, and area under the curve blood glucose and plasma insulin levels after an i.v. bolus of glucose were greater (P<0.05 and P<0.01, respectively) in offspring deprived of estrogen in utero than in untreated animals and partially or completely restored in letrozole plus estradiol-treated baboons. The value for the homeostasis model assessment of insulin resistance was 2.5-fold greater (P<0.02) and quantitative insulin sensitivity check index lower (P<0.01) in offspring of letrozole-treated versus untreated animals and returned to almost normal in letrozole plus estradiol-treated animals. The exaggerated rise in glucose and insulin levels after glucose challenge in baboon offspring deprived of estrogen in utero indicates that pancreatic beta cells had the capacity to secrete insulin, but that peripheral glucose uptake and/or metabolism were impaired, indicative of insulin resistance and glucose intolerance. We propose that estrogen normally programs mechanisms in utero within the developing primate fetus that lead to insulin sensitivity, normal glucose tolerance, and the capacity to metabolize glucose after birth.
Collapse
Affiliation(s)
- Adina Maniu
- Department of ObstetricsGynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Graham W Aberdeen
- Department of ObstetricsGynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Terrie J Lynch
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia, USA
| | - Jerry L Nadler
- Department of Internal MedicineEastern Virginia Medical School, Norfolk, Virginia, USA
| | - Soon O K Kim
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia, USA
| | - Michael J Quon
- Department of MedicineUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gerald J Pepe
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia, USA
| | - Eugene D Albrecht
- Department of ObstetricsGynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Andreozzi F, Raciti GA, Nigro C, Mannino GC, Procopio T, Davalli AM, Beguinot F, Sesti G, Miele C, Folli F. The GLP-1 receptor agonists exenatide and liraglutide activate Glucose transport by an AMPK-dependent mechanism. J Transl Med 2016; 14:229. [PMID: 27473212 PMCID: PMC4967343 DOI: 10.1186/s12967-016-0985-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/20/2016] [Indexed: 01/11/2023] Open
Abstract
Aims/hypothesis Potentiation of glucose-induced insulin secretion is the main mechanism of exenatide (EXE) antidiabetic action, however, increased glucose utilization by peripheral tissues has been also reported. We here studied the effect of EXE on glucose uptake by skeletal muscle cells. Methods 2-deoxy-glucose (2DG) uptake and intracellular signal pathways were measured in rat L6 skeletal muscle myotubes exposed to 100 nmol/l EXE for up to 48 h. Mechanisms of EXE action were explored by inhibiting AMPK activity with compound C (CC, 40 μmol/l) or siRNAs (2 μmol/l). Results Time course experiments show that EXE increases glucose uptake up to 48 h achieving its maximal effect, similar to that induced by insulin, after 20 min (2- vs 2.5-fold-increase, respectively). Differently from insulin, EXE does not stimulate: (i) IR β-subunit- and IRS1 tyrosine phosphorylation and binding to p85 regulatory subunit of PI-3kinase; (ii) AKT activation; and (iii) ERK1/2 and JNK1/2 phosphorylation. Conversely, EXE increases phosphorylation of α-subunit of AMPK at Thr172 by 2.5-fold (p < 0.01). Co-incubation of EXE and insulin does not induce additive effects on 2DG-uptake. Inhibition of AMPK with CC, and reduction of AMPK protein expression by siRNA, completely abolish EXE-induced 2DG-uptake. Liraglutide, another GLP-1 receptor agonist, also stimulates AMPK phosphorylation and 2DG-uptake. Moreover, EXE stimulates 2DG-uptake also by L6 myotubes rendered insulin-resistant with methylglyoxal. Finally, EXE also induces glucose transporter Glut-4 translocation to the plasma membrane. Conclusions/interpretation In L6 myotubes, EXE and liraglutide increase glucose uptake in an insulin-independent manner by activating AMPK. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0985-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francesco Andreozzi
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy. .,Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Gregory Alexander Raciti
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Cecilia Nigro
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Gaia Chiara Mannino
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Teresa Procopio
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Alberto M Davalli
- Department of Medicine Endocrinology Unit, Ospedale San Raffaele, Milan, Italy
| | - Francesco Beguinot
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giorgio Sesti
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna-Graecia", Catanzaro, Italy
| | - Claudia Miele
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA. .,Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
18
|
Owston MA, LaRue MK, Dick EJ, Ambrus A, Porter BF. Pancreatic neuroendocrine tumors in twelve baboons (Papio spp.). J Med Primatol 2016; 45:85-91. [PMID: 26899153 DOI: 10.1111/jmp.12210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic neuroendocrine tumors (PNETs) are rare in nonhuman primates and in humans. METHODS Twenty-one PNETs from twelve female baboons (Papio spp.) from the Southwest National Primate Research Center were evaluated using histopathology and immunohistochemistry. RESULTS Histologically, all tumors were benign and had neuroendocrine packeting. Immunohistochemical staining for synaptophysin and chromogranin was positive in all tumors evaluated (17/17). Insulin was positive in 16 of 21 tumors. Somatostatin was positive in 9 of 20 tumors. Multifocal staining for glucagon and pancreatic polypeptide was evident in a minority of tumors (6/20 and 2/17, respectively). Gastrin and vasoactive intestinal peptide were negative in all tumors evaluated. Nine tumors expressed more than one hormone marker. CONCLUSIONS This is the first detailed pathologic study of pancreatic endocrine tumors in the baboon. The findings suggest that these tumors are generally benign and have similar morphologic and immunohistochemical features as those described in people, including the ability to express multiple hormones.
Collapse
Affiliation(s)
- M A Owston
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - M K LaRue
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - E J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - A Ambrus
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - B F Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
19
|
Guardado Mendoza R, Perego C, Finzi G, La Rosa S, Capella C, Jimenez-Ceja LM, Velloso LA, Saad MJA, Sessa F, Bertuzzi F, Moretti S, Dick EJ, Davalli AM, Folli F. Delta cell death in the islet of Langerhans and the progression from normal glucose tolerance to type 2 diabetes in non-human primates (baboon, Papio hamadryas). Diabetologia 2015; 58:1814-26. [PMID: 26049399 PMCID: PMC5603258 DOI: 10.1007/s00125-015-3625-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/21/2015] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS The cellular composition of the islet of Langerhans is essential to ensure its physiological function. Morphophysiological islet abnormalities are present in type 2 diabetes but the relationship between fasting plasma glucose (FPG) and islet cell composition, particularly the role of delta cells, is unknown. We explored these questions in pancreases from baboons (Papio hamadryas) with FPG ranging from normal to type 2 diabetic values. METHODS We measured the volumes of alpha, beta and delta cells and amyloid in pancreatic islets of 40 baboons (Group 1 [G1]: FPG < 4.44 mmol/l [n = 10]; G2: FPG = 4.44-5.26 mmol/l [n = 9]; G3: FPG = 5.27-6.94 mmol/l [n = 9]; G4: FPG > 6.94 mmol/l [n = 12]) and correlated islet composition with metabolic and hormonal variables. We also performed confocal microscopy including TUNEL, caspase-3, and anti-caspase cleavage product of cytokeratin 18 (M30) immunostaining, electron microscopy, and immuno-electron microscopy with anti-somatostatin antibodies in baboon pancreases. RESULTS Amyloidosis preceded the decrease in beta cell volume. Alpha cell volume increased ∼ 50% in G3 and G4 (p < 0.05), while delta cell volume decreased in these groups by 31% and 39%, respectively (p < 0.05). In G4, glucagon levels were higher, while insulin and HOMA index of beta cell function were lower than in the other groups. Immunostaining of G4 pancreatic sections with TUNEL, caspase-3 and M30 showed apoptosis of beta and delta cells, which was also confirmed by immuno-electron microscopy with anti-somatostatin antibodies. CONCLUSIONS/INTERPRETATION In diabetic baboons, changes in islet composition correlate with amyloid deposition, with increased alpha cell and decreased beta and delta cell volume and number due to apoptosis. These data argue for an important role of delta cells in type 2 diabetes.
Collapse
Affiliation(s)
- Rodolfo Guardado Mendoza
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Frost PA, Chen S, Mezzles MJ, Voruganti VS, Nava-Gonzalez EJ, Arriaga-Cazares HE, Freed KA, Comuzzie AG, DeFronzo RA, Kent JW, Grayburn PA, Bastarrachea RA. Successful pharmaceutical-grade streptozotocin (STZ)-induced hyperglycemia in a conscious tethered baboon (Papio hamadryas) model. J Med Primatol 2015; 44:202-17. [PMID: 26122701 DOI: 10.1111/jmp.12182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Non-human primate (NHP) diabetic models using chemical ablation of β-cells with STZ have been achieved by several research groups. Chemotherapeutic STZ could lead to serious adverse events including nephrotoxicity, hepatotoxicity, and mortality. METHODS We implemented a comprehensive therapeutic strategy that included the tether system, permanent indwelling catheter implants, an aggressive hydration protocol, management for pain with IV nubain and anxiety with IV midazolam, moment-by-moment monitoring of glucose levels post-STZ administration, and continuous intravenous insulin therapy. RESULTS A triphasic response in blood glucose after STZ administration was fully characterized. A dangerous hypoglycemic phase was also detected in all baboons. Other significant findings were hyperglycemia associated with low levels of plasma leptin, insulin and C-peptide concentrations, hyperglucagonemia, and elevated non-esterified fatty acids (NEFA) concentrations. CONCLUSIONS We successfully induced frank diabetes by IV administering a single dose of pharmaceutical-grade STZ safely and without adverse events in conscious tethered baboons.
Collapse
Affiliation(s)
- Patrice A Frost
- Southwest National Primate Research Center, San Antonio, TX, USA
| | | | - Marguerite J Mezzles
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Edna J Nava-Gonzalez
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA.,University of Nuevo Leon School of Nutrition and Public Health, Monterrey, Mexico
| | - Hector E Arriaga-Cazares
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA.,Hospital Infantil de Tamaulipas, Ciudad Victoria, México
| | - Katy A Freed
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Anthony G Comuzzie
- Southwest National Primate Research Center, San Antonio, TX, USA.,Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Ralph A DeFronzo
- The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jack W Kent
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Paul A Grayburn
- Baylor Research Institute, Dallas, TX, USA.,Baylor University Medical Center, Dallas, TX, USA
| | - Raul A Bastarrachea
- Southwest National Primate Research Center, San Antonio, TX, USA.,Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
21
|
Velloso LA, Folli F, Saad MJ. TLR4 at the Crossroads of Nutrients, Gut Microbiota, and Metabolic Inflammation. Endocr Rev 2015; 36:245-71. [PMID: 25811237 DOI: 10.1210/er.2014-1100] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity is accompanied by the activation of low-grade inflammatory activity in metabolically relevant tissues. Studies have shown that obesity-associated insulin resistance results from the inflammatory targeting and inhibition of key proteins of the insulin-signaling pathway. At least three apparently distinct mechanisms-endoplasmic reticulum stress, toll-like receptor (TLR) 4 activation, and changes in gut microbiota-have been identified as triggers of obesity-associated metabolic inflammation; thus, they are expected to represent potential targets for the treatment of obesity and its comorbidities. Here, we review the data that place TLR4 in the center of the events that connect the consumption of dietary fats with metabolic inflammation and insulin resistance. Changes in the gut microbiota can lead to reduced integrity of the intestinal barrier, leading to increased leakage of lipopolysaccharides and fatty acids, which can act upon TLR4 to activate systemic inflammation. Fatty acids can also trigger endoplasmic reticulum stress, which can be further stimulated by cross talk with active TLR4. Thus, the current data support a connection among the three main triggers of metabolic inflammation, and TLR4 emerges as a link among all of these mechanisms.
Collapse
Affiliation(s)
- Licio A Velloso
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Franco Folli
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Mario J Saad
- Department of Internal Medicine (L.A.V., F.F., M.J.S.), University of Campinas, 13084-970 Campinas SP, Brazil; and Department of Medicine (F.F.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
22
|
Blanco CL, McGill-Vargas LL, Gastaldelli A, Seidner SR, McCurnin DC, Leland MM, Anzueto DG, Johnson MC, Liang H, DeFronzo RA, Musi N. Peripheral insulin resistance and impaired insulin signaling contribute to abnormal glucose metabolism in preterm baboons. Endocrinology 2015; 156:813-23. [PMID: 25560831 PMCID: PMC4330304 DOI: 10.1210/en.2014-1757] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Premature infants develop hyperglycemia shortly after birth, increasing their morbidity and death. Surviving infants have increased incidence of diabetes as young adults. Our understanding of the biological basis for the insulin resistance of prematurity and developmental regulation of glucose production remains fragmentary. The objective of this study was to examine maturational differences in insulin sensitivity and the insulin-signaling pathway in skeletal muscle and adipose tissue of 30 neonatal baboons using the euglycemic hyperinsulinemic clamp. Preterm baboons (67% gestation) had reduced peripheral insulin sensitivity shortly after birth (M value 12.5 ± 1.5 vs 21.8 ± 4.4 mg/kg · min in term baboons) and at 2 weeks of age (M value 12.8 ± 2.6 vs 16.3 ± 4.2, respectively). Insulin increased Akt phosphorylation, but these responses were significantly lower in preterm baboons during the first week of life (3.2-fold vs 9.8-fold). Preterm baboons had lower glucose transporter-1 protein content throughout the first 2 weeks of life (8%-12% of term). In preterm baboons, serum free fatty acids (FFAs) did not decrease in response to insulin, whereas FFAs decreased by greater than 80% in term baboons; the impaired suppression of FFAs in the preterm animals was paired with a decreased glucose transporter-4 protein content in adipose tissue. In conclusion, peripheral insulin resistance and impaired non-insulin-dependent glucose uptake play an important role in hyperglycemia of prematurity. Impaired insulin signaling (reduced Akt) contributes to the defect in insulin-stimulated glucose disposal. Counterregulatory hormones are not major contributors.
Collapse
Affiliation(s)
- Cynthia L Blanco
- Department of Pediatrics (C.L.B., L.L.M.-V., S.R.S., D.C.M., M.M.L., D.G.A., M.C.J.), Division of Neonatology, University of Texas Health Science Center at San Antonio, Department of Medicine (A.G., H.L., R.A.D., N.M.), Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229; Texas Diabetes Institute (H.L., R.A.D., N.M.), San Antonio, Texas 78207; San Antonio Geriatric, Research and Education Center and Barshop Institute for Longevity and Aging Studies (N.M.), San Antonio, Texas 78245, and Institute of Clinical Physiology, CNR, Pisa, Italy (A.G.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Maslak E, Zabielski P, Kochan K, Kus K, Jasztal A, Sitek B, Proniewski B, Wojcik T, Gula K, Kij A, Walczak M, Baranska M, Chabowski A, Holland RJ, Saavedra JE, Keefer LK, Chlopicki S. The liver-selective NO donor, V-PYRRO/NO, protects against liver steatosis and improves postprandial glucose tolerance in mice fed high fat diet. Biochem Pharmacol 2015; 93:389-400. [PMID: 25534988 DOI: 10.1016/j.bcp.2014.12.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/30/2022]
Affiliation(s)
- Edyta Maslak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Piotr Zabielski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland.
| | - Kamila Kochan
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Krakow, Poland.
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Barbara Sitek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Tomasz Wojcik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Katarzyna Gula
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland.
| | - Maria Walczak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| | - Małgorzata Baranska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; Faculty of Chemistry, Jagiellonian University, Ingardena 3, 30-060 Krakow, Poland.
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland.
| | - Ryan J Holland
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States.
| | - Joseph E Saavedra
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland 21702, United States.
| | - Larry K Keefer
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States.
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; Department of Experimental Pharmacology (Chair of Pharmacology), Jagiellonian University Medical College, Grzegorzecka 16, 31-531 Krakow, Poland.
| |
Collapse
|
24
|
Fiorentino TV, Owston M, Abrahamian G, La Rosa S, Marando A, Perego C, Di Cairano ES, Finzi G, Capella C, Sessa F, Casiraghi F, Paez A, Adivi A, Davalli A, Fiorina P, Guardado Mendoza R, Comuzzie AG, Sharp M, DeFronzo RA, Halff G, Dick EJ, Folli F. Chronic continuous exenatide infusion does not cause pancreatic inflammation and ductal hyperplasia in non-human primates. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:139-50. [PMID: 25447052 PMCID: PMC4278248 DOI: 10.1016/j.ajpath.2014.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 08/26/2014] [Accepted: 09/09/2014] [Indexed: 12/16/2022]
Abstract
In this study, we aimed to evaluate the effects of exenatide (EXE) treatment on exocrine pancreas of nonhuman primates. To this end, 52 baboons (Papio hamadryas) underwent partial pancreatectomy, followed by continuous infusion of EXE or saline (SAL) for 14 weeks. Histological analysis, immunohistochemistry, Computer Assisted Stereology Toolbox morphometry, and immunofluorescence staining were performed at baseline and after treatment. The EXE treatment did not induce pancreatitis, parenchymal or periductal inflammatory cell accumulation, ductal hyperplasia, or dysplastic lesions/pancreatic intraepithelial neoplasia. At study end, Ki-67-positive (proliferating) acinar cell number did not change, compared with baseline, in either group. Ki-67-positive ductal cells increased after EXE treatment (P = 0.04). However, the change in Ki-67-positive ductal cell number did not differ significantly between the EXE and SAL groups (P = 0.13). M-30-positive (apoptotic) acinar and ductal cell number did not change after SAL or EXE treatment. No changes in ductal density and volume were observed after EXE or SAL. Interestingly, by triple-immunofluorescence staining, we detected c-kit (a marker of cell transdifferentiation) positive ductal cells co-expressing insulin in ducts only in the EXE group at study end, suggesting that EXE may promote the differentiation of ductal cells toward a β-cell phenotype. In conclusion, 14 weeks of EXE treatment did not exert any negative effect on exocrine pancreas, by inducing either pancreatic inflammation or hyperplasia/dysplasia in nonhuman primates.
Collapse
Affiliation(s)
- Teresa Vanessa Fiorentino
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Michael Owston
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Gregory Abrahamian
- Department of Surgery, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Stefano La Rosa
- Department of Pathology, Ospedale di Circolo and Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Alessandro Marando
- Department of Pathology, Ospedale di Circolo and Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Carla Perego
- Department of Pharmacology and Biomolecular Science, Universitádegli Studi di Milano, Milan, Italy
| | - Eliana S Di Cairano
- Department of Pharmacology and Biomolecular Science, Universitádegli Studi di Milano, Milan, Italy
| | - Giovanna Finzi
- Department of Pathology, Ospedale di Circolo and Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Carlo Capella
- Department of Pathology, Ospedale di Circolo and Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Fausto Sessa
- Department of Pathology, Ospedale di Circolo and Department of Surgical and Morphological Sciences, University of Insubria, Varese, Italy
| | - Francesca Casiraghi
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Biomedical Sciences for Health, Universitádegli Studi di Milano, Milan, Italy
| | - Ana Paez
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Ashwin Adivi
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Alberto Davalli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Internal and Specialized Medicine, Ospedale San Raffaele, Milan, Italy
| | - Paolo Fiorina
- Department of Pediatrics, Children's Hospital Harvard Medical School, Boston, Massachusetts
| | - Rodolfo Guardado Mendoza
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; Division of Health Sciences, Department of Medicine and Nutrition, University of Guanajuato, Campus León, México, and the Research Department, Hospital Regional de Alta Especialidad del Bajío, León, Mexico
| | - Anthony G Comuzzie
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas
| | - Mark Sharp
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Ralph A DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Glenn Halff
- Department of Surgery, Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Edward J Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas; Faculdade de Ciencias Medicas (FCM), Departamento de Clinica Medica, Obesity and Comorbidities Research Center (O.C.R.C.), Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
25
|
McGill-Vargas LL, Johnson-Pais T, Johnson MC, Blanco CL. Developmental regulation of key gluconeogenic molecules in nonhuman primates. Physiol Rep 2014; 2:2/12/e12243. [PMID: 25524279 PMCID: PMC4332221 DOI: 10.14814/phy2.12243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aberrant glucose regulation is common in preterm and full‐term neonates leading to short and long‐term morbidity/mortality; however, glucose metabolism in this population is understudied. The aim of this study was to investigate developmental differences in hepatic gluconeogenic pathways in fetal/newborn baboons. Fifteen fetal baboons were delivered at 125 day (d) gestational age (GA), 140d GA, and 175d GA (term = 185d GA) via cesarean section and sacrificed at birth. Term and healthy adult baboons were used as controls. Protein content and gene expression of key hepatic gluconeogenic molecules were measured: cytosolic and mitochondrial phosphoenolpyruvate carboxykinase (PEPCK‐C and PEPCK‐M), glucose‐6‐phosphatase‐alpha (G6Pase‐α), G6Pase‐β, fructose‐1,6‐bisphosphatase (FBPase), and forkhead box‐O1 (FOXO1). Protein content of PEPCK‐M increased with advancing gestation in fetal baboons (9.6 fold increase from 125d GA to 175d GA, P < 0.001). PEPCK‐C gene expression was consistent with these developmental differences. Phosphorylation of FOXO1 was significantly lower in preterm fetal baboons compared to adults, and gene expression of FOXO1 was lower in all neonates when compared to adults (10% and 62% of adults respectively, P < 0.05). The FOXO1 target gene G6Pase expression was higher in preterm animals compared to term animals. No significant differences were found in G6Pase‐α, G6Pase‐β, FOXO1, and FBPase during fetal development. In conclusion, significant developmental differences are found in hepatic gluconeogenic molecules in fetal and neonatal baboons, which may impact the responses to insulin during the neonatal period. Further studies under insulin‐stimulated conditions are required to understand the physiologic impact of these maturational differences. Significant developmental differences were found in several hepatic gluconeogenic molecules. In particular, phosphorylated FOXO1 was significantly reduced in the liver of premature fetal baboons compared to adults and may contribute the increased incidence of hyperglycemia seen in prematurity. In addition, PEPCK increased with advancing gestational age and may play a key role in glucose regulation during the newborn period.
Collapse
Affiliation(s)
- Lisa L McGill-Vargas
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Teresa Johnson-Pais
- Department of Pediatrics, Division of Child Neurology, Developmental Pediatrics & Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Marney C Johnson
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Cynthia L Blanco
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
26
|
Moreira A, Koele-Schmidt L, Leland M, Seidner S, Blanco C. Neonatal intubation with direct laryngoscopy vs videolaryngoscopy: an extremely premature baboon model. Paediatr Anaesth 2014; 24:840-4. [PMID: 24916063 DOI: 10.1111/pan.12435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To compare the ability to successfully intubate extremely preterm baboons using conventional direct laryngoscopy (DL) vs videolaryngoscopy. METHODS A prospective randomized crossover study using experienced and inexperienced neonatal intubators. All participants were shown an educational video on intubation with each device, followed by attempt of the procedure. The time for successful intubation was the primary outcome. RESULTS Seven subjects comprised the experienced group, while 10 individuals were in the inexperienced group. The overall intubation success rate was comparable between both devices (53% vs 26%, P = 0.09); however, mean time to intubate with the conventional laryngoscope was faster (25.5 vs 39.4 s, P = 0.02). Although both groups intubated faster with DL, it only reached statistical significance in the inexperienced group (27.0 vs 48.7 s, P < 0.05). CONCLUSION Conventional DL and videolaryngoscopy are suitable modes for intubating extremely preterm baboons. Although experienced intubators prefer DL, intubation success rate and time to intubate with both devices were comparable. In inexperienced intubators, participants preferred and intubated faster with DL.
Collapse
Affiliation(s)
- Alvaro Moreira
- Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center, San Antonio, TX, USA
| | | | | | | | | |
Collapse
|
27
|
Haeno S, Maeda N, Yagi T, Tahata S, Sato M, Sakaguchi K, Miyasho T, Ueda H, Yokota H. Diethylstilbestrol decreased adrenal cholesterol and corticosterone in rats. J Endocrinol 2014; 221:261-72. [PMID: 24578295 DOI: 10.1530/joe-13-0460] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The synthetic oestrogen diethylstilbestrol (DES), which is known to bind oestrogen receptors (ERs), has been reported to have adverse effects on endocrine homeostasis; however, the molecular mechanisms underlying these effects are poorly understood. In this study, we treated rats with DES and found high levels of this compound in the liver, adrenal glands and pituitary gland, as compared with other tissues. We have also detected early adverse effects of DES in the adrenal glands. The adrenal glands of rats treated with DES (340 μg/kg body weight every 2 days) for 2 weeks showed increased weight and size and a decreased fat droplet size. Following 1 week of treatment with DES, the blood and adrenal corticosterone levels were substantially decreased without any histological alterations. The levels of the precursors for corticosteroid biosynthesis in the adrenal glands were also decreased, as determined using mass spectroscopy. Cholesterol, the principal material of corticosteroid biosynthesis, decreased substantially in the adrenal glands after only 1 week of treatment with DES. In conclusion, cholesterol insufficiency results in a reduction in adrenal corticosterone biosynthesis, which may lead to endocrine dysfunction, such as reproductive toxicity.
Collapse
Affiliation(s)
- Satoko Haeno
- Laboratory of Veterinary Biochemistry Laboratory of Veterinary Pathology Laboratory of Veterinary Anatomy, School of Veterinary Medicine Laboratory Animal Nutrition, School of Veterinary Science, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan Japan Meat Science and Technology Institute, Ebisu, Shibuya-ku, Tokyo 150-0013, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
BLANCO CL, MOREIRA AG, McGILL LL, ANZUETO DG, NATHANIELSZ P, MUSI N. Antenatal corticosteroids alter insulin signaling pathways in fetal baboon skeletal muscle. J Endocrinol 2014; 221:253-60. [PMID: 24756099 PMCID: PMC4347920 DOI: 10.1530/joe-13-0504] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We hypothesize that prenatal exposure to glucocorticoids (GCs) negatively alters the insulin signal transduction pathway and has differing effects on the fetus according to gestational age (GA) at exposure. Twenty-three fetal baboons were delivered from 23 healthy, nondiabetic mothers. Twelve preterm (0.67 GA) and 11 near-term (0.95 GA) baboons were killed immediately after delivery. Half of the pregnant baboons at each gestation received two doses of i.m. betamethasone 24 h apart (170 μg/kg) before delivery, while the other half received no intervention. Vastus lateralis muscle was obtained from postnatal animals to measure the protein content and gene expression of insulin receptor β (IRβ; INSR), IRβ Tyr 1361 phosphorylation (pIRβ), IR substrate 1 (IRS1), IRS1 tyrosine phosphorylation (pIRS1), p85 subunit of PI3-kinase, AKT (protein kinase B), phospho-AKT Ser473 (pAKT), AKT1, AKT2, and glucose transporters (GLUT1 and GLUT4). Skeletal muscle from preterm baboons exposed to GCs had markedly reduced protein content of AKT and AKT1 (respectively, 73 and 72% from 0.67 GA control, P<0.001); IRβ and pIRβ were also decreased (respectively, 94 and 85%, P<0.01) in the muscle of premature GC-exposed fetuses but not in term fetuses. GLUT1 and GLUT4 tended to increase with GC exposure in preterm animals (P=0.09), while GLUT4 increased sixfold in term animals after exposure to GC (P<0.05). In conclusion, exposure to a single course of antenatal GCs during fetal life alters the insulin signaling pathway in fetal muscle in a manner dependent on the stage of gestation.
Collapse
Affiliation(s)
- Cynthia L. BLANCO
- University of Texas Health Science Center, Department of Pediatrics, Neonatology Division, San Antonio, TX, USA 78229
- Corresponding Author: Cynthia L. Blanco MD, MSCI, UTHSCSA, 7703 Floyd Curl Drive, MC-7812, San Antonio, TX, 78229, , phone: 210-567-5247, fax: 210-567-5169
| | - Alvaro G. MOREIRA
- University of Texas Health Science Center, Department of Pediatrics, Neonatology Division, San Antonio, TX, USA 78229
| | - Lisa L. McGILL
- University of Texas Health Science Center, Department of Pediatrics, Neonatology Division, San Antonio, TX, USA 78229
| | - Diana G. ANZUETO
- University of Texas Health Science Center, Department of Pediatrics, Neonatology Division, San Antonio, TX, USA 78229
| | - Peter NATHANIELSZ
- University of Texas Health Science Center, Department of Obstetrics, Center for Pregnancy and Newborn Research, San Antonio, TX, USA 78229
| | - Nicolas MUSI
- University of Texas Health Science Center, Department of Medicine, Diabetes Division, San Antonio, TX, USA 78229
- Texas Diabetes Institute, 701 S. Zarzamora, San Antonio, TX, USA 78207
- Barshop Institute for Longevity and Aging Studies, 15355 Lambda Drive, San Antonio, TX, USA 78245
| |
Collapse
|
29
|
Chen S, Bastarrachea RA, Roberts BJ, Voruganti VS, Frost PA, Nava-Gonzalez EJ, Arriaga-Cazares HE, Chen J, Huang P, DeFronzo RA, Comuzzie AG, Grayburn PA. Successful β cells islet regeneration in streptozotocin-induced diabetic baboons using ultrasound-targeted microbubble gene therapy with cyclinD2/CDK4/GLP1. Cell Cycle 2014; 13:1145-51. [PMID: 24553120 DOI: 10.4161/cc.27997] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Both major forms of diabetes mellitus (DM) involve β-cell destruction and dysfunction. New treatment strategies have focused on replenishing the deficiency of β-cell mass common to both major forms of diabetes by islet transplantation or β-cell regeneration. The pancreas, not the liver, is the ideal organ for islet regeneration, because it is the natural milieu for islets. Since islet mass is known to increase during obesity and pregnancy, the concept of stimulating pancreatic islet regeneration in vivo is both rational and physiologic. This paper proposes a novel approach in which non-viral gene therapy is targeted to pancreatic islets using ultrasound targeted microbubble destruction (UTMD) in a non-human primate model (NHP), the baboon. Treated baboons received a gene cocktail comprised of cyclinD2, CDK, and GLP1, which in rats results in robust and durable islet regeneration with normalization of blood glucose, insulin, and C-peptide levels. We were able to generate important preliminary data indicating that gene therapy by UTMD can achieve in vivo normalization of the intravenous (IV) glucose tolerance test (IVGTT) curves in STZ hyperglycemic-induced conscious tethered baboons. Immunohistochemistry clearly demonstrated evidence of islet regeneration and restoration of β-cell mass.
Collapse
Affiliation(s)
| | - Raul A Bastarrachea
- Texas Biomedical Research Institute; San Antonio, TX USA; Southwest National Primate Research Center; San Antonio, TX USA
| | - Brad J Roberts
- Baylor Research Institute; Dallas, TX USA; Department of Internal Medicine; Division of Cardiology; Baylor Heart and Vascular Institute; Baylor University Medical Center; Dallas, TX USA
| | | | - Patrice A Frost
- Texas Biomedical Research Institute; San Antonio, TX USA; Southwest National Primate Research Center; San Antonio, TX USA
| | - Edna J Nava-Gonzalez
- Texas Biomedical Research Institute; San Antonio, TX USA; University of Nuevo Leon School of Nutrition and Public Health; Monterrey, Mexico
| | - Hector E Arriaga-Cazares
- Texas Biomedical Research Institute; San Antonio, TX USA; Hospital Infantil de Tamaulipas; Ciudad Victoria, Mexico
| | - Jiaxi Chen
- Baylor Research Institute; Dallas, TX USA
| | - Pintong Huang
- Department of Ultrasonography; The 2nd Affiliated Hospital of Zhejiang University College of Medicine; Hangzhou, Zhejiang Province, PR China
| | - Ralph A DeFronzo
- Diabetes Division; Department of Medicine; The University of Texas Health Science Center at San Antonio; San Antonio, TX USA
| | - Anthony G Comuzzie
- Texas Biomedical Research Institute; San Antonio, TX USA; Southwest National Primate Research Center; San Antonio, TX USA
| | - Paul A Grayburn
- Department of Internal Medicine; Division of Cardiology; Baylor Heart and Vascular Institute; Baylor University Medical Center; Dallas, TX USA
| |
Collapse
|
30
|
Lee JI, Shin JS, Jung WY, Lee G, Kim MS, Kim YS, Choi JW, Park CG, Kim SJ. Porcine islet adaptation to metabolic need of monkeys in pig-to-monkey intraportal islet xenotransplantation. Transplant Proc 2014; 45:1866-8. [PMID: 23769059 DOI: 10.1016/j.transproceed.2013.01.090] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/24/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Physiologic regulation of glucose metabolism is different between donor and recipient of xenogeneic pancreatic islet transplantation. We sought to assess whether the capacity of donor islets to adapt to recipient metabolic requirements should be considered in determining the success of pancreatic islet xenotransplantation. METHODS Rhesus macaque hosts rendered diabetic by streptozotocin were transplanted with porcine islets into the liver. Porcine c-peptide and insulin levels as well as intravenous glucose tolerance test (IVGTT) were measured at intervals. RESULTS At 2 months after islet transplantation, glucose responses on IVGTT showed a normoglycemic pattern. There was a 2.48 fold increase in C-peptide level during the initial 15 minutes of IVGTT in normal monkeys: from 3.122 ng/mL at baseline to 7.728 ng/mL at 15 minutes. Monkeys transplanted with porcine islets showed 2.38- and 2.45-folds the initial increases in C-peptide on IVGTT at 2 and 4 months after transplantation, respectively. Histopathologic evaluation identified the host endothelial cells having well lined the vessels of the porcine islets in the monkey liver. CONCLUSIONS The glucose response on IVGTT of porcine islets engrafted in the monkey liver resembled the normal monkey pattern rather than that of pigs. The presence of monkey endothelial cells suggested that porcine islets were well adapted to the local environment of the recipient.
Collapse
Affiliation(s)
- J-I Lee
- Xenotransplantation Research Center, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Higgins PB, Rodriguez PJ, Voruganti VS, Mattern V, Bastarrachea RA, Rice K, Raabe T, Comuzzie AG. Body composition and cardiometabolic disease risk factors in captive baboons (Papio hamadryas sp.): sexual dimorphism. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2013; 153:9-14. [PMID: 24318937 DOI: 10.1002/ajpa.22357] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 11/06/2022]
Abstract
Baboons (Papio hamadryas sp.) exhibit significant sexual dimorphism in body size. Sexual dimorphism is also exhibited in a number of circulating factors associated with risk of cardiometabolic disease. We investigated whether sexual dimorphism in body size and composition underlie these differences. We examined data from 28 male and 24 female outdoor group-housed young adult baboons enrolled in a longitudinal observational study of cardiometabolic disease risk factors. Animals were sedated with ketamine HCl (10 mg/kg) before undergoing venous blood draws, basic body measurements, and dual-energy X-ray absorptiometry body composition scans. Percentage glycated hemoglobin A1c (%HbA1c ) was measured in whole blood. Serum samples were analyzed for glucose, insulin, C-peptide, high-density lipoprotein, and triglyceride concentrations. Males were heavier and had greater body length and lean tissue mass than females. Females had a greater body fat percentage relative to males (10.8 ± 6.4 vs. 6.9 ± 4.0, P = 0.01). Although C-peptide, fasting glucose, and %HbA1c did not differ between the sexes, females had greater fasting insulin and triglyceride compared to their male counterparts. Insulin and percentage body fat were significantly correlated in males (r = 0.61, P = 0.001) and to a lesser extent in females (r = 0.43, P = 0.04). Overall, relations between adiposity and fasting insulin and fasting triglyceride were stronger in males. After accounting for differences in percentage body fat, fasting insulin and triglyceride were no longer statistically different between males and females. Despite stronger correlations between relative adiposity and insulin and triglyceride in males, the higher fasting insulin and triglyceride of female baboons may be underlain by their greater relative body fat masses.
Collapse
Affiliation(s)
- Paul B Higgins
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Fiori JL, Shin YK, Kim W, Krzysik-Walker SM, González-Mariscal I, Carlson OD, Sanghvi M, Moaddel R, Farhang K, Gadkaree SK, Doyle ME, Pearson KJ, Mattison JA, de Cabo R, Egan JM. Resveratrol prevents β-cell dedifferentiation in nonhuman primates given a high-fat/high-sugar diet. Diabetes 2013; 62:3500-13. [PMID: 23884882 PMCID: PMC3781448 DOI: 10.2337/db13-0266] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Eating a "Westernized" diet high in fat and sugar leads to weight gain and numerous health problems, including the development of type 2 diabetes mellitus (T2DM). Rodent studies have shown that resveratrol supplementation reduces blood glucose levels, preserves β-cells in islets of Langerhans, and improves insulin action. Although rodent models are helpful for understanding β-cell biology and certain aspects of T2DM pathology, they fail to reproduce the complexity of the human disease as well as that of nonhuman primates. Rhesus monkeys were fed a standard diet (SD), or a high-fat/high-sugar diet in combination with either placebo (HFS) or resveratrol (HFS+Resv) for 24 months, and pancreata were examined before overt dysglycemia occurred. Increased glucose-stimulated insulin secretion and insulin resistance occurred in both HFS and HFS+Resv diets compared with SD. Although islet size was unaffected, there was a significant decrease in β-cells and an increase in α-cells containing glucagon and glucagon-like peptide 1 with HFS diets. Islets from HFS+Resv monkeys were morphologically similar to SD. HFS diets also resulted in decreased expression of essential β-cell transcription factors forkhead box O1 (FOXO1), NKX6-1, NKX2-2, and PDX1, which did not occur with resveratrol supplementation. Similar changes were observed in human islets where the effects of resveratrol were mediated through Sirtuin 1. These findings have implications for the management of humans with insulin resistance, prediabetes, and diabetes.
Collapse
Affiliation(s)
- Jennifer L. Fiori
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Yu-Kyong Shin
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
- Biochemistry Department, Boston University School of Medicine, Boston, Massachusetts
| | - Wook Kim
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
- Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Susan M. Krzysik-Walker
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Isabel González-Mariscal
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Olga D. Carlson
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Mitesh Sanghvi
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Kathleen Farhang
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Shekhar K. Gadkaree
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Maire E. Doyle
- Division of Endocrinology, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Kevin J. Pearson
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
- Graduate Center for Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Julie A. Mattison
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Josephine M. Egan
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
- Corresponding author: Josephine M. Egan,
| |
Collapse
|
33
|
Chu MP, Klopfenstein BJ, Krisky CM, Urbanski HF, Rooney WD, Kohama SG, Purnell JQ. Intrahepatic lipid, not visceral or muscle fat, is correlated with insulin resistance in older, female rhesus macaques. Obesity (Silver Spring) 2013; 21:2021-8. [PMID: 23408675 PMCID: PMC3661746 DOI: 10.1002/oby.20339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 12/11/2012] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Little is known of the effect of body composition on glucose metabolism in the aging female non-human primate. These variables in older female Rhesus macaques were studied. DESIGN AND METHODS Female Rhesus macaques (Macaca mulatta, n = 19, age range 23-30 years) underwent magnetic resonance imaging and (1) H spectroscopy to quantify total abdominal fat, visceral fat (VF), subcutaneous fat (SF) area, extramyocellular lipid (EMCL), intramyocellular lipid (IMCL) and intrahepatic lipid (IHL) content, and DEXA scan for whole body composition. A subgroup (n = 12) underwent a fasting blood draw and intravenous glucose tolerance test. RESULTS SF correlated with homeostatic model assessment of insulin resistance (HOMAIR ) and quantitative insulin sensitivity check index (QUICKI), but not after adjustment for fat mass. IHL demonstrated the strongest correlation with HOMAIR , QUICKI and calculated insulin sensitivity index (CSI ), and remained significant after adjustment for fat mass. VF, IMCL, and EMCL did not correlate with any of our measures of insulin sensitivity. CONCLUSIONS Despite a greater amount of VF compared to SF, VF was not associated with markers of insulin resistance (IR) in the older female monkey. Instead, IHL is a marker for IR in the fasting and post-prandial state in these animals.
Collapse
Affiliation(s)
- Michael P Chu
- Department of Medicine, Division of Endocrinology, Diabetes, and Clinical Nutrition, Oregon Health and Science University, Portland, OR, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Casiraghi F, Lertwattanarak R, Luzi L, Chavez AO, Davalli AM, Naegelin T, Comuzzie AG, Frost P, Musi N, Folli F. Energy expenditure evaluation in humans and non-human primates by SenseWear Armband. Validation of energy expenditure evaluation by SenseWear Armband by direct comparison with indirect calorimetry. PLoS One 2013; 8:e73651. [PMID: 24069218 PMCID: PMC3777938 DOI: 10.1371/journal.pone.0073651] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/22/2013] [Indexed: 11/25/2022] Open
Abstract
Introduction The purpose of this study was to compare and validate the use of SenseWear Armband (SWA) placed on the arm (SWA ARM) and on the back (SWA BACK) in healthy humans during resting and a cycle-ergometer exercise and to evaluate the SWA to estimate Resting Energy Expenditure (REE) and Total Energy Expenditure (TEE) in healthy baboons. Methods We studied 26 (15F/11M) human subjects wearing SWA in two different anatomical sites (arm and back) during resting and a cycle-ergometer test and directly compared these results with indirect calorimetry evaluation (IC), performed at the same time. We then inserted the SWA in a metabolic jacket for baboons and evaluated the TEE and REE in free living condition for 6 days in 21 (8F/13M) non-human primates. Results In humans we found a good correlation between SWA place on the ARM and on the BACK with IC during the resting experiment (1.1±0.3 SWAs, 1±0.2 IC kcal/min) and a slight underestimation in the SWAs data compared with IC during the cycle-ergometer exercise (5±1.9 SWA ARM, 4.5±1.5 SWA BACK and 5.4±2.1 IC kcal/min). In the non-human primate (baboons) experiment SWA estimated a TEE of 0.54±0.009 kcal/min during free living and a REE of 0.82±0.06 kcal/min. Conclusion SWA, an extremely simple and inexpensive apparatus, provides quite accurate measurements of energy expenditure in humans and in baboons. Energy expenditure data obtained with SWA are highly correlated with the data obtained with “gold standard”, IC, in humans.
Collapse
Affiliation(s)
- Francesca Casiraghi
- Department of Medicine/Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
- Metabolism Research Center, I.R.C.C.S. Policlinico San Donato Hospital, Milano, Italy
| | - Raweewan Lertwattanarak
- Department of Medicine/Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Texas Diabetes Institute, San Antonio, Texas, United States of America
| | - Livio Luzi
- Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
- Metabolism Research Center, I.R.C.C.S. Policlinico San Donato Hospital, Milano, Italy
| | - Alberto O. Chavez
- Department of Medicine/Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | | | - Terry Naegelin
- Southwest National Primate Research Center, San Antonio, Texas, United States of America
| | - Anthony G. Comuzzie
- Southwest National Primate Research Center, San Antonio, Texas, United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Patricia Frost
- Southwest National Primate Research Center, San Antonio, Texas, United States of America
| | - Nicolas Musi
- Department of Medicine/Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Texas Diabetes Institute, San Antonio, Texas, United States of America
| | - Franco Folli
- Department of Medicine/Division of Diabetes, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
35
|
Guardado-Mendoza R, Jimenez-Ceja L, Majluf-Cruz A, Kamath S, Fiorentino TV, Casiraghi F, Velazquez AOC, DeFronzo RA, Dick E, Davalli A, Folli F. Impact of obesity severity and duration on pancreatic β- and α-cell dynamics in normoglycemic non-human primates. Int J Obes (Lond) 2013; 37:1071-8. [PMID: 23229736 PMCID: PMC3906680 DOI: 10.1038/ijo.2012.205] [Citation(s) in RCA: 22] [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: 08/16/2012] [Revised: 11/05/2012] [Accepted: 11/08/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Obesity is associated with high insulin and glucagon plasma levels. Enhanced β-cell function and β-cell expansion are responsible for insulin hypersecretion. It is unknown whether hyperglucagonemia is due to α-cell hypersecretion or to an increase in α-cell mass. In this study, we investigated the dynamics of the β-cell and α-cell function and mass in pancreas of obese normoglycemic baboons. METHODS Pancreatic β- and α-cell volumes were measured in 51 normoglycemic baboons divided into six groups according to overweight severity or duration. Islets morphometric parameters were correlated to overweight and to diverse metabolic and laboratory parameters. RESULTS Relative α-cell volume (RαV) and relative islet α-cell volume (RIαV) increased significantly with both overweight duration and severity. Conversely, in spite of the induction of insulin resistance, overweight produced only modest effects on relative β-cell volume (RβV) and relative islet β-cell volume (RIβV). Of note, RIβV did not increase neither with overweight duration nor with overweight severity, supposedly because of the concomitant, greater increase in RIαV. Baboons' body weights correlated with serum levels of interleukin-6 and tumor necrosis factor-α soluble receptors, demonstrating that overweight induces abnormal activation of the signaling of two cytokines known to impact differently β- and α-cell viability and replication. CONCLUSION In conclusion, overweight and insulin resistance induce in baboons a significant increase in α-cell volumes (RαV, RIαV), whereas have minimal effects on the β cells. This study suggests that an increase in the α-cell mass may precede the loss of β cells and the transition to overt hyperglycemia and diabetes.
Collapse
Affiliation(s)
- R Guardado-Mendoza
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liang H, Tantiwong P, Sriwijitkamol A, Shanmugasundaram K, Mohan S, Espinoza S, Defronzo RA, Dubé JJ, Musi N. Effect of a sustained reduction in plasma free fatty acid concentration on insulin signalling and inflammation in skeletal muscle from human subjects. J Physiol 2013; 591:2897-909. [PMID: 23529132 DOI: 10.1113/jphysiol.2012.247510] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Free fatty acids (FFAs) have been implicated in the pathogenesis of insulin resistance. Reducing plasma FFA concentration in obese and type 2 diabetic (T2DM) subjects improves insulin sensitivity. However, the molecular mechanism by which FFA reduction improves insulin sensitivity in human subjects is not fully understood. In the present study, we tested the hypothesis that pharmacological FFA reduction enhances insulin action by reducing local (muscle) inflammation, leading to improved insulin signalling. Insulin-stimulated total glucose disposal (TGD), plasma FFA species, muscle insulin signalling, IBα protein, c-Jun phosphorylation, inflammatory gene (toll-like receptor 4 and monocyte chemotactic protein 1) expression, and ceramide and diacylglycerol (DAG) content were measured in muscle from a group of obese and T2DM subjects before and after administration of the antilipolytic drug acipimox for 7 days, and the results were compared to lean individuals. We found that obese and T2DM subjects had elevated saturated and unsaturated FFAs in plasma, and acipimox reduced all FFA species. Acipimox-induced reductions in plasma FFAs improved TGD and insulin signalling in obese and T2DM subjects. Acipimox increased IBα protein (an indication of decreased IB kinase-nuclear factor B signalling) in both obese and T2DM subjects, but did not affect c-Jun phosphorylation in any group. Acipimox also decreased inflammatory gene expression, although this reduction only occurred in T2DM subjects. Ceramide and DAG content did not change. To summarize, pharmacological FFA reduction improves insulin signalling in muscle from insulin-resistant subjects. This beneficial effect on insulin action could be related to a decrease in local inflammation. Notably, the improvements in insulin action were more pronounced in T2DM, indicating that these subjects are more susceptible to the toxic effect of FFAs.
Collapse
Affiliation(s)
- Hanyu Liang
- N. Musi: Audie L. Murphy VA Medical Center, 7400 Merton Minter, San Antonio, TX 78229, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Blanco CL, McGill-Vargas LL, McCurnin D, Quinn AR. Hyperglycemia increases the risk of death in extremely preterm baboons. Pediatr Res 2013; 73:337-43. [PMID: 23364173 PMCID: PMC4112412 DOI: 10.1038/pr.2012.184] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Transient neonatal hyperglycemia (HG) has been reported in up to 80% of extremely preterm human infants. We hypothesize that severe HG is associated with increased morbidity and mortality in preterm baboons. METHODS Sixty-six baboons born at 67% of gestation were studied. HG was defined as serum glucose level ≥150 mg/dl during the first week of life. Animals were stratified into two groups: severe HG (≥8 events) and nonsevere HG (<8 events). RESULTS HG developed in 65 of the 66 (98%) baboons that were included. A total of 3,386 glucose measurements were obtained. The mean serum glucose level was 159 ± 69 mg/dl for the severe HG group and 130 ± 48 mg/dl for the nonsevere HG group during the first week of life. No differences were found in gender, birth weight, sepsis, patent ductus arteriosus, or oxygenation/ventilation indexes between groups. Severe HG was associated with early death even after controlling for sepsis, postnatal steroid exposure, and catecholamine utilization. CONCLUSION HG is common in preterm baboons and is not associated with short-term morbidity. Severe HG occurring in the first week of life is associated with early death in preterm baboons.
Collapse
Affiliation(s)
- Cynthia L. Blanco
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA,Corresponding Author: 7703 Floyd Curl Drive, MC-7812 San Antonio, TX, 78229 phone: 210-567-5225, fax: 210-567-5169
| | - Lisa L. McGill-Vargas
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Donald McCurnin
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Amy R. Quinn
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
38
|
Hansen BC. Investigation and treatment of type 2 diabetes in nonhuman primates. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2013; 933:177-85. [PMID: 22893407 DOI: 10.1007/978-1-62703-068-7_11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Nonhuman primates provide the ideal animal model for discovering and examining further the mechanisms underlying human type 2 diabetes mellitus. In all aspects studied to date the nonhuman primate has been shown to develop the same disease with the same features that develop in overweight middle-aged humans. This includes the progressive development of the known complications of diabetes, all of which are extraordinarily like those identified in humans. In addition, for the development and evaluation of new therapeutic agents, the translation of findings from nonhuman primates to application in humans has been highly predictable. Both therapeutic efficacy and identification of potential adverse responses can be effectively examined in nonhuman primates due to their great similarity to humans at the molecular, biochemical, and physiological levels. This chapter provides guidance for the development and management of a colony of monkeys with naturally occurring type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Barbara C Hansen
- Departments of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
39
|
Ma Y, Luo T, Xu W, Ye Z, Hong A. A new recombinant pituitary adenylate cyclase-activating peptide-derived peptide efficiently promotes glucose uptake and glucose-dependent insulin secretion. Acta Biochim Biophys Sin (Shanghai) 2012; 44:948-56. [PMID: 23052710 DOI: 10.1093/abbs/gms078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The recombinant peptide, DBAYL, a promising therapeutic peptide for type 2 diabetes, is a new, potent, and highly selective agonist for VPAC2 generated through site-directed mutagenesis based on sequence alignments of pituitary adenylate cyclase-activating peptide (PACAP), vasoactive intestinal peptide (VIP), and related analogs. The recombinant DBAYL was used to evaluate its effect and mechanism in blood glucose metabolism and utilization. As much as 28.9 mg recombinant DBAYL peptide with purity over 98% can be obtained from 1 l of Luria-Bertani medium culture by the method established in this study and the prepared DBAYL with four mutations (N10Q, V18L, N29Q, and M added to the N-terminal) were much more stable than BAY55-9837. The half-life of recombinant DBAYL was about 25 folds compared with that of BAY55-9837 in vitro. The bioactivity assay of DBAYL showed that it displaced [(125)I]PACAP38 and [(125)I]VIP from VPAC2 with a half-maximal inhibitory concentration of 48.4 ± 6.9 and 47.1 ± 4.9 nM, respectively, which were significantly lower than that of BAY55-9837, one established VPAC2 agonists. DBAYL enhances the cAMP accumulation in CHO cells expressing human VPAC2 with a half-maximal stimulatory concentration (EC(50)) of 0.68 nM, whereas the receptor potency of DBAYL at human VPAC1 (EC(50) of 737 nM) was only 1/1083 of that at human VPAC2, and DBAYL had no activity toward human PAC1 receptor. Western blot analysis of the key proteins of insulin receptor signaling pathway: insulin receptor substrate 1 (IRS-1) and glucose transporter 4 (GLUT4) indicated that the DBAYL could significantly induce the insulin-stimulated IRS-1 and GLUT4 expression more efficiently than BAY55-9837 and VIP in adipocytes. Compared with BAY55-9837 and PACAP38, the recombinant peptide DBAYL can more efficiently promote insulin release and decrease plasma glucose level in Institute of Cancer Research (ICR) mice. These results suggested that DBAYL could efficiently improve glucose uptake and glucose-dependent insulin secretion by VPAC2-mediated effect.
Collapse
Affiliation(s)
- Yi Ma
- Department of Cell Biology, Institute of Biological Medicine, Jinan University, Guangzhou 510632, China
| | | | | | | | | |
Collapse
|
40
|
Quinn AR, Blanco CL, Perego C, Finzi G, La Rosa S, Capella C, Guardado-Mendoza R, Casiraghi F, Gastaldelli A, Johnson M, Dick EJ, Folli F. The ontogeny of the endocrine pancreas in the fetal/newborn baboon. J Endocrinol 2012; 214:289-99. [PMID: 22723715 PMCID: PMC3686495 DOI: 10.1530/joe-12-0070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Erratic regulation of glucose metabolism including hyperglycemia is a common condition in premature infants and is associated with increased morbidity and mortality. The objective of this study was to examine histological and ultrastructural differences in the endocrine pancreas in fetal (throughout gestation) and neonatal baboons. Twelve fetal baboons were delivered at 125 days (d) gestational age (GA), 140d GA, or 175d GA. Eight animals were delivered at term (185d GA); half were fed for 5 days. Seventy-three nondiabetic adult baboons were used for comparison. Pancreatic tissue was studied using light microscopy, confocal imaging, and electron microscopy. The fetal and neonatal endocrine pancreas islet architecture became more organized as GA advanced. The percent areas of α-β-δ-cell type were similar within each fetal and newborn GA (NS) but were higher than the adults (P<0.05) regardless of GA. The ratio of β cells within the islet (whole and core) increased with gestation (P<0.01). Neonatal baboons, which survived for 5 days (feeding), had a 2.5-fold increase in pancreas weight compared with their counterparts killed at birth (P=0.01). Endocrine cells were also found in exocrine ductal and acinar cells in 125, 140 and 175d GA fetuses. Subpopulation of tissue that coexpressed trypsin and glucagon/insulin shows the presence of cells with mixed endo-exocrine lineage in fetuses. In summary, the fetal endocrine pancreas has no prevalence of a α-β-δ-cell type with larger endocrine cell percent areas than adults. Cells with mixed endocrine/exocrine phenotype occur during fetal development. Developmental differences may play a role in glucose homeostasis during the neonatal period and may have long-term implications.
Collapse
Affiliation(s)
- Amy R. Quinn
- Department of Pediatrics, Neonatology Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| | - Cynthia L. Blanco
- Department of Pediatrics, Neonatology Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| | - Carla Perego
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20134 Milan, Italy
| | - Giovanna Finzi
- Department of Pathology, Ospedale di Circolo, Department of Human Morphology, and Centro Insubre di Biotecnologie per la Salute Umana, 21100 Varese, Italy
| | - Stefano La Rosa
- Department of Pathology, Ospedale di Circolo, Department of Human Morphology, and Centro Insubre di Biotecnologie per la Salute Umana, 21100 Varese, Italy
| | - Carlo Capella
- Department of Pathology, Ospedale di Circolo, Department of Human Morphology, and Centro Insubre di Biotecnologie per la Salute Umana, 21100 Varese, Italy
| | - Rodolfo Guardado-Mendoza
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| | - Francesca Casiraghi
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| | - Amalia Gastaldelli
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
- Fondazione G. Monasterio and Institute of Clinical Physiology, National Research Council, 56126 Pisa, Italy
| | - Marney Johnson
- Department of Pediatrics, Neonatology Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| | - Edward J. Dick
- Texas Biomedical Research Institute, San Antonio, TX, 78245
| | - Franco Folli
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229
| |
Collapse
|
41
|
Vaidyanathan V, Bastarrachea RA, Higgins PB, Voruganti VS, Kamath S, DiPatrizio NV, Piomelli D, Comuzzie AG, Parks EJ. Selective cannabinoid-1 receptor blockade benefits fatty acid and triglyceride metabolism significantly in weight-stable nonhuman primates. Am J Physiol Endocrinol Metab 2012; 303:E624-34. [PMID: 22761159 PMCID: PMC3468508 DOI: 10.1152/ajpendo.00072.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The goal of this study was to determine whether administration of the CB₁ cannabinoid receptor antagonist rimonabant would alter fatty acid flux in nonhuman primates. Five adult baboons (Papio Sp) aged 12.1 ± 4.7 yr (body weight: 31.9 ± 2.1 kg) underwent repeated metabolic tests to determine fatty acid and TG flux before and after 7 wk of treatment with rimonabant (15 mg/day). Animals were fed ad libitum diets, and stable isotopes were administered via diet (d₃₁-tripalmitin) and intravenously (¹³C₄-palmitate, ¹³C₁-acetate). Plasma was collected in the fed and fasted states, and blood lipids were analyzed by GC-MS. DEXA was used to assess body composition and a hyperinsulinemic euglycemic clamp used to assess insulin-mediated glucose disposal. During the study, no changes were observed in food intake, body weight, plasma, and tissue endocannabinoid concentrations or the quantity of liver-TG fatty acids originating from de novo lipogenesis (19 ± 6 vs. 16 ± 5%, for pre- and posttreatment, respectively, P = 0.39). However, waist circumference was significantly reduced 4% in the treated animals (P < 0.04), glucose disposal increased 30% (P = 0.03), and FFA turnover increased 37% (P = 0.02). The faster FFA flux was consistent with a 43% reduction in these fatty acids used for TRL-TG synthesis (40 ± 3 vs. 23 ± 4%, P = 0.02) and a twofold increase in TRL-TG turnover (1.5 ± 0.9 vs. 3.1 ± 1.4 μmol·kg⁻¹·h⁻¹, P = 0.03). These data support the potential for a strong effect of CB₁ receptor antagonism at the level of adipose tissue, resulting in improvements in fasting turnover of fatty acids at the whole body level, central adipose storage, and significant improvements in glucose homeostasis.
Collapse
Affiliation(s)
- Vidya Vaidyanathan
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9052, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rincon-Choles H, Abboud HE, Lee S, Shade RE, Rice KS, Carey KD, Comuzzie AG, Barnes JL. Renal histopathology of a baboon model with type 2 diabetes. Toxicol Pathol 2012; 40:1020-30. [PMID: 22552392 DOI: 10.1177/0192623312444025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Naturally occurring type 2 diabetes has been found in a colony of baboons. Ongoing characterization of the baboon colony maintained at the Southwest National Primate Research Center has revealed a significant range of glucose sensitivity with some animals clearly diabetic. Seven baboons, four with diabetes and three without diabetes, underwent histopathological investigation. Three diabetic animals were diagnosed using fasting blood glucose, hemoglobin A1C, and intravenous glucose tolerance test, and a fourth one was known to have hyperglycemia. One control baboon and three baboons with diabetes had microalbuminuria. On kidney biopsy, diabetic baboons had thickening of the glomerular basement membrane and mesangial matrix expansion compared to controls. Immunohistochemistry showed the diabetic animals had increased mesangial expression of cellular fibronectin ED-A. Two diabetic animals with microalbuminuria had evidence of mesangiolysis with the formation of an early nodule. One diabetic animal had a Kimmestiel-Wilson nodule. We conclude that the baboon represents a useful primate model of diabetes and nephropathy that resembles the nephropathy associated with type 2 diabetes in humans.
Collapse
Affiliation(s)
- Hernan Rincon-Choles
- Audie L Murphy Division, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Folli F, Okada T, Perego C, Gunton J, Liew CW, Akiyama M, D'Amico A, La Rosa S, Placidi C, Lupi R, Marchetti P, Sesti G, Hellerstein M, Perego L, Kulkarni RN. Altered insulin receptor signalling and β-cell cycle dynamics in type 2 diabetes mellitus. PLoS One 2011; 6:e28050. [PMID: 22140505 PMCID: PMC3227614 DOI: 10.1371/journal.pone.0028050] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 10/31/2011] [Indexed: 12/31/2022] Open
Abstract
Insulin resistance, reduced β-cell mass, and hyperglucagonemia are consistent features in type 2 diabetes mellitus (T2DM). We used pancreas and islets from humans with T2DM to examine the regulation of insulin signaling and cell-cycle control of islet cells. We observed reduced β-cell mass and increased α-cell mass in the Type 2 diabetic pancreas. Confocal microscopy, real-time PCR and western blotting analyses revealed increased expression of PCNA and down-regulation of p27-Kip1 and altered expression of insulin receptors, insulin receptor substrate-2 and phosphorylated BAD. To investigate the mechanisms underlying these findings, we examined a mouse model of insulin resistance in β-cells – which also exhibits reduced β-cell mass, the β-cell-specific insulin receptor knockout (βIRKO). Freshly isolated islets and β-cell lines derived from βIRKO mice exhibited poor cell-cycle progression, nuclear restriction of FoxO1 and reduced expression of cell-cycle proteins favoring growth arrest. Re-expression of insulin receptors in βIRKO β-cells reversed the defects and promoted cell cycle progression and proliferation implying a role for insulin-signaling in β-cell growth. These data provide evidence that human β- and α-cells can enter the cell-cycle, but proliferation of β-cells in T2DM fails due to G1-to-S phase arrest secondary to defective insulin signaling. Activation of insulin signaling, FoxO1 and proteins in β-cell-cycle progression are attractive therapeutic targets to enhance β-cell regeneration in the treatment of T2DM.
Collapse
Affiliation(s)
- Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail: (RNK); (FF)
| | - Terumasa Okada
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Carla Perego
- Department of Molecular Science Applied to Biosystems, Università degli Studi di Milano, Milan, Italy
| | - Jenny Gunton
- Garvan Institute of Medical Research, Sydney, Australia
| | - Chong Wee Liew
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Masaru Akiyama
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anna D'Amico
- Department of Molecular Science Applied to Biosystems, Università degli Studi di Milano, Milan, Italy
| | - Stefano La Rosa
- Department of Pathology, Ospedale di Circolo and Department of Human Morphology, University of Insubria, Varese, Italy
| | - Claudia Placidi
- Department of Pathology, Ospedale di Circolo and Department of Human Morphology, University of Insubria, Varese, Italy
| | - Roberto Lupi
- Division of Endocrinology, University of Pisa, Pisa, Italy
| | | | - Giorgio Sesti
- Department of Clinical and Experimental Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Marc Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States of America
| | - Lucia Perego
- Department of Molecular Science Applied to Biosystems, Università degli Studi di Milano, Milan, Italy
| | - Rohit N. Kulkarni
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (RNK); (FF)
| |
Collapse
|
44
|
Kamath S, Chavez AO, Gastaldelli A, Casiraghi F, Halff GA, Abrahamian GA, Davalli AM, Bastarrachea RA, Comuzzie AG, Guardado-Mendoza R, Jimenez-Ceja LM, Mattern V, Paez AM, Ricotti A, Tejero ME, Higgins PB, Rodriguez-Sanchez IP, Tripathy D, DeFronzo RA, Dick EJ, Cline GW, Folli F. Coordinated defects in hepatic long chain fatty acid metabolism and triglyceride accumulation contribute to insulin resistance in non-human primates. PLoS One 2011; 6:e27617. [PMID: 22125617 PMCID: PMC3220682 DOI: 10.1371/journal.pone.0027617] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 10/20/2011] [Indexed: 01/07/2023] Open
Abstract
Non-Alcoholic fatty liver disease (NAFLD) is characterized by accumulation of triglycerides (TG) in hepatocytes, which may also trigger cirrhosis. The mechanisms of NAFLD are not fully understood, but insulin resistance has been proposed as a key determinant.
Collapse
Affiliation(s)
- Subhash Kamath
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Alberto O. Chavez
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Amalia Gastaldelli
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Francesca Casiraghi
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Glenn A. Halff
- The UT Transplant Center, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Gregory A. Abrahamian
- The UT Transplant Center, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Alberto M. Davalli
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Internal Medicine, Diabetes & Endocrinology Unit, San Raffaele Scientific Institute, Milano, Italy
| | - Raul A. Bastarrachea
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Anthony G. Comuzzie
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Rodolfo Guardado-Mendoza
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lilia M. Jimenez-Ceja
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Vicki Mattern
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Ana Maria Paez
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Andrea Ricotti
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Mary E. Tejero
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Paul B. Higgins
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Iram Pablo Rodriguez-Sanchez
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Devjit Tripathy
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ralph A. DeFronzo
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Edward J. Dick
- Southwest National Primate Research Center/Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Gary W. Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Franco Folli
- Department of Medicine/Division of Diabetes. The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Internal Medicine, Diabetes & Endocrinology Unit, San Raffaele Scientific Institute, Milano, Italy
- * E-mail:
| |
Collapse
|
45
|
Andreozzi F, Procopio C, Greco A, Mannino GC, Miele C, Raciti GA, Iadicicco C, Beguinot F, Pontiroli AE, Hribal ML, Folli F, Sesti G. Increased levels of the Akt-specific phosphatase PH domain leucine-rich repeat protein phosphatase (PHLPP)-1 in obese participants are associated with insulin resistance. Diabetologia 2011; 54:1879-87. [PMID: 21461637 DOI: 10.1007/s00125-011-2116-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 02/17/2011] [Indexed: 10/18/2022]
Abstract
AIMS/HYPOTHESIS We determined the contribution to insulin resistance of the PH domain leucine-rich repeat protein phosphatase (PHLPP), which dephosphorylates Akt at Ser473, inhibiting its activity. We measured the abundance of PHLPP in fat and skeletal muscle from obese participants. To study the effect of PHLPP on insulin signalling, PHLPP (also known as PHLPP1) was overexpressed in HepG2 and L6 cells. METHODS Subcutaneous fat samples were obtained from 82 morbidly obese and ten non-obese participants. Skeletal muscle samples were obtained from 12 obese and eight non-obese participants. Quantification of PHLPP-1 in human tissues was performed by immunoblotting. The functional consequences of recombinant PHLPP1 overexpression in hepatoma HepG2 cells and L6 myoblasts were investigated. RESULTS Of the 82 obese participants, 31 had normal fasting glucose, 33 impaired fasting glucose and 18 type 2 diabetes. PHLPP-1 abundance was twofold higher in the three obese groups than in non-obese participants (p = 0.004). No differences were observed between obese participants with normal fasting glucose, impaired fasting glucose or type 2 diabetes. PHLPP-1 abundance was correlated with basal Akt Ser473 phosphorylation (r = -0.48; p = 0.001), BMI (r = 0.44; p < 0.0001), insulin (r = 0.35; p < 0.0001) and HOMA (r = 0.38; p < 0.0001). PHLPP-1 abundance was twofold higher in the skeletal muscle of 12 obese participants than in that of eight non-obese participants (p < 0.0001). Insulin treatment of HepG2 cells resulted in a dose- and time-dependent upregulation of PHLPP-1. Overexpression of PHLPP1 in HepG2 cells and L6 myoblasts resulted in impaired insulin signalling involving Akt/glycogen synthase kinase 3, glycogen synthesis and glucose transport. CONCLUSIONS/INTERPRETATION Increased abundance of PHLPP-1, production of which is regulated by insulin, may represent a new molecular defect in insulin-resistant states such as obesity.
Collapse
Affiliation(s)
- F Andreozzi
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Choi J, Li C, McDonald TJ, Comuzzie A, Mattern V, Nathanielsz PW. Emergence of insulin resistance in juvenile baboon offspring of mothers exposed to moderate maternal nutrient reduction. Am J Physiol Regul Integr Comp Physiol 2011; 301:R757-62. [PMID: 21653880 DOI: 10.1152/ajpregu.00051.2011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Developmental programming of postnatal pancreatic β-cell and peripheral insulin function by maternal nutrient reduction (MNR) has been extensively investigated in rodents and sheep, but no data exist from nonhuman primate offspring of MNR mothers. We hypothesized that moderate levels of MNR would result in developmental programming of postnatal β-cell function and peripheral insulin sensitivity that lead to emergence of a prediabetic state prior to puberty. Prepregnancy phenotype of 18 nonpregnant baboons was matched. During pregnancy and lactation 12 mothers ate chow ad libitum (controls), while six ate 70% of chow consumed by controls (weight-adjusted MNR). Weaned offspring ate normal chow. At 3.5 ± 0.18 yr (mean ± SE) in an intravenous glucose tolerance test, conscious, tethered MNR juvenile offspring (2 females and 4 males) showed increased fasting glucose (P < 0.04), fasting insulin (P < 0.04), and insulin area under the curve (AUC; P < 0.01) compared with controls (8 females and 4 males). Insulin AUC also increased following an arginine challenge (P < 0.02). Baseline homeostatic model assessment insulin β-cell sensitivity was greater in MNR offspring than controls (P < 0.03). In a hyperinsulinemic, euglycemic clamp, the glucose disposal rate decreased 26% in MNR offspring. Changes observed were not sex dependent. MNR in pregnancy and lactation programs offspring metabolic responses, increasing insulin resistance and β-cell responsiveness, resulting in emergence of an overall phenotype that would predispose to later life type-2 diabetes, especially, should other dietary challenges such as a Westernized diet be experienced.
Collapse
Affiliation(s)
- Jaehyek Choi
- Center for Pregnancy and Newborn Research, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|
47
|
Bastarrachea RA, Veron SM, Vaidyanathan V, Garcia-Forey M, Voruganti VS, Higgins PB, Parks EJ. Protocol for the measurement of fatty acid and glycerol turnover in vivo in baboons. J Lipid Res 2011; 52:1272-1280. [PMID: 21415122 PMCID: PMC3090248 DOI: 10.1194/jlr.d012385] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 03/15/2011] [Indexed: 01/22/2023] Open
Abstract
Recognition of the strength of nonhuman primate models in investigating metabolic disorders has resulted in an expanded need for in vivo research techniques. We studied adipose metabolism in 10 baboons (13.0 ± 4.2 years old, 29.5 ± 5.5 kg). Part 1 evaluated the effect of different sedatives on the rate of appearance of plasma free fatty acids (RaFFA), assessed using ¹³C₄-labeled palmitate infusion (7 µmol/kg/min). Animals, were studied with no sedation, with complete isoflurane sedation, and with minimal midazolam infusion (0.04 mg/kg/h), with the last scheme allowing for the most consistent values and animals that were visually more calm. In Part 2, RaFFA and RaGlycerol (D₅-glycerol, 5 mg/kg lean body mass/h) were measured. From midnight to 0300, flux fell and came to a steady state between 0500 and 0700 h (RaFFA, 39.4 ± 29.8 μmol/kg fat mass/min; and RaGlycerol, 26.9 ± 7.3 μmol/kg/min). The RaFFA-to-RaGlycerol ratio was 1.5 ± 0.8 (49% reesterification). The decline in turnover throughout the night reflects natural circadian processes and was mirrored by reductions in FFA and glycerol to 0.62 and ± 0.14 and 0.16 and ± 0.03 mmol/l, respectively. The concurrent changes in both FFA and glycerol kinetics indicate physiologic validity of the method. These techniques will support needed research to determine mechanisms by which treatments act upon the adipocyte in vivo.
Collapse
Affiliation(s)
- Raul A Bastarrachea
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - Sonya M Veron
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX
| | - Vidya Vaidyanathan
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX
| | - Maggie Garcia-Forey
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - V Saroja Voruganti
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - Paul B Higgins
- Department of Genetics, Texas Biomedical Research Institute, San Antonio, TX
| | - Elizabeth J Parks
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
48
|
Ortmeyer HK, Sajan MP, Miura A, Kanoh Y, Rivas J, Li Y, Standaert ML, Ryan AS, Bodkin NL, Farese RV, Hansen BC. Insulin signaling and insulin sensitizing in muscle and liver of obese monkeys: peroxisome proliferator-activated receptor gamma agonist improves defective activation of atypical protein kinase C. Antioxid Redox Signal 2011; 14:207-19. [PMID: 20518698 PMCID: PMC3014763 DOI: 10.1089/ars.2010.3234] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Obesity, the metabolic syndrome, and aging share several pathogenic features in both humans and non-human primates, including insulin resistance and inflammation. Since muscle and liver are considered key integrators of metabolism, we sought to determine in biopsies from lean and obese aging rhesus monkeys the nature of defects in insulin activation and, further, the potential for mitigation of such defects by an in vivo insulin sensitizer, rosiglitazone, and a thiazolidinedione activator of the peroxisome proliferator-activated receptor gamma. The peroxisome proliferator-activated receptor gamma agonist reduced hyperinsulinemia, improved insulin sensitivity, lowered plasma triglycerides and free fatty acids, and increased plasma adiponectin. In muscle of obese monkeys, previously shown to exhibit defective insulin signaling, the insulin sensitizer improved insulin activation of atypical protein kinase C (aPKC), the defective direct activation of aPKC by phosphatidylinositol (PI)-3,4,5-(PO₄)₃, and 5'-AMP-activated protein kinase and increased carnitine palmitoyltransferase-1 mRNA expression, but it did not improve insulin activation of insulin receptor substrate (IRS)-1-dependent PI 3-kinase (IRS-1/PI3K), protein kinase B, or glycogen synthase. We found that, although insulin signaling was impaired in muscle, insulin activation of IRS-1/PI3K, IRS-2/PI3K, protein kinase B, and aPKC was largely intact in liver and that rosiglitazone improved insulin signaling to aPKC in muscle by improving responsiveness to PI-3,4,5-(PO₄)₃.
Collapse
Affiliation(s)
- Heidi K Ortmeyer
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Eight week exposure to a high sugar high fat diet results in adiposity gain and alterations in metabolic biomarkers in baboons (Papio hamadryas sp.). Cardiovasc Diabetol 2010; 9:71. [PMID: 21034486 PMCID: PMC2988722 DOI: 10.1186/1475-2840-9-71] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Accepted: 10/29/2010] [Indexed: 01/03/2023] Open
Abstract
Background Baboons (Papio hamadryas Sp.) develop features of the cardiometabolic syndrome and represent a clinically-relevant animal model in which to study the aetiology of the disorder. To further evaluate the baboon as a model for the study of the cardiometabolic syndrome, we developed a high sugar high fat diet and hypothesized that it could be used to induce adiposity gain and affect associated circulating biomarkers. Methods We developed a diet enriched with monosaccharides and saturated fatty acids that was composed of solid and liquid energy sources. We provided a group of baboons (n = 9) ad libitum access to this diet for 8 weeks. Concurrently, a control group (n = 6) was maintained with ad libitum access to a low sugar low fat baseline diet and normal water for 8 weeks. Body composition was determined by dual-energy X-ray absorptiometry and circulating metabolic biomarkers were measured using standard methodology before and after the 8 week study period. Results Neither body composition nor circulating biomarkers changed in the control group. Following the 8 weeks, the intervention group had a significant increase in fat mass (1.71 ± 0.98 vs. 3.23 ± 1.70 kg, p = 0.004), triglyceride (55 ± 13 vs. 109 ± 67 mg/dL, p = 0.006,), and leptin (1.19 ± 1.40 vs. 3.29 ± 2.32 ng/mL, p = 0.001) and a decline in adiponectin concentrations (33530 ± 9744 vs. 23330 ± 7863 ng/mL, p = 0.002). Percentage haemoglobin A1C (4.0 ± 0.3 vs. 6.0 ± 1.4, p = 0.002) also increased in the intervention group. Conclusions Our findings indicate that when exposed to a high sugar high fat diet, young adult male baboons develop increased body fat and triglyceride concentrations, altered adipokine concentrations, and evidence of altered glucose metabolism. Our findings are in keeping with observations in humans and further demonstrate the potential utility of this highly clinically-relevant animal model for studying diet-induced metabolic dysregulation.
Collapse
|
50
|
Appt SE, Ethun KF. Reproductive aging and risk for chronic disease: Insights from studies of nonhuman primates. Maturitas 2010; 67:7-14. [PMID: 20430541 PMCID: PMC2941880 DOI: 10.1016/j.maturitas.2010.03.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 03/30/2010] [Accepted: 03/31/2010] [Indexed: 01/08/2023]
Abstract
Reproductive aging and ovarian senescence have considerable public health relevance because they are associated with increased risk for coronary heart disease (CHD), osteoporosis and other degenerative conditions including cognitive decline and potentially the metabolic syndrome. It has been suggested that the hormonal dysregulation that occurs during the perimenopausal transition may play a role in the initiation of pathobiological changes (e.g., adverse lipid profiles, atherosclerotic plaques) that will increase risk for chronic disease (e.g., CHD) during the postmenopausal years. Moreover, these early changes are suspected to establish a trajectory of disease progression that may be difficult to alter if interventions are not begun until after menopause. Even a slight increase in the rate of disease progression during the pre- or perimenopausal years could have substantial consequences for health and quality of life over the postmenopausal lifespan. Thus, the years leading up to menopause may offer a "critical window" for interventions aimed at reducing the postmenopausal disease burden. The relationship between perimenopausal hormonal dysregulation and the risk for chronic disease is poorly understood due, in large part, to the lack of appropriate animal models of the perimenopausal transition and natural menopause. In this review we assesses studies of nonhuman primates (NHPs) evaluated in various reproductive stages (naturally pre-, peri- and postmenopausal, surgically menopausal) and their contribution to our understanding about risk factors for chronic disease. Finally, because large numbers of naturally perimenopausal and menopausal NHPs are not available for research at present, experimental approaches that have the potential to hasten the onset of the perimenopausal transition will be described.
Collapse
Affiliation(s)
- Susan E Appt
- Wake Forest University School of Medicine, Department of Pathology (Comparative Medicine), Wake Forest University Primate Center, Winston-Salem, NC 27157-1040, USA.
| | | |
Collapse
|