1
|
Roy S, Pokharel P, Piganelli JD. Decoding the immune dance: Unraveling the interplay between beta cells and type 1 diabetes. Mol Metab 2024; 88:101998. [PMID: 39069156 PMCID: PMC11342121 DOI: 10.1016/j.molmet.2024.101998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by the specific destruction of insulin-producing beta cells in the pancreas by the immune system, including CD4 cells which orchestrate the attack and CD8 cells which directly destroy the beta cells, resulting in the loss of glucose homeostasis. SCOPE OF REVIEW This comprehensive document delves into the complex interplay between the immune system and beta cells, aiming to shed light on the mechanisms driving their destruction in T1D. Insights into the genetic predisposition, environmental triggers, and autoimmune responses provide a foundation for understanding the autoimmune attack on beta cells. From the role of viral infections as potential triggers to the inflammatory response of beta cells, an intricate puzzle starts to unfold. This exploration highlights the importance of beta cells in breaking immune tolerance and the factors contributing to their targeted destruction. Furthermore, it examines the potential role of autophagy and the impact of cytokine signaling on beta cell function and survival. MAJOR CONCLUSIONS This review collectively represents current research findings on T1D which offers valuable perspectives on novel therapeutic approaches for preserving beta cell mass, restoring immune tolerance, and ultimately preventing or halting the progression of T1D. By unraveling the complex dynamics between the immune system and beta cells, we inch closer to a comprehensive understanding of T1D pathogenesis, paving the way for more effective treatments and ultimately a cure.
Collapse
Affiliation(s)
- Saptarshi Roy
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Pravil Pokharel
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jon D Piganelli
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States.
| |
Collapse
|
2
|
Ukegjini K, Müller PC, Warschkow R, Tarantino I, Petrowsky H, Gutschow CA, Schmied BM, Steffen T. Partial pancreatoduodenectomy versus total pancreatectomy in patients with preoperative diabetes mellitus: Comparison of surgical outcomes and quality of life. Langenbecks Arch Surg 2024; 409:254. [PMID: 39160361 DOI: 10.1007/s00423-024-03444-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/04/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
PURPOSE To reduce perioperative risks among patients with a preoperative diabetes mellitus (DM) a total pancreatectomy (TP) might be a alternative to pancreatoduodenectomy (PD). This study aimed to compare the postoperative quality of life (QoL) of patients with preoperative DM undergoing PD or TP. METHODS A single-centre retrospective study was conducted, all consecutive patients with preoperative DM undergoing PD or TP between 2011 and 2023 were identified in a prospective database. The primary endpoint was QoL, prospectively assessed using EORTC QLQ-C30 questionnaires at 3, 6, and 12 months after surgery and then annually until death. Secondary endpoints were morbidity and mortality. RESULTS Seventy-one patients were included, 17 after TP and 54 after PD. Insulin-dependent DM occurred in 21 (39%) of the PD patients. QoL was worse after TP, especially in terms of physical functioning (-31.7 points; 95% CI: -50.0 to -13.3; P < 0.001), role functioning (-41.3 points; 95% CI: -61.3 to -21.3; P < 0.001), emotional functioning (-27.5 points; 95% CI: -50.4 to -4.6; P = 0.019), fatigue symptoms (20 points; 95% CI: 2.7 to 37.4; P = 0.024) and pain symptoms (30.2 points; 95% CI: 4.1 to 56.3; P = 0.024). The rates of postoperative major complications (29% vs. 35%; P = 0.853) and mortality (11% vs. 7%; P = 0.857) were similar between TP and PD. CONCLUSION Postoperative morbidity and mortality were comparable between PD and TP, however QoL is significantly lower after TP. Importantly, patients with preoperative DM have a 60% chance of remaining noninsulin-dependent after PD.
Collapse
Affiliation(s)
- Kristjan Ukegjini
- Department of General, Visceral, Endocrine and Transplant Surgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, CH-9007 St, Gallen, Switzerland.
| | - Philip C Müller
- Department of Surgery, Clarunis - University Centre for Gastrointestinal and Hepatopancreatobiliary Diseases, Basel, Switzerland
| | - Rene Warschkow
- Department of General, Visceral, Endocrine and Transplant Surgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, CH-9007 St, Gallen, Switzerland
| | - Ignazio Tarantino
- Department of General, Visceral, Endocrine and Transplant Surgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, CH-9007 St, Gallen, Switzerland
| | - Henrik Petrowsky
- Department of Surgery and Transplantation, Swiss HPB & Transplant Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Christian A Gutschow
- Department of Surgery and Transplantation, Swiss HPB & Transplant Center Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Bruno M Schmied
- Department of General, Visceral, Endocrine and Transplant Surgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, CH-9007 St, Gallen, Switzerland
| | - Thomas Steffen
- Department of General, Visceral, Endocrine and Transplant Surgery, Kantonsspital St. Gallen, Rorschacherstrasse 95, CH-9007 St, Gallen, Switzerland
| |
Collapse
|
3
|
Guillemain G, Lacapere JJ, Khemtemourian L. Targeting hIAPP fibrillation: A new paradigm to prevent β-cell death? BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184002. [PMID: 35868406 DOI: 10.1016/j.bbamem.2022.184002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Loss of pancreatic β-cell mass is deleterious for type 2 diabetes patients since it reduces insulin production, critical for glucose homeostasis. The main research axis developed over the last few years was to generate new pancreatic β-cells or to transplant pancreatic islets as occurring for some specific type 1 diabetes patients. We evaluate here a new paradigm consisting in preservation of β-cells by prevention of human islet amyloid polypeptide (hIAPP) oligomers and fibrils formation leading to pancreatic β-cell death. We review the hIAPP physiology and the pathology that contributes to β-cell destruction, deciphering the various cellular steps that could be involved. Recent progress in understanding other amyloidosis such as Aβ, Tau, α-synuclein or prion, involved in neurodegenerative processes linked with inflammation, has opened new research lines of investigations to preserve neuronal cells. We evaluate and estimate their transposition to the pancreatic β-cells preservation. Among them is the control of reactive oxygen species (ROS) production occurring with inflammation and the possible implication of the mitochondrial translocator protein as a diagnostic and therapeutic target. The present review also focuses on other amyloid forming proteins from molecular to physiological and physiopathological points of view that could help to better decipher hIAPP-induced β-cell death mechanisms and to prevent hIAPP fibril formation.
Collapse
Affiliation(s)
- Ghislaine Guillemain
- Sorbonne Université, Institut Hospitalo-Universitaire, Inserm UMR_S938, Institute of Cardio metabolism and Nutrition (ICAN), Centre de recherche de St-Antoine (CRSA), 27 rue de Chaligny, F-75012 Paris, France.
| | - Jean-Jacques Lacapere
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS UMR 7203, Laboratoire des BioMolécules (LBM), 4 place Jussieu, F-75005 Paris, France.
| | - Lucie Khemtemourian
- CBMN, CNRS UMR 5248, IPB, Univ. Bordeaux, Allée Geoffroy Saint-Hilaire, F-33600 Pessac, France.
| |
Collapse
|
4
|
Weir GC, Butler PC, Bonner-Weir S. The β-cell glucose toxicity hypothesis: Attractive but difficult to prove. Metabolism 2021; 124:154870. [PMID: 34480921 PMCID: PMC8530963 DOI: 10.1016/j.metabol.2021.154870] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/11/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022]
Abstract
β cells in the hyperglycemic environment of diabetes have marked changes in phenotype and function that are largely reversible if glucose levels can be returned to normal. A leading hypothesis is that these changes are caused by the elevated glucose levels leading to the concept of glucose toxicity. Support for the glucose toxicity hypothesis is largely circumstantial, but little progress has been made in defining the responsible mechanisms. Then questions emerge that are difficult to answer. In the very earliest stages of diabetes development, there is a dramatic loss of glucose-induced first-phase insulin release (FPIR) with only trivial elevations of blood glucose levels. A related question is how impaired insulin action on target tissues such as liver, muscle and fat can cause increased insulin secretion. The existence of a sophisticated feedback mechanism between insulin secretion and insulin action on peripheral tissues driven by glucose has been postulated, but it has been difficult to measure increases in blood glucose levels that might have been expected. These complexities force us to challenge the simplicity of the glucose toxicity hypothesis and feedback mechanisms. It may turn out that glucose is somehow driving all of these changes, but we must develop new questions and experimental approaches to test the hypothesis.
Collapse
Affiliation(s)
- Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Peter C Butler
- Larry l. Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susan Bonner-Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Yun R, Javed AA, Jarrell AS, Crow J, Wright MJ, Burkhart RA, Rybny J, Wolfgang CL, Kruer RM. Impact of Postoperative Glycemic Control on Postoperative Morbidity in Patients Undergoing Open Pancreaticoduodenectomy. Pancreas 2021; 50:834-840. [PMID: 34347733 DOI: 10.1097/mpa.0000000000001856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE To evaluate the impact of postoperative glycemic control on postoperative morbidity in patients undergoing a pancreaticoduodenectomy. METHODS A retrospective study was performed on patients at The Johns Hopkins Hospital between April 2015 and April 2016. Data were collected on postoperative insulin regimens, blood glucose, rates of hyperglycemia and hypoglycemia, and postoperative complications and were evaluated. RESULTS Out of 244 patients, 114 (46.7%) experienced at least 1 hyperglycemic (>180 mg/dL) episode and 16 (6.6%) experienced at least 1 hypoglycemic episode (<70 mg/dL) during the first postoperative 24 hours. Early postoperative hyperglycemia (>180 mg/dL) was associated with a significantly higher rate of surgical site infections (15.7% vs 7%; P = 0.031). Late postoperative hyperglycemia (>180 mg/dL) was associated with a significantly higher rate of fistulas (4.3% vs 14.6%; P = 0.021). CONCLUSIONS Early hyperglycemia (>180 mg/dL) is associated with a higher risk of surgical site infections while late hyperglycemia is associated with a higher risk of fistulas. Intensive glucose control (<150 mg/dL) was not demonstrated to decrease the risk of postoperative complications. Similar to other critically ill populations, targeting a glucose goal of <180 mg/dL may be an appropriate target to reduce morbidity without increasing the risk of hypoglycemia.
Collapse
Affiliation(s)
- Regina Yun
- From the Department of Pharmacy, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ammar A Javed
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery
| | - Andrew S Jarrell
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | - Jessica Crow
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | - Michael J Wright
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery
| | | | - Joseph Rybny
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| | | | - Rachel M Kruer
- Department of Pharmacy, The Johns Hopkins Hospital, Baltimore, MD
| |
Collapse
|
6
|
Donovan AL, Furlan A, Borhani AA, Kalor A, Wang L, Epelboym Rossmer I, Marsh JW. Evaluation of clinical and imaging biomarkers for the prediction of new onset diabetes following pancreatic resection. Abdom Radiol (NY) 2021; 46:2628-2636. [PMID: 33474575 DOI: 10.1007/s00261-020-02943-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE To identify clinical and imaging biomarkers that can predict the new onset of diabetes mellitus (NODM) within 1 year of pancreatic resection. METHODS A retrospective chart review was conducted of 426 non-diabetic patients who underwent a pancreaticoduodenectomy or distal pancreatectomy at the University of Pittsburgh Medical Center between 2006 and 2016. Clinical characteristics and the patient's diabetic status at 1-year post resection were collected from the EMR. Imaging biomarkers included hepatic and pancreatic fat replacement, pancreatic calcifications, pancreatic duct diameter, pancreatic volume and body composition. Univariate and multivariable analyses were performed to demonstrate any predictive biomarkers of diabetes occurrence within 1 year of pancreatic resection. RESULTS 135/426 (31.7%) patients developed NODM. The only significant clinical predictor was older age (OR 1.02, 95% CI 1.002-1.039, p = 0.032). Imaging characteristics found to be significant included hepatic steatosis (OR 1.777, 95% CI 1.094-2.886, p = 0.02), larger reduction in pancreas volume (OR 0.989, 95% CI 0.979-0.999, p = 0.027), and greater preoperative visceral fat (OR 1.004, 95% CI 1.001-1.006, p = 0.001). CONCLUSION Age, presence of hepatic steatosis, change in pancreatic volume, and preoperative visceral fat are independent predictive biomarkers for NODM following pancreatic resection.
Collapse
Affiliation(s)
- Ashley L Donovan
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Alessandro Furlan
- Department of Radiology, University of Pittsburgh Medical Center, UPMC Presbyterian Campus, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - Amir A Borhani
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Ashley Kalor
- Department of Radiology, University of Pittsburgh Medical Center, UPMC Presbyterian Campus, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Li Wang
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - J Wallis Marsh
- Department of Surgery, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
7
|
Laurenti MC, Dalla Man C, Varghese RT, Andrews JC, Jones JG, Barosa C, Rizza RA, Matveyenko A, De Nicolao G, Bailey KR, Cobelli C, Vella A. Insulin Pulse Characteristics and Insulin Action in Non-diabetic Humans. J Clin Endocrinol Metab 2021; 106:1702-1709. [PMID: 33606017 PMCID: PMC8344841 DOI: 10.1210/clinem/dgab100] [Citation(s) in RCA: 3] [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] [Received: 10/28/2020] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Pulsatile insulin secretion is impaired in diseases such as type 2 diabetes that are characterized by insulin resistance. This has led to the suggestion that changes in insulin pulsatility directly impair insulin signaling. We sought to examine the effects of pulse characteristics on insulin action in humans, hypothesizing that a decrease in pulse amplitude or frequency is associated with impaired hepatic insulin action. METHODS We studied 29 nondiabetic subjects on two occasions. On 1 occasion, hepatic and peripheral insulin action was measured using a euglycemic clamp. The deuterated water method was used to estimate the contribution of gluconeogenesis to endogenous glucose production. On a separate study day, we utilized nonparametric stochastic deconvolution of frequently sampled peripheral C-peptide concentrations during fasting to reconstruct portal insulin secretion. In addition to measuring basal and pulsatile insulin secretion, we used approximate entropy to measure orderliness and Fourier transform to measure the average, and the dispersion of, insulin pulse frequencies. RESULTS In univariate analysis, basal insulin secretion (R2 = 0.16) and insulin pulse amplitude (R2 = 0.09) correlated weakly with insulin-induced suppression of gluconeogenesis. However, after adjustment for age, sex, and weight, these associations were no longer significant. The other pulse characteristics also did not correlate with the ability of insulin to suppress endogenous glucose production (and gluconeogenesis) or to stimulate glucose disappearance. CONCLUSIONS Overall, our data demonstrate that insulin pulse characteristics, considered independently of other factors, do not correlate with measures of hepatic and peripheral insulin sensitivity in nondiabetic humans.
Collapse
Affiliation(s)
- Marcello C Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Ron T Varghese
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN, USA
| | - James C Andrews
- Vascular and Interventional Radiology, Mayo Clinic, Rochester, MN, USA
| | - John G Jones
- Center for Neurosciences, University of Coimbra, Coimbra, Portugal
| | - Cristina Barosa
- Center for Neurosciences, University of Coimbra, Coimbra, Portugal
| | - Robert A Rizza
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN, USA
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Giuseppe De Nicolao
- Department of Computer Engineering and Systems Science, University of Pavia, Pavia, Italy
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN, USA
- Correspondence: Adrian Vella MD, Endocrine Research Unit, Mayo Clinic College of Medicine, 200 First ST SW, 5–194 Joseph, Rochester, MN 55905, USA.
| |
Collapse
|
8
|
Mezza T, Cefalo CMA, Cinti F, Quero G, Pontecorvi A, Alfieri S, Holst JJ, Giaccari A. Endocrine and Metabolic Insights from Pancreatic Surgery. Trends Endocrinol Metab 2020; 31:760-772. [PMID: 32830029 DOI: 10.1016/j.tem.2020.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/25/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023]
Abstract
Although it is well established that diabetes can also develop as a result of diseases or maneuvers on the exocrine pancreas, the complex relationship between glucose disorders and underlying pancreatic disease is still debated. There is evidence that several features linked to pancreatic diseases can modify endocrine and metabolic conditions before and after surgery. However, pancreatic surgery provides a rare opportunity to correlate in vivo endocrine and metabolic pathways with ex vivo pancreatic samples, to examine the endocrine and metabolic effects of acute islet removal, and finally to clarify the pathogenesis of diabetes. This approach could therefore represent a unique method to shed light on the molecular mechanisms, predicting factors, and metabolic consequences of insulin resistance, islet plasticity, β cell failure, and type 2 diabetes.
Collapse
Affiliation(s)
- Teresa Mezza
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara M A Cefalo
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesca Cinti
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Quero
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alfredo Pontecorvi
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sergio Alfieri
- Chirurgia Digestiva, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jens J Holst
- Novo Nordisk Foundation (NNF) Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Giaccari
- Endocrinologia e Diabetologia, Fondazione Policlinico Universitario Agostino Gemelli Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
9
|
Naveed A, Farrukh L, Sana MK, Naveed B, Randhawa FA. Pharmacological Primary Prevention of Diabetes Mellitus Type II: A Narrative Review. Cureus 2020; 12:e10033. [PMID: 32999773 PMCID: PMC7521466 DOI: 10.7759/cureus.10033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022] Open
Abstract
The evolving epidemic of type 2 diabetes mellitus has challenged health-care professionals. It stands among the leading causes of mortality in the present world. It warrants new and versatile approaches to improve mortality and the associated huge quality-adjusted life years lost to it once diagnosed. A possible venue to lower the incidence is to assess the safety and efficacy of various diabetes prevention strategies. Diet and exercise have a well-developed role in the prevention of weight gain and, ultimately, diabetes mellitus type II in high-risk individuals. However, high-risk individuals can also benefit from adjunct pharmacotherapy. In light of this information, we decided to conduct a systematic review of randomized controlled trials. This article summarizes the evidence in the literature on the pharmacological prevention of diabetes in high-risk individuals.
Collapse
Affiliation(s)
- Ali Naveed
- Internal Medicine, King Edward Medical University & Mayo Hospital, Lahore, PAK
| | - Larabe Farrukh
- Internal Medicine, King Edward Medical University & Mayo Hospital, Lahore, PAK
| | | | - Bazigh Naveed
- Internal Medicine, King Edward Medical University & Mayo Hospital, Lahore, PAK
| | - Fawad Ahmad Randhawa
- Diabetes and Endocrinology, King Edward Medical University & Mayo Hospital, Lahore, PAK
| |
Collapse
|
10
|
Nagpal SJS, Kandlakunta H, Her T, Sharma A, Sannapaneni S, Smyrk TC, Velamala P, Garg SK, Rakshit K, Majumder S, Chari S, Matveyenko A. Pancreatic ductal adenocarcinoma is associated with a unique endocrinopathy distinct from type 2 diabetes mellitus. Pancreatology 2020; 20:929-935. [PMID: 32620407 DOI: 10.1016/j.pan.2020.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/10/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The majority of patients with pancreatic ductal adenocarcinoma (PC) display either impaired fasting glucose/glucose intolerance or overt diabetes. However, the pathophysiologic basis of this association remains largely unexplained. METHODS In this case-control study we aimed to study the morphological changes in the islets of patients with PC, compared to control patients with and without type 2 diabetes mellitus (T2DM). T2DM controls and PC cases had a lower β-cell area and average islet size and density compared to non-T2DM controls (p < 0.05). RESULTS Compared to both T2DM and non-T2DM controls, mean α-cell area was significantly lower and β/α-ratio was higher in PC cases (p < 0.05). Furthermore, whereas islets in T2DM controls were characterized by disrupted islet architecture and presence of islet amyloid aggregates, islet composition in PC islets was not significantly different compared to non-T2DM controls (p > 0.05 vs. Control). CONCLUSIONS Our data shows that PC is associated with a unique pattern of islet pathology characterized by preserved architecture, absence of amyloid aggregates, and relative α-cell loss indicating that distinct mechanisms are likely involved in the pathophysiology of islet failure in PC-induced DM. Insights into the mechanisms mediating β-cell failure in PC can be important for our understanding of pathophysiology of PC.
Collapse
Affiliation(s)
- Sajan Jiv Singh Nagpal
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, UChicago Medicine, Chicago, IL, USA
| | - Harika Kandlakunta
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Tracy Her
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Ayush Sharma
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Shilpa Sannapaneni
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Thomas C Smyrk
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Pruthvi Velamala
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sushil K Garg
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Shounak Majumder
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Suresh Chari
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Gastroenterology, Hepatology and Nutrition, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, and Nutrition, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
| |
Collapse
|
11
|
|
12
|
Weir GC, Gaglia J, Bonner-Weir S. Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes. Lancet Diabetes Endocrinol 2020; 8:249-256. [PMID: 32006519 PMCID: PMC7098467 DOI: 10.1016/s2213-8587(20)30022-x] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
Abstract
For patients with type 1 diabetes, it is accepted among the scientific community that there is a marked reduction in β-cell mass; however, with type 2 diabetes, there is disagreement as to whether this reduction in mass occurs in every case. Some have argued that β-cell mass in some patients with type 2 diabetes is normal and that the cause of the hyperglycaemia in these patients is a functional abnormality of insulin secretion. In this Personal View, we argue that a deficient β-cell mass is essential for the development of type 2 diabetes. The main point is that there are enormous (≥10 fold) variations in insulin sensitivity and insulin secretion in the general population, with a very close correlation between these two factors for any individual. Although β-cell mass cannot be accurately measured in living patients, it is highly likely that it too is highly correlated with insulin sensitivity and secretion. Thus, our argument is that a person with type 2 diabetes can have a β-cell mass that is the same as a person without type 2 diabetes, but because they are insulin resistant, the mass is inadequate and responsible for their diabetes. Because the abnormal insulin secretion of diabetes is caused by dysglycaemia and can be largely reversed with glycaemic control, it is a less serious problem than the reduction in β-cell mass, which is far more difficult to restore.
Collapse
Affiliation(s)
- Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Jason Gaglia
- Section on Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susan Bonner-Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
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: 1.3] [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
|
14
|
Westermeier F, Holyoak T, Asenjo JL, Gatica R, Nualart F, Burbulis I, Bertinat R. Gluconeogenic Enzymes in β-Cells: Pharmacological Targets for Improving Insulin Secretion. Trends Endocrinol Metab 2019; 30:520-531. [PMID: 31213347 DOI: 10.1016/j.tem.2019.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/14/2019] [Accepted: 05/16/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic β-cells express the gluconeogenic enzymes glucose 6-phosphatase (G6Pase), fructose 1,6-bisphosphatase (FBP), and phosphoenolpyruvate (PEP) carboxykinase (PCK), which modulate glucose-stimulated insulin secretion (GSIS) through their ability to reverse otherwise irreversible glycolytic steps. Here, we review current knowledge about the expression and regulation of these enzymes in the context of manipulating them to improve insulin secretion in diabetics. Because the regulation of gluconeogenic enzymes in β-cells is so poorly understood, we propose novel research avenues to study these enzymes as modulators of insulin secretion and β-cell dysfunction, with especial attention to FBP, which constitutes an attractive target with an inhibitor under clinical evaluation at present.
Collapse
Affiliation(s)
- Francisco Westermeier
- FH JOANNEUM Gesellschaft mbH University of Applied Sciences, Institute of Biomedical Science, Eggenberger Allee 13, 8020 Graz, Austria
| | - Todd Holyoak
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Joel L Asenjo
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Independencia 631, 5110566 Valdivia, Chile
| | - Rodrigo Gatica
- Escuela de Veterinaria, Facultad de Ciencias, Universidad Mayor, La Pirámide 5750, 8580745 Santiago, Chile
| | - Francisco Nualart
- Centro de Microscopía Avanzada, CMA BIO, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160 C, 4030000 Concepción, Chile
| | - Ian Burbulis
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Jordan Hall Room 6022, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA; Escuela de Medicina, Universidad San Sebastián, Sede Patagonia, Lago Panguipulli 1390, 5501842 Puerto Montt, Chile
| | - Romina Bertinat
- Centro de Microscopía Avanzada, CMA BIO, Facultad de Ciencias Biológicas, Universidad de Concepción, Casilla 160 C, 4030000 Concepción, Chile.
| |
Collapse
|
15
|
Laurenti MC, Vella A, Varghese RT, Andrews JC, Sharma A, Kittah NE, Rizza RA, Matveyenko A, De Nicolao G, Cobelli C, Dalla Man C. Assessment of pulsatile insulin secretion derived from peripheral plasma C-peptide concentrations by nonparametric stochastic deconvolution. Am J Physiol Endocrinol Metab 2019; 316:E687-E694. [PMID: 30807214 PMCID: PMC6580177 DOI: 10.1152/ajpendo.00519.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/01/2019] [Accepted: 02/17/2019] [Indexed: 01/08/2023]
Abstract
The characteristics of pulsatile insulin secretion are important determinants of type 2 diabetes pathophysiology, but they are understudied due to the difficulties in measuring pulsatile insulin secretion noninvasively. Deconvolution of either peripheral C-peptide or insulin concentrations offers an appealing alternative to hepatic vein catheterization. However, to do so, there are a series of methodological challenges to overcome. C-peptide has a relatively long half-life and accumulates in the circulation. On the other hand, peripheral insulin concentrations reflect relatively fast clearance and hepatic extraction as it leaves the portal circulation to enter the systemic circulation. We propose a method based on nonparametric stochastic deconvolution of C-peptide concentrations, using individually determined C-peptide kinetics, to overcome these limitations. The use of C-peptide (instead of insulin) concentrations allows estimation of portal (and not post-hepatic) insulin pulses, whereas nonparametric stochastic deconvolution allows evaluation of pulsatile signals without any a priori assumptions of pulse shape and occurrence. The only assumption required is the degree of smoothness of the (unknown) secretion rate. We tested this method first on simulated data and then on 29 nondiabetic subjects studied during euglycemia and hyperglycemia and compared our estimates with the profiles obtained from hepatic vein insulin concentrations. This method produced satisfactory results both in the ability to fit the data and in providing reliable estimates of pulsatile secretion, in agreement with hepatic vein measurements. In conclusion, the proposed method enables reliable and noninvasive measurement of pulsatile insulin secretion. Future studies will be needed to validate this method in people with type 2 diabetes.
Collapse
Affiliation(s)
- Marcello C Laurenti
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
- Department of Information Engineering, University of Padua , Padua , Italy
| | - Adrian Vella
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
| | - Ron T Varghese
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
| | - James C Andrews
- Vascular and Interventional Radiology, Mayo Clinic , Rochester, Minnesota
| | - Anu Sharma
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
| | - Nana Esi Kittah
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
| | - Robert A Rizza
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic , Rochester, Minnesota
- Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Giuseppe De Nicolao
- Department of Computer Engineering and Systems Science, University of Pavia , Pavia , Italy
| | - Claudio Cobelli
- Department of Information Engineering, University of Padua , Padua , Italy
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padua , Padua , Italy
| |
Collapse
|
16
|
Diabetes after pancreaticoduodenectomy: can we predict it? J Surg Res 2018; 227:211-219. [PMID: 29804855 DOI: 10.1016/j.jss.2018.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 12/07/2017] [Accepted: 02/13/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND There is limited literature about the perioperative factors which can predict endocrine insufficiency after pancreaticoduodenectomy (PD). The primary aim was to correlate percentage pancreatic remnant volume (%RV) after PD in nondiabetic patients with the development of new-onset impaired glucose tolerance/diabetes mellitus (IGT/DM). The secondary aim was to identify the risk factors for new-onset IGT/DM. METHODS In this prospective study, all consecutive patients with resectable periampullary carcinoma and without IGT/DM were evaluated with fasting and postprandial plasma glucose, HbA1c, insulin, and C-peptide levels preoperatively and at 3 mo postoperatively. After that, all patients were followed up with fasting and postprandial plasma glucose level assessed at 3-mo intervals for 24 mo or till death, whichever occurred earlier. The %RV was determined from computed tomography measurements preoperatively. RESULTS Of the 50 patients, 11 (22%) patients developed IGT/DM after median follow-up of 32 mo. The patients' with/without IGT/DM were similar in demographic/perioperative variables. The %RV was found to be an independent factor associated with new-onset IGT/DM. A %RV of <48.8% was found to be a predictor of new-onset IGT/DM (sensitivity, 89.7%; specificity, 73.6%). Plasma sugar and glycosylated hemoglobin levels were significantly higher postoperatively after PD than the preoperative levels. Insulin and C-peptide levels were significantly lower after PD, irrespective of new-onset IGT/DM. CONCLUSIONS The incidence of IGT/DM after PD was 22%, and %RV < 48.8% was found to be a significant risk factor for new-onset IGT/DM. (CTRI/2013/12/004233).
Collapse
|
17
|
Hou J, Li Z, Zhong W, Hao Q, Lei L, Wang L, Zhao D, Xu P, Zhou Y, Wang Y, Xu T. Temporal Transcriptomic and Proteomic Landscapes of Deteriorating Pancreatic Islets in Type 2 Diabetic Rats. Diabetes 2017; 66:2188-2200. [PMID: 28559245 DOI: 10.2337/db16-1305] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 05/17/2017] [Indexed: 11/13/2022]
Abstract
Progressive reduction in β-cell mass and function comprise the core of the pathogenesis mechanism of type 2 diabetes. The process of deteriorating pancreatic islets, in which a complex network of molecular events is involved, is not yet fully characterized. We used RNA sequencing and tandem mass tag-based quantitative proteomics technology to measure the temporal mRNA and protein expression changes of pancreatic islets in Goto-Kakizaki (GK) rats from 4 to 24 weeks of age. Our omics data set outlines the dynamics of the molecular network during the deterioration of GK islets as two stages: The early stage (4-6 weeks) is characterized by anaerobic glycolysis, inflammation priming, and compensation for insulin synthesis, and the late stage (8-24 weeks) is characterized by inflammation amplification and compensation failure. Further time course analysis allowed us to reveal 5,551 differentially expressed genes, a large portion of which have not been reported before. Our comprehensive and temporal transcriptome and proteome data offer a valuable resource for the diabetes research community and for quantitative biology.
Collapse
Affiliation(s)
- Junjie Hou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zonghong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Northeast Normal University, Changchun, China
| | - Wen Zhong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Science and Technology, HuaZhong University of Science and Technology, Wuhan, China
| | - Qiang Hao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lei Lei
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Linlin Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Dongyu Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingyong Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yifa Zhou
- School of Life Sciences, Northeast Normal University, Changchun, China
| | - You Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Science and Technology, HuaZhong University of Science and Technology, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc 2017. [DOI: 10.1111/brv.12349] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sara Al-Khawaga
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Bushra Memon
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Alexandra E. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine; University of California; Los Angeles CA 90095 U.S.A
| | - Shahrad Taheri
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Abdul B. Abou-Samra
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Essam M. Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
19
|
Md Moin AS, Dhawan S, Cory M, Butler PC, Rizza RA, Butler AE. Increased Frequency of Hormone Negative and Polyhormonal Endocrine Cells in Lean Individuals With Type 2 Diabetes. J Clin Endocrinol Metab 2016; 101:3628-3636. [PMID: 27472443 PMCID: PMC5052343 DOI: 10.1210/jc.2016-2496] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
CONTEXT It has been suggested that beta cell loss in type 2 diabetes (T2D) may be due to beta cell degranulation and/or altered cell identity. While shown to have a minor role in obese T2D, this has not been evaluated in lean T2D. OBJECTIVE To establish the contribution of altered beta cell identity in lean T2D and, using a rodent model of lean T2D, whether changes in beta cell identity precede hyperglycemia. DESIGN, SETTING, AND PARTICIPANTS We investigated the frequency of chromogranin A positive hormone negative (CPHN) and polyhormonal endocrine cells in pancreas from 10 lean nondiabetic and 10 lean T2D subjects and in pancreas from wild-type and human IAPP transgenic rats at the prediabetic and diabetic stages. RESULTS CPHN cells and polyhormonal-expressing cells were comparably increased in lean T2D and human IAPP transgenic rats, in the latter both before and at onset of diabetes. However, the extent of these cells could only account for approximately 2% of beta cell loss. CONCLUSION Degranulation and altered identity play at most a minor role in the beta cell deficit in lean T2D. Because the increase in CPHN and polyhormonal cells precede diabetes onset, these changes are likely a response to stress rather than hyperglycemia, and may reflect attempted regeneration.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Sangeeta Dhawan
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Megan Cory
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Robert A Rizza
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| | - Alexandra E Butler
- Larry L. Hillblom Islet Research Center (A.S.M.M., S.D., M.C., P.C.B., A.E.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, MN 55905
| |
Collapse
|
20
|
Yagihashi S, Inaba W, Mizukami H. Dynamic pathology of islet endocrine cells in type 2 diabetes: β-Cell growth, death, regeneration and their clinical implications. J Diabetes Investig 2016; 7:155-65. [PMID: 27042265 PMCID: PMC4773678 DOI: 10.1111/jdi.12424] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 08/28/2015] [Accepted: 08/30/2015] [Indexed: 12/12/2022] Open
Abstract
Diabetes is defined as a disease of hyperglycemic metabolic disorder caused by impaired insulin action or low insulin secretion, resulting in the occurrence of vascular complications. Based on this definition, diabetes therapy has long been oriented to correct hyperglycemia against the specific complications of diabetes. This definition has posed some difficulties, however, in understanding of the pathophysiology of this complicated disease and as such in the establishment of an effective treatment. With continuing efforts to explore the structural basis for diabetes onset and methodological development of immunohistochemistry, progressive decline of β-cells is now established as a salient feature of type 2 diabetes. Accordingly, diabetes therapy has now turned out to protect β-cells concurrently with the correction of hyperglycemia. Together with this effort, exploration of the means to regenerate β-cells or to supply new β-cells by, for example, induced pluripotential stem cells, are vigorously made with the search for the mechanism of β-cell decline in diabetes. In the present review, we describe the advances in the islet pathology in type 2 diabetes with special reference to the dynamic alterations of islet endocrine cells in the milieu of maturation, obesity, aging and ethnic differences. The effect of amyloid deposition is also discussed. We hope it will help with understanding the pathophysiology of diabetes, and suggest the future direction of diabetes treatment.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Wataru Inaba
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hiroki Mizukami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
21
|
Butler AE, Dhawan S, Hoang J, Cory M, Zeng K, Fritsch H, Meier JJ, Rizza RA, Butler PC. β-Cell Deficit in Obese Type 2 Diabetes, a Minor Role of β-Cell Dedifferentiation and Degranulation. J Clin Endocrinol Metab 2016; 101:523-32. [PMID: 26700560 PMCID: PMC4880126 DOI: 10.1210/jc.2015-3566] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONTEXT Type 2 diabetes is characterized by a β-cell deficit and a progressive defect in β-cell function. It has been proposed that the deficit in β-cells may be due to β-cell degranulation and transdifferentiation to other endocrine cell types. OBJECTIVE The objective of the study was to establish the potential impact of β-cell dedifferentiation and transdifferentiation on β-cell deficit in type 2 diabetes and to consider the alternative that cells with an incomplete identity may be newly forming rather than dedifferentiated. DESIGN, SETTING, AND PARTICIPANTS Pancreata obtained at autopsy were evaluated from 14 nondiabetic and 13 type 2 diabetic individuals, from four fetal cases, and from 10 neonatal cases. RESULTS Whereas there was a slight increase in islet endocrine cells expressing no hormone in type 2 diabetes (0.11 ± 0.03 cells/islet vs 0.03 ± 0.01 cells/islet, P < .01), the impact on the β-cell deficit would be minimal. Furthermore, we established that the deficit in β-cells per islet cannot be accounted for by an increase in other endocrine cell types. The distribution of hormone negative endocrine cells in type 2 diabetes (most abundant in cells scattered in the exocrine pancreas) mirrors that in developing (embryo and neonatal) pancreas, implying that these may represent newly forming cells. CONCLUSIONS Therefore, although we concur that in type 2 diabetes there are endocrine cells with altered cell identity, this process does not account for the deficit in β-cells in type 2 diabetes but may reflect, in part, attempted β-cell regeneration.
Collapse
Affiliation(s)
- Alexandra E Butler
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Sangeeta Dhawan
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Jonathan Hoang
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Megan Cory
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Kylie Zeng
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Helga Fritsch
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Juris J Meier
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Robert A Rizza
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center (A.E.B., S.D., J.H., M.C., K.Z., P.C.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095-7073; Institute of Pathology (H.F.), Division of Clinical and Functional Anatomy, Medical University of Innsbruck, A-6020 Innsbruck, Austria; St Josef Hospital of the Ruhr-University Bochum (J.J.M.), 44791 Bochum, Germany; and Division of Endocrinology, Diabetes, Metabolism, and Nutrition (R.A.R.), Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|
22
|
Glucose Homeostatic Law: Insulin Clearance Predicts the Progression of Glucose Intolerance in Humans. PLoS One 2015; 10:e0143880. [PMID: 26623647 PMCID: PMC4666631 DOI: 10.1371/journal.pone.0143880] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/10/2015] [Indexed: 12/31/2022] Open
Abstract
Homeostatic control of blood glucose is regulated by a complex feedback loop between glucose and insulin, of which failure leads to diabetes mellitus. However, physiological and pathological nature of the feedback loop is not fully understood. We made a mathematical model of the feedback loop between glucose and insulin using time course of blood glucose and insulin during consecutive hyperglycemic and hyperinsulinemic-euglycemic clamps in 113 subjects with variety of glucose tolerance including normal glucose tolerance (NGT), impaired glucose tolerance (IGT) and type 2 diabetes mellitus (T2DM). We analyzed the correlation of the parameters in the model with the progression of glucose intolerance and the conserved relationship between parameters. The model parameters of insulin sensitivity and insulin secretion significantly declined from NGT to IGT, and from IGT to T2DM, respectively, consistent with previous clinical observations. Importantly, insulin clearance, an insulin degradation rate, significantly declined from NGT, IGT to T2DM along the progression of glucose intolerance in the mathematical model. Insulin clearance was positively correlated with a product of insulin sensitivity and secretion assessed by the clamp analysis or determined with the mathematical model. Insulin clearance was correlated negatively with postprandial glucose at 2h after oral glucose tolerance test. We also inferred a square-law between the rate constant of insulin clearance and a product of rate constants of insulin sensitivity and secretion in the model, which is also conserved among NGT, IGT and T2DM subjects. Insulin clearance shows a conserved relationship with the capacity of glucose disposal among the NGT, IGT and T2DM subjects. The decrease of insulin clearance predicts the progression of glucose intolerance.
Collapse
|
23
|
Saisho Y. β-cell dysfunction: Its critical role in prevention and management of type 2 diabetes. World J Diabetes 2015; 6:109-124. [PMID: 25685282 PMCID: PMC4317303 DOI: 10.4239/wjd.v6.i1.109] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 08/17/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2DM) is characterized by insulin resistance and β-cell dysfunction. Although, in contrast to type 1 diabetes, insulin resistance is assumed to be a major pathophysiological feature of T2DM, T2DM never develops unless β-cells fail to compensate insulin resistance. Recent studies have revealed that a deficit of β-cell functional mass is an essential component of the pathophysiology of T2DM, implying that β-cell deficit is a common feature of both type 1 and type 2 diabetes. β-cell dysfunction is present at the diagnosis of T2DM and progressively worsens with disease duration. β-cell dysfunction is associated with worsening of glycemic control and treatment failure; thus, it is important to preserve or recover β-cell functional mass in the management of T2DM. Since β-cell regenerative capacity appears somewhat limited in humans, reducing β-cell workload appears to be the most effective way to preserve β-cell functional mass to date, underpinning the importance of lifestyle modification and weight loss for the treatment and prevention of T2DM. This review summarizes the current knowledge on β-cell functional mass in T2DM and discusses the treatment strategy for T2DM.
Collapse
|
24
|
Farmer TD, Jenkins EC, O'Brien TP, McCoy GA, Havlik AE, Nass ER, Nicholson WE, Printz RL, Shiota M. Comparison of the physiological relevance of systemic vs. portal insulin delivery to evaluate whole body glucose flux during an insulin clamp. Am J Physiol Endocrinol Metab 2015; 308:E206-22. [PMID: 25516552 PMCID: PMC4312835 DOI: 10.1152/ajpendo.00406.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To understand the underlying pathology of metabolic diseases, such as diabetes, an accurate determination of whole body glucose flux needs to be made by a method that maintains key physiological features. One such feature is a positive differential in insulin concentration between the portal venous and systemic arterial circulation (P/S-IG). P/S-IG during the determination of the relative contribution of liver and extra-liver tissues/organs to whole body glucose flux during an insulin clamp with either systemic (SID) or portal (PID) insulin delivery was examined with insulin infusion rates of 1, 2, and 5 mU·kg(-1)·min(-1) under either euglycemic or hyperglycemic conditions in 6-h-fasted conscious normal rats. A P/S-IG was initially determined with endogenous insulin secretion to exist with a value of 2.07. During an insulin clamp, while inhibiting endogenous insulin secretion by somatostatin, P/S-IG remained at 2.2 with PID, whereas, P/S-IG disappeared completely with SID, which exhibited higher arterial and lower portal insulin levels compared with PID. Consequently, glucose disappearance rates and muscle glycogen synthetic rates were higher, but suppression of endogenous glucose production and liver glycogen synthetic rates were lower with SID compared with PID. When the insulin clamp was performed with SID at 2 and 5 mU·kg(-1)·min(-1) without managing endogenous insulin secretion under euglycemic but not hyperglycemic conditions, endogenous insulin secretion was completely suppressed with SID, and the P/S-IG disappeared. Thus, compared with PID, an insulin clamp with SID underestimates the contribution of liver in response to insulin to whole body glucose flux.
Collapse
Affiliation(s)
- Tiffany D Farmer
- Diabetes Research Training Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Erin C Jenkins
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Tracy P O'Brien
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Gregory A McCoy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Allison E Havlik
- Diabetes Research Training Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Erik R Nass
- Diabetes Research Training Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wendell E Nicholson
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Richard L Printz
- Diabetes Research Training Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Masakazu Shiota
- Diabetes Research Training Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| |
Collapse
|
25
|
Pulsatile insulin secretion, impaired glucose tolerance and type 2 diabetes. Mol Aspects Med 2015; 42:61-77. [PMID: 25637831 DOI: 10.1016/j.mam.2015.01.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 12/28/2022]
Abstract
Type 2 diabetes (T2DM) results when increases in beta cell function and/or mass cannot compensate for rising insulin resistance. Numerous studies have documented the longitudinal changes in metabolism that occur during the development of glucose intolerance and lead to T2DM. However, the role of changes in insulin secretion, both amount and temporal pattern, has been understudied. Most of the insulin secreted from pancreatic beta cells of the pancreas is released in a pulsatile pattern, which is disrupted in T2DM. Here we review the evidence that changes in beta cell pulsatility occur during the progression from glucose intolerance to T2DM in humans, and contribute significantly to the etiology of the disease. We review the evidence that insulin pulsatility improves the efficacy of secreted insulin on its targets, particularly hepatic glucose production, but also examine evidence that pulsatility alters or is altered by changes in peripheral glucose uptake. Finally, we summarize our current understanding of the biophysical mechanisms responsible for oscillatory insulin secretion. Understanding how insulin pulsatility contributes to normal glucose homeostasis and is altered in metabolic disease states may help improve the treatment of T2DM.
Collapse
|
26
|
Robertson RP, Raymond RH, Lee DS, Calle RA, Ghosh A, Savage PJ, Shankar SS, Vassileva MT, Weir GC, Fryburg DA. Arginine is preferred to glucagon for stimulation testing of β-cell function. Am J Physiol Endocrinol Metab 2014; 307:E720-7. [PMID: 25159323 PMCID: PMC4200308 DOI: 10.1152/ajpendo.00149.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A key aspect of research into the prevention and treatment of type 2 diabetes is the availability of reproducible clinical research methodology to assess β-cell function. One commonly used method employs nonglycemic secretagogues like arginine (arg) or glucagon (glgn). This study was designed to quantify the insulin response to arg and glgn and determine test repeatability and tolerability. Obese overnight-fasted subjects with normal glucose tolerance were studied on 4 separate days: twice using arg (5 g iv) and twice with glgn (1 mg iv). Pre- and postinfusion samples for plasma glucose, insulin, and C-peptide were acquired. Arg and glgn challenges were repeated in the last 10 min of a 60-min glucose (900 mg/min) infusion. Insulin and C-peptide secretory responses were estimated under baseline fasting glucose conditions (AIRarg and AIRglgn) and hyperglycemic (AIRargMAX AIRglgnMAX) states. Relative repeatability was estimated by intraclass correlation coefficient (ICC). Twenty-three (12 men and 11 women) subjects were studied (age: 42.4 ± 8.3 yr; BMI: 31.4 ± 2.8 kg/m²). Geometric means (95% CI) for baseline-adjusted values AIRarg and AIRglgn were 84 (75-95) and 102 (90-115) μU/ml, respectively. After the glucose infusion, AIRargMAX and AIRglgnMAX were 395 (335-466) and 483 (355-658) μU/ml, respectively. ICC values were >0.90 for AIRarg andAIRargMAX. Glucagon ICCs were 0.83, 0.34, and 0.36, respectively, although the exclusion of one outlier increased the latter two values (to 0.84 and 0.86). Both glgn and arg induced mild adverse events that were transient. Glucagon, but not arginine, induced moderate adverse events due to nausea. Taken together, arginine is preferred to glucagon for assessment of β-cell function.
Collapse
Affiliation(s)
- R Paul Robertson
- Pacific Northwest Diabetes Research Institute, Seattle, Washington
| | | | | | | | | | - Peter J Savage
- National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | | | - Maria T Vassileva
- Foundation for the National Institutes of Health, Bethesda, Maryland
| | | | | | | |
Collapse
|
27
|
Abstract
The early occurrence of β‐cell dysfunction has been broadly recognized as a critical determinant of the development and progression of type 2 diabetes. β‐cell dysfunction might be induced by insufficient β‐cell mass, by a dysfunction of the β‐cells, or both. Whether or not β‐cell dysfunction constitutes a cause of reduced β‐cells or vice‐versa currently remains unclear. The results of some studies have measured the loss of β‐cells in type 2 diabetic patients at between 22 and 63% by planimetric measurements. Because β‐cell hypertrophy has been noted in type 2 diabetic patients, the loss of β‐cell number should prove more profound than what has thus far been reported. Furthermore, β‐cell volumes are reduced even in patients with impaired fasting glucose. Such defects in β‐cell mass are associated with increased apoptosis rather than insufficient replication or neogenesis of β‐cells. With these results, although they still require clarification, the peak β‐cell mass might be determined at quite an early stage of life, and then might decline progressively over time as the result of exposure to harmful environmental influences over one’s lifetime. In this review, we have summarized the relevant studies regarding β‐cell mass in patients with type 2 diabetes, and then presented a review of the various causes of β‐cell loss in adults. (J Diabetes Invest, doi: 10.1111/j.2040‐1124.2010.00072.x, 2010)
Collapse
Affiliation(s)
- Jae-Hyoung Cho
- Department of Endocrinology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Ji-Won Kim
- Department of Endocrinology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jeong-Ah Shin
- Department of Endocrinology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Juyoung Shin
- Department of Endocrinology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Kun-Ho Yoon
- Department of Endocrinology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
28
|
Malapan K, Chang PC, Huang CK. Metabolic surgery for diabetes mellitus after pancreatectomy. Surg Obes Relat Dis 2013; 10:e43-5. [PMID: 24238731 DOI: 10.1016/j.soard.2013.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 10/26/2022]
Affiliation(s)
- Kirubakaran Malapan
- Department of Bariatric and Metabolic International Surgery Center, E-Da Hospital, Kaohsiung City, Taiwan
| | - Po-Chih Chang
- Department of Bariatric and Metabolic International Surgery Center, E-Da Hospital, Kaohsiung City, Taiwan
| | - Chih-Kun Huang
- Department of Bariatric and Metabolic International Surgery Center, E-Da Hospital, Kaohsiung City, Taiwan.
| |
Collapse
|
29
|
Meier JJ, Bonadonna RC. Role of reduced β-cell mass versus impaired β-cell function in the pathogenesis of type 2 diabetes. Diabetes Care 2013; 36 Suppl 2:S113-9. [PMID: 23882035 PMCID: PMC3920783 DOI: 10.2337/dcs13-2008] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Juris J Meier
- Division of Diabetology and Gastrointestinal Endocrinology, St. Josef-Hospital, Ruhr-University of Bochum, Bochum, Germany.
| | | |
Collapse
|
30
|
Meier JJ, Pennartz C, Schenker N, Menge BA, Schmidt WE, Heise T, Kapitza C, Veldhuis JD. Hyperglycaemia is associated with impaired pulsatile insulin secretion: effect of basal insulin therapy. Diabetes Obes Metab 2013; 15:258-63. [PMID: 23039360 DOI: 10.1111/dom.12022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 08/17/2012] [Accepted: 10/01/2012] [Indexed: 01/09/2023]
Abstract
AIM Postprandial insulin pulsatility is impaired in patients with type 2 diabetes, but the effects of exogenous insulin therapy on pulsatile insulin secretion are not known. We addressed, whether pulsatile insulin secretion is related to glycaemic control, whether basal insulin supplementation increases postprandial insulin secretion, and if so, is this accomplished by a specific improvement in pulsatile insulin secretion? METHODS Fourteen patients with type 2 diabetes underwent a mixed meal test before and after an 8-week treatment period with insulin glargine. Glucose, insulin and C-peptide levels were measured, and insulin pulsatility was determined by deconvolution analysis. RESULTS Insulin treatment lowered fasting glycaemia from 179.6 ± 7.5 mg/dl to 117.6 ± 6.5 mg/dl (p < 0.001). Postprandial insulin and C-peptide levels increased significantly after the treatment period (p < 0.0001). The total calculated insulin secretion rate increased with insulin treatment (p = 0.0039), with non-significant increases in both pulsatile and non-pulsatile insulin secretion. Insulin pulse frequency was unchanged by the intervention. There was an inverse relationship between fasting and postprandial glycaemia and insulin pulse mass (r(2) = 0.51 and 0.56, respectively), whereas non-pulsatile insulin secretion was unrelated to either fasting or postprandial glucose concentrations (r(2) = 0.0073 and 0.031). CONCLUSIONS Hyperglycaemia in type 2 diabetes is associated with a reduction in postprandial insulin secretion, specifically through a reduction in insulin pulsatility. Reducing chronic hyperglycaemia by basal insulin therapy enhances endogenous β-cell function in the postprandial state. These data support the use of basal insulin regimens in the pharmacotherapy of overtly hyperglycaemic patients with type 2 diabetes.
Collapse
Affiliation(s)
- J J Meier
- Diabetes Division, Department of Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Type 1 and some forms of type 2 diabetes mellitus are caused by deficiency of insulin-secretory islet β cells. An ideal treatment for these diseases would therefore be to replace β cells, either by transplanting donated islets or via endogenous regeneration (and controlling the autoimmunity in type 1 diabetes). Unfortunately, the poor availability of donor islets has severely restricted the broad clinical use of islet transplantation. The ability to differentiate embryonic stem cells into insulin-expressing cells initially showed great promise, but the generation of functional β cells has proven extremely difficult and far slower than originally hoped. Pancreatic stem cells (PSC) or transdifferentiation of other cell types in the pancreas may hence provide an alternative renewable source of surrogate β cells. However, the existence of PSC has been hotly debated for many years. In this review, we will discuss the latest development and future perspectives of PSC research, giving readers an overview of this controversial but important area.
Collapse
Affiliation(s)
- Fang-Xu Jiang
- Centre for Diabetes Research, Western Australian Institute for Medical Research, The University of Western Australia, 50 Murray St (Rear), Perth, WA 6000, Australia.
| | | |
Collapse
|
32
|
Patterson ME, Mao CS, Yeh MW, Ipp E, Cortina G, Barank D, Vasinrapee P, Pawlikowska-Haddal A, Lee WNP, Yee JK. Hyperinsulinism presenting in childhood and treatment by conservative pancreatectomy. Endocr Pract 2012; 18:e52-6. [PMID: 22548943 DOI: 10.4158/ep11232.cr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To describe the uncommon presentation of hyperinsulinism in an 8-year-old boy. METHODS We describe the patient's clinical findings, results from biochemical and imaging studies, surgical approach, and outcome. The discussion encompasses a review of literature that provided the basis for the diagnostic and surgical approach applied to this patient's case. RESULTS An obese 8.5-year-old boy initially presented with hypoglycemic seizures after initiation of dietary changes to treat obesity. Biochemical analysis indicated hyperinsulinism. Endoscopic ultrasonography showed no pancreatic lesions suggestive of insulinoma. Genetic studies identified no known mutations in the ABCC8, KCNJ11, GCK, or GLUD1 genes. Selective arterial calcium stimulation and hepatic venous sampling did not document a focal source for hyperinsulinism in the pancreas, and positron emission tomography with 18-fluoro-L-3,4-dihydroxyphenylalanine showed diffusely increased uptake in the pancreas. The patient ultimately required partial pancreatectomy because of continued hypoglycemia while taking diazoxide and octreotide. Intraoperative glucose monitoring directed the extent of surgical resection. A 45% pancreatectomy was performed, which resolved the hypoglycemia but led to impaired glucose tolerance after surgery. CONCLUSION The unusual presentation of hyperinsulinism in childhood required a personalized approach to diagnosis and surgical management using intraoperative glucose monitoring that resulted in a conservative pancreatectomy.
Collapse
Affiliation(s)
- Mary E Patterson
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, California 90509, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Matveyenko AV, Liuwantara D, Gurlo T, Kirakossian D, Dalla Man C, Cobelli C, White MF, Copps KD, Volpi E, Fujita S, Butler PC. Pulsatile portal vein insulin delivery enhances hepatic insulin action and signaling. Diabetes 2012; 61:2269-79. [PMID: 22688333 PMCID: PMC3425431 DOI: 10.2337/db11-1462] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Insulin is secreted as discrete insulin secretory bursts at ~5-min intervals into the hepatic portal vein, these pulses being attenuated early in the development of type 1 and type 2 diabetes mellitus (T2DM). Intraportal insulin infusions (pulsatile, constant, or reproducing that in T2DM) indicated that the pattern of pulsatile insulin secretion delivered via the portal vein is important for hepatic insulin action and, therefore, presumably for hepatic insulin signaling. To test this, we examined hepatic insulin signaling in rat livers exposed to the same three patterns of portal vein insulin delivery by use of sequential liver biopsies in anesthetized rats. Intraportal delivery of insulin in a constant versus pulsatile pattern led to delayed and impaired activation of hepatic insulin receptor substrate (IRS)-1 and IRS-2 signaling, impaired activation of downstream insulin signaling effector molecules AKT and Foxo1, and decreased expression of glucokinase (Gck). We further established that hepatic Gck expression is decreased in the HIP rat model of T2DM, a defect that correlated with a progressive defect of pulsatile insulin secretion. We conclude that the physiological pulsatile pattern of insulin delivery is important in hepatic insulin signaling and glycemic control. Hepatic insulin resistance in diabetes is likely in part due to impaired pulsatile insulin secretion.
Collapse
Affiliation(s)
- Aleksey V. Matveyenko
- Larry Hillblom Islet Research Center, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - David Liuwantara
- Larry Hillblom Islet Research Center, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tatyana Gurlo
- Larry Hillblom Islet Research Center, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - David Kirakossian
- Larry Hillblom Islet Research Center, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Chiara Dalla Man
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Morris F. White
- Howard Hughes Medical Institute, Division of Endocrinology, Children’s Hospital, Boston, Massachusetts
| | - Kyle D. Copps
- Howard Hughes Medical Institute, Division of Endocrinology, Children’s Hospital, Boston, Massachusetts
| | - Elena Volpi
- Department of Internal Medicine and Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| | - Satoshi Fujita
- Department of Internal Medicine and Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| | - Peter C. Butler
- Larry Hillblom Islet Research Center, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Corresponding author: Peter C. Butler,
| |
Collapse
|
34
|
Kavanagh K, Wylie AT, Chavanne TJ, Jorgensen MJ, Voruganti VS, Comuzzie AG, Kaplan JR, McCall CE, Kritchevsky SB. Aging does not reduce heat shock protein 70 in the absence of chronic insulin resistance. J Gerontol A Biol Sci Med Sci 2012; 67:1014-21. [PMID: 22403054 DOI: 10.1093/gerona/gls008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heat shock protein (HSP)70 decreases with age. Often aging is associated with coincident insulin resistance and higher blood glucose levels, which also associate with lower HSP70. We aimed to understand how these factors interrelate through a series of experiments using vervet monkeys (Chlorocebus aethiops sabaeous). Monkeys (n = 284, 4-25 years) fed low-fat diets showed no association of muscle HSP70 with age (r = .04, p = .53), but levels were highly heritable. Insulin resistance was induced in vervet monkeys with high-fat diets, and muscle biopsies were taken after 0.3 or 6 years. HSP70 levels were significantly greater after 0.3 years (+72%, p < .05) but were significantly lower following 6 years of high-fat diet (-77%, p < .05). Associations with glucose also switched from being positive (r = .44, p = .03) to strikingly negative (r = -.84, p < .001) with increasing insulin resistance. In conclusion, a low-fat diet may preserve tissue HSP70 and health with aging, whereas high-fat diets, insulin resistance, and genetic factors may be more important than age for determining HSP70 levels.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kanatsuna N, Papadopoulos GK, Moustakas AK, Lenmark Å. Etiopathogenesis of insulin autoimmunity. ANATOMY RESEARCH INTERNATIONAL 2012; 2012:457546. [PMID: 22567309 PMCID: PMC3335545 DOI: 10.1155/2012/457546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 01/12/2012] [Indexed: 12/12/2022]
Abstract
Autoimmunity against pancreatic islet beta cells is strongly associated with proinsulin, insulin, or both. The insulin autoreactivity is particularly pronounced in children with young age at onset of type 1 diabetes. Possible mechanisms for (pro)insulin autoimmunity may involve beta-cell destruction resulting in proinsulin peptide presentation on HLA-DR-DQ Class II molecules in pancreatic draining lymphnodes. Recent data on proinsulin peptide binding to type 1 diabetes-associated HLA-DQ2 and -DQ8 is reviewed and illustrated by molecular modeling. The importance of the cellular immune reaction involving cytotoxic CD8-positive T cells to kill beta cells through Class I MHC is discussed along with speculations of the possible role of B lymphocytes in presenting the proinsulin autoantigen over and over again through insulin-carrying insulin autoantibodies. In contrast to autoantibodies against other islet autoantigens such as GAD65, IA-2, and ZnT8 transporters, it has not been possible yet to standardize the insulin autoantibody test. As islet autoantibodies predict type 1 diabetes, it is imperative to clarify the mechanisms of insulin autoimmunity.
Collapse
Affiliation(s)
- Norio Kanatsuna
- Department of Clinical Sciences, Skåne University Hospital (SUS), Lund University, CRC Ing 72 Building 91:10, 205 02 Malmö, Sweden
| | - George K. Papadopoulos
- Laboratory of Biochemistry and Biophysics, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, 47100 Arta, Greece
| | - Antonis K. Moustakas
- Department of Organic Farming, Technological Educational Institute of Ionian Islands, 27100 Argostoli, Greece
| | - Åke Lenmark
- Department of Clinical Sciences, Skåne University Hospital (SUS), Lund University, CRC Ing 72 Building 91:10, 205 02 Malmö, Sweden
| |
Collapse
|
36
|
Shirakawa S, Matsumoto I, Toyama H, Shinzeki M, Ajiki T, Fukumoto T, Ku Y. Pancreatic volumetric assessment as a predictor of new-onset diabetes following distal pancreatectomy. J Gastrointest Surg 2012; 16:2212-9. [PMID: 23054900 PMCID: PMC3508270 DOI: 10.1007/s11605-012-2039-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 09/19/2012] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Pancreatogenic diabetes after pancreatectomy is of growing importance due to the increasing life expectancy of pancreatectomized patients. Although reduction of pancreatic volume is thought to affect glucose metabolism, a consistent relationship has yet to be determined. This study aimed to investigate functional consequences of distal pancreatectomy (DP) in preoperatively non-diabetic patients. METHODS This study included 61 non-diabetic patients who underwent DP. Clinical data were obtained, and the percent resected volume (PRV) of each pancreas was determined via multi-detector row computed tomography volumetry. RESULTS During the follow-up period (median 26 months), 22 patients (36 %) developed new-onset diabetes within a median onset time of 8 months (range 0.5-42 months) postoperatively. The remaining 39 patients also showed impaired glucose metabolism. Multivariate analysis identified preoperative hemoglobin A1c ≥ 5.7 % (odds ratio 15.6, p = 0.001) and PRV > 44 % (odds ratio 11.3, p = 0.004) as independent risk factors for new-onset diabetes. CONCLUSIONS Key determinants of postoperative glycemic control include preoperative functional reserve of the endocrine pancreas and the volume reduction of pancreatic parenchyma. Our findings enable reliable preoperative evaluation of the risk of postoperative diabetes and appropriate postoperative surveillance, which is helpful for early intervention in high risk patients.
Collapse
Affiliation(s)
- Sachiyo Shirakawa
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Ippei Matsumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Makoto Shinzeki
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Tetsuo Ajiki
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Takumi Fukumoto
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| | - Yonson Ku
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017 Japan
| |
Collapse
|
37
|
Saisho Y, Manesso E, Butler AE, Galasso R, Kavanagh K, Flynn M, Zhang L, Clark P, Gurlo T, Toffolo GM, Cobelli C, Wagner JD, Butler PC. Ongoing beta-cell turnover in adult nonhuman primates is not adaptively increased in streptozotocin-induced diabetes. Diabetes 2011; 60:848-56. [PMID: 21270238 PMCID: PMC3046845 DOI: 10.2337/db09-1368] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE β-Cell turnover and its potential to permit β-cell regeneration in adult primates are unknown. Our aims were 1) to measure β-cell turnover in adult nonhuman primates; 2) to establish the relative contribution of β-cell replication and formation of new β-cells from other precursors (defined thus as β-cell neogenesis); and 3) to establish whether there is an adaptive increase in β-cell formation (attempted regeneration) in streptozotocin (STZ)-induced diabetes in adult nonhuman primates. RESEARCH DESIGN AND METHODS Adult (aged 7 years) vervet monkeys were administered STZ (45-55 mg/kg, n = 7) or saline (n = 9). Pancreas was obtained from each animal twice, first by open surgical biopsy and then by euthanasia. β-Cell turnover was evaluated by applying a mathematic model to measured replication and apoptosis rates. RESULTS β-Cell turnover is present in adult nonhuman primates (3.3 ± 0.9 mg/month), mostly (~80%) derived from β-cell neogenesis. β-Cell formation was minimal in STZ-induced diabetes. Despite marked hyperglycemia, β-cell apoptosis was not increased in monkeys administered STZ. CONCLUSIONS There is ongoing β-cell turnover in adult nonhuman primates that cannot be accounted for by β-cell replication. There is no evidence of β-cell regeneration in monkeys administered STZ. Hyperglycemia does not induce β-cell apoptosis in nonhuman primates in vivo.
Collapse
Affiliation(s)
- Yoshifumi Saisho
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Erica Manesso
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Alexandra E. Butler
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Ryan Galasso
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Kylie Kavanagh
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Mickey Flynn
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Li Zhang
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Paige Clark
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Tatyana Gurlo
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Gianna M. Toffolo
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Claudio Cobelli
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Janice D. Wagner
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Peter C. Butler
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, California
- Corresponding author: Peter C. Butler,
| |
Collapse
|
38
|
Kavanagh K, Flynn DM, Nelson C, Zhang L, Wagner JD. Characterization and validation of a streptozotocin-induced diabetes model in the vervet monkey. J Pharmacol Toxicol Methods 2011; 63:296-303. [PMID: 21356321 DOI: 10.1016/j.vascn.2011.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 02/03/2011] [Accepted: 02/17/2011] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Streptozotocin (STZ), preferentially toxic to pancreatic beta cells, is commonly used to model Type 1 diabetes mellitus (DM) in numerous species, including nonhuman primates. METHODS We induced DM in twenty vervet monkeys (Chlorocebus aethiops) by intravenous administration of either 45 (n=8, STZ-45) or 55 mg/kg STZ (n=12, STZ-55); ten control (CTL) monkeys received saline. RESULTS Overall there was 15% mortality, likely secondary to renal toxicity. Twice-daily insulin therapy was initiated to maintain comparable glycemic control, confirmed by comparable glycated hemoglobin levels. Exogenous insulin requirements increased rapidly for 4weeks; STZ-45 insulin doses stabilized thereafter while STZ-55 doses continued to increase through 16weeks. Glucose tolerance testing and arginine-stimulated insulin secretion confirmed 80-90% reduction in pancreatic beta cell function in both groups. Body weight was reduced in all STZ monkeys, with return to baseline only in STZ-45 at 16 wks. Elevated blood urea nitrogen (BUN) and creatinine were noted in the STZ-55 group. Alkaline phosphatase (ALKP) was also increased with STZ-55 (p < 0.05 vs. CTL) whereas STZ-45 ALKP elevation resolved by study end. Red cell parameters were reduced in all STZ monkeys, but more severely in the STZ-55 group. DISCUSSION We have demonstrated that a model of DM can be induced and maintained in vervets with a single dose of STZ. The lower dose of STZ (45 mg/kg) significantly improved the toxicity profile without altering efficacy in inducing DM. Finally, sufficient time following induction is recommended to allow transient renal, hepatic and hematologic parameters to resolve.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | | | | | |
Collapse
|
39
|
Mirasierra M, Fernández-Pérez A, Díaz-Prieto N, Vallejo M. Alx3-deficient mice exhibit decreased insulin in beta cells, altered glucose homeostasis and increased apoptosis in pancreatic islets. Diabetologia 2011; 54:403-14. [PMID: 21104068 DOI: 10.1007/s00125-010-1975-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Accepted: 10/20/2010] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Homeodomain transcription factors play an important role in the regulation of pancreatic islet function. In previous studies we determined that aristaless-like homeobox 3 (ALX3) is produced in islet cells, binds to the promoter of the insulin gene and regulates its expression. The purpose of the present study was to investigate the functional role of ALX3 in pancreatic islets and its possible involvement in the regulation of glucose homeostasis in vivo. METHODS Alx3-knockout mice were used. Glucose and insulin tolerance tests were carried out, and serum insulin concentrations were determined. Isolated islets were used to test insulin secretion and gene expression. The pancreatic islets were also studied using both confocal and conventional microscopy. RESULTS ALX3 deficiency resulted in increased blood glucose levels and impaired glucose tolerance in the presence of normal serum insulin concentrations. Insulin, glucagon and glucokinase expression were reduced in Alx3-null pancreatic islets. Reduced insulin content was reflected by decreased insulin secretion from isolated islets. Alx3-deficient islets also showed increased apoptosis, and morphometric analyses indicated that they were, on average, of smaller size than islets from control mice. ALX3 deficiency resulted in reduced beta cell mass. Finally, mature Alx3-null mice developed age-dependent insulin resistance due to impaired peripheral insulin receptor signalling. CONCLUSIONS/INTERPRETATION ALX3 participates in the regulation of the expression of essential genes for the function of pancreatic islets, and its deficiency alters the regulation of glucose homeostasis in vivo. We suggest that ALX3 constitutes a potential candidate to consider in the aetiopathogenesis of diabetes mellitus.
Collapse
Affiliation(s)
- M Mirasierra
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas /Universidad Autónoma de Madrid, Calle Arturo Duperier 4, 28029 Madrid, Spain
| | | | | | | |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW To provide an overview of the genetics of type 2 diabetes in the context of recent progress in the understanding of the genetic architecture of the disease and its applicability to the pathogenesis of the disease as well as efforts to individualize therapy in type 2 diabetes. Efforts are underway to understand how these loci alter measurable physiologic processes in nondiabetic humans. However, it is important to understand the potential pitfalls in such studies and the limitations underlying measurement of insulin secretion and action using qualitative methodologies. RECENT FINDINGS The availability of large population-based cohorts and the ease with which large numbers of common genetic variants can be genotyped has enabled the discovery of multiple loci and pathways associated with type 2 diabetes. Recent efforts examining quantitative traits such as fasting glucose concentrations have led to the discovery of other genes likely to be important in the development of diabetes. SUMMARY The past 4 years have witnessed a significant increase in our understanding of genetic predisposition to type 2 diabetes. Hopefully more progress will be made in applying this knowledge to the pathophysiology of type 2 diabetes in the coming years.
Collapse
Affiliation(s)
- Galina Smushkin
- Division of Endocrinology & Metabolism, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
41
|
Ingelsson E, Langenberg C, Hivert MF, Prokopenko I, Lyssenko V, Dupuis J, Mägi R, Sharp S, Jackson AU, Assimes TL, Shrader P, Knowles JW, Zethelius B, Abbasi FA, Bergman RN, Bergmann A, Berne C, Boehnke M, Bonnycastle LL, Bornstein SR, Buchanan TA, Bumpstead SJ, Böttcher Y, Chines P, Collins FS, Cooper CC, Dennison EM, Erdos MR, Ferrannini E, Fox CS, Graessler J, Hao K, Isomaa B, Jameson KA, Kovacs P, Kuusisto J, Laakso M, Ladenvall C, Mohlke KL, Morken MA, Narisu N, Nathan DM, Pascoe L, Payne F, Petrie JR, Sayer AA, Schwarz PEH, Scott LJ, Stringham HM, Stumvoll M, Swift AJ, Syvänen AC, Tuomi T, Tuomilehto J, Tönjes A, Valle TT, Williams GH, Lind L, Barroso I, Quertermous T, Walker M, Wareham NJ, Meigs JB, McCarthy MI, Groop L, Watanabe RM, Florez JC. Detailed physiologic characterization reveals diverse mechanisms for novel genetic Loci regulating glucose and insulin metabolism in humans. Diabetes 2010; 59:1266-75. [PMID: 20185807 PMCID: PMC2857908 DOI: 10.2337/db09-1568] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Recent genome-wide association studies have revealed loci associated with glucose and insulin-related traits. We aimed to characterize 19 such loci using detailed measures of insulin processing, secretion, and sensitivity to help elucidate their role in regulation of glucose control, insulin secretion and/or action. RESEARCH DESIGN AND METHODS We investigated associations of loci identified by the Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC) with circulating proinsulin, measures of insulin secretion and sensitivity from oral glucose tolerance tests (OGTTs), euglycemic clamps, insulin suppression tests, or frequently sampled intravenous glucose tolerance tests in nondiabetic humans (n = 29,084). RESULTS The glucose-raising allele in MADD was associated with abnormal insulin processing (a dramatic effect on higher proinsulin levels, but no association with insulinogenic index) at extremely persuasive levels of statistical significance (P = 2.1 x 10(-71)). Defects in insulin processing and insulin secretion were seen in glucose-raising allele carriers at TCF7L2, SCL30A8, GIPR, and C2CD4B. Abnormalities in early insulin secretion were suggested in glucose-raising allele carriers at MTNR1B, GCK, FADS1, DGKB, and PROX1 (lower insulinogenic index; no association with proinsulin or insulin sensitivity). Two loci previously associated with fasting insulin (GCKR and IGF1) were associated with OGTT-derived insulin sensitivity indices in a consistent direction. CONCLUSIONS Genetic loci identified through their effect on hyperglycemia and/or hyperinsulinemia demonstrate considerable heterogeneity in associations with measures of insulin processing, secretion, and sensitivity. Our findings emphasize the importance of detailed physiological characterization of such loci for improved understanding of pathways associated with alterations in glucose homeostasis and eventually type 2 diabetes.
Collapse
Affiliation(s)
- Erik Ingelsson
- Corresponding authors: Erik Ingelsson, ; Leif Groop, ; Richard M. Watanabe, ; Jose C. Florez,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Leif Groop
- Corresponding authors: Erik Ingelsson, ; Leif Groop, ; Richard M. Watanabe, ; Jose C. Florez,
| | - Richard M. Watanabe
- Corresponding authors: Erik Ingelsson, ; Leif Groop, ; Richard M. Watanabe, ; Jose C. Florez,
| | - Jose C. Florez
- Corresponding authors: Erik Ingelsson, ; Leif Groop, ; Richard M. Watanabe, ; Jose C. Florez,
| | | |
Collapse
|
42
|
Saisho Y, Butler AE, Manesso E, Galasso R, Zhang L, Gurlo T, Toffolo GM, Cobelli C, Kavanagh K, Wagner JD, Butler PC. Relationship between fractional pancreatic beta cell area and fasting plasma glucose concentration in monkeys. Diabetologia 2010; 53:111-4. [PMID: 19847395 PMCID: PMC2789926 DOI: 10.1007/s00125-009-1552-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 08/27/2009] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS We sought to establish the relationship between fasting plasma glucose concentrations and pancreatic fractional beta cell area in adult cynomolgus monkeys (Macaca fascicularis). METHODS Fasting plasma glucose and pancreatic fractional beta cell area were measured in 18 control and 17 streptozotocin-treated adult primates (17.0 +/- 1.2 vs 15.4 +/- 1.2 years old). RESULTS Fasting plasma glucose was increased (12.0 +/- 2.0 vs 3.4 +/- 0.1 mmol/l, p < 0.01) and fractional beta cell area was decreased (0.62 +/- 0.13% vs 2.49 +/- 0.35%, p < 0.01) in streptozotocin-treated monkeys. The relationship between fasting plasma glucose and pancreatic fractional beta cell area was described by a wide range of beta cell areas in controls. In streptozotocin-treated monkeys there was an inflection of fasting blood glucose at approximately 50% of the mean beta cell area in controls with a steep increase in blood glucose for each further decrement in beta cell area. CONCLUSIONS/INTERPRETATION In adult non-human primates a decrement in fractional beta cell area of approximately 50% or more leads to loss of glycaemic control.
Collapse
Affiliation(s)
- Y. Saisho
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
| | - A. E. Butler
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
| | - E. Manesso
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
- Department of Information Engineering, University of Padova, Padua, Italy
| | - R. Galasso
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
| | - L. Zhang
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - T. Gurlo
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
| | - G. M. Toffolo
- Department of Information Engineering, University of Padova, Padua, Italy
| | - C. Cobelli
- Department of Information Engineering, University of Padova, Padua, Italy
| | - K. Kavanagh
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - J. D. Wagner
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC USA
| | - P. C. Butler
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, 900 Weyburn Place #A, Los Angeles, CA 90024-2852 USA
| |
Collapse
|
43
|
Li X, Shu YH, Xiang AH, Trigo E, Kuusisto J, Hartiala J, Swift AJ, Kawakubo M, Stringham HM, Bonnycastle LL, Lawrence JM, Laakso M, Allayee H, Buchanan TA, Watanabe RM. Additive effects of genetic variation in GCK and G6PC2 on insulin secretion and fasting glucose. Diabetes 2009; 58:2946-53. [PMID: 19741163 PMCID: PMC2780888 DOI: 10.2337/db09-0228] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Glucokinase (GCK) and glucose-6-phosphatase catalytic subunit 2 (G6PC2) regulate the glucose-cycling step in pancreatic beta-cells and may regulate insulin secretion. GCK rs1799884 and G6PC2 rs560887 have been independently associated with fasting glucose, but their interaction on glucose-insulin relationships is not well characterized. RESEARCH DESIGN AND METHODS We tested whether these variants are associated with diabetes-related quantitative traits in Mexican Americans from the BetaGene Study and attempted to replicate our findings in Finnish men from the METabolic Syndrome in Men (METSIM) Study. RESULTS rs1799884 was not associated with any quantitative trait (corrected P > 0.1), whereas rs560887 was significantly associated with the oral glucose tolerance test 30-min incremental insulin response (30' Deltainsulin, corrected P = 0.021). We found no association between quantitative traits and the multiplicative interaction between rs1799884 and rs560887 (P > 0.26). However, the additive effect of these single nucleotide polymorphisms was associated with fasting glucose (corrected P = 0.03) and 30' Deltainsulin (corrected P = 0.027). This additive association was replicated in METSIM (fasting glucose, P = 3.5 x 10(-10) 30' Deltainsulin, P = 0.028). When we examined the relationship between fasting glucose and 30' Deltainsulin stratified by GCK and G6PC2, we noted divergent changes in these quantitative traits for GCK but parallel changes for G6PC2. We observed a similar pattern in METSIM. CONCLUSIONS Our data suggest that variation in GCK and G6PC2 have additive effects on both fasting glucose and insulin secretion.
Collapse
Affiliation(s)
- Xia Li
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yu-Hsiang Shu
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Anny H. Xiang
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Enrique Trigo
- Department of Medicine, Division of Endocrinology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Johanna Kuusisto
- Department of Medicine, University of Kuopio and Kuopio University Hospital, Kuopio, Finland
| | - Jaana Hartiala
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Amy J. Swift
- Genome Technology Branch, National Human Genome Research Institute, Bethesda, Maryland
| | - Miwa Kawakubo
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Heather M. Stringham
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Lori L. Bonnycastle
- Genome Technology Branch, National Human Genome Research Institute, Bethesda, Maryland
| | - Jean M. Lawrence
- Research and Evaluation, Kaiser Permanente of Southern California, Pasadena, California
| | - Markku Laakso
- Department of Medicine, University of Kuopio and Kuopio University Hospital, Kuopio, Finland
| | - Hooman Allayee
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Institute for Genetic Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Thomas A. Buchanan
- Department of Medicine, Division of Endocrinology, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Richard M. Watanabe
- Department of Preventive Medicine, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Corresponding author: Richard M. Watanabe,
| |
Collapse
|
44
|
|
45
|
Schrader H, Menge BA, Breuer TGK, Ritter PR, Uhl W, Schmidt WE, Holst JJ, Meier JJ. Impaired glucose-induced glucagon suppression after partial pancreatectomy. J Clin Endocrinol Metab 2009; 94:2857-63. [PMID: 19491219 DOI: 10.1210/jc.2009-0826] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The glucose-induced decline in glucagon levels is often lost in patients with type 2 diabetes. It is unclear whether this is due to an independent defect in alpha-cell function or secondary to the impairment in insulin secretion. We examined whether a partial pancreatectomy in humans would also impair postchallenge glucagon concentrations and, if so, whether this could be attributed to the reduction in insulin levels. PATIENTS AND METHODS Thirty-six patients with pancreatic tumours or chronic pancreatitis were studied before and after approximately 50% pancreatectomy with a 240-min oral glucose challenge, and the plasma concentrations of glucose, insulin, C-peptide, and glucagon were determined. RESULTS Fasting and postchallenge insulin and C-peptide levels were significantly lower after partial pancreatectomy (P < 0.0001). Likewise, fasting glucagon concentrations tended to be lower after the intervention (P = 0.11). Oral glucose ingestion elicited a decline in glucagon concentrations before surgery (P < 0.0001), but this was lost after partial pancreatectomy (P < 0.01 vs. preoperative values). The loss of glucose-induced glucagon suppression was found after both pancreatic head (P < 0.001) and tail (P < 0.05) resection. The glucose-induced changes in glucagon levels were closely correlated to the respective increments in insulin and C-peptide concentrations (P < 0.01). CONCLUSIONS The glucose-induced suppression in glucagon levels is lost after a 50% partial pancreatectomy in humans. This suggests that impaired alpha-cell function in patients with type 2 diabetes may also be secondary to reduced beta-cell mass. Alterations in glucagon regulation should be considered as a potential side effect of partial pancreatectomies.
Collapse
Affiliation(s)
- Henning Schrader
- Department of Medicine I, St. Josef-Hospital, Ruhr-University Bochum, Gudrunstrasse 56, Bochum, Germany
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Delghingaro-Augusto V, Nolan CJ, Gupta D, Jetton TL, Latour MG, Peshavaria M, Madiraju SRM, Joly E, Peyot ML, Prentki M, Leahy J. Islet beta cell failure in the 60% pancreatectomised obese hyperlipidaemic Zucker fatty rat: severe dysfunction with altered glycerolipid metabolism without steatosis or a falling beta cell mass. Diabetologia 2009; 52:1122-32. [PMID: 19294363 DOI: 10.1007/s00125-009-1317-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Accepted: 01/07/2009] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS The Zucker fatty (ZF) rat subjected to 60% pancreatectomy (Px) develops moderate diabetes by 3 weeks. We determined whether a progressive fall in beta cell mass and/or beta cell dysfunction contribute to beta cell failure in this type 2 diabetes model. METHODS Partial (60%) or sham Px was performed in ZF and Zucker lean (ZL) rats. At 3 weeks post-surgery, beta cell mass and proliferation, proinsulin biosynthesis, pancreatic insulin content, insulin secretion, and islet glucose and lipid metabolism were measured. RESULTS ZL-Px rats maintained normal glycaemia and glucose-stimulated insulin secretion (GSIS) despite incomplete recovery of beta cell mass possibly due to compensatory enhanced islet glucose metabolism and lipolysis. ZF-Px rats developed moderate hyperglycaemia (14 mmol/l), hypertriacylglycerolaemia and relative hypoinsulinaemia. Despite beta cell mass recovery and normal arginine-induced insulin secretion, GSIS and pancreatic insulin content were profoundly lowered in ZF-Px rats. Proinsulin biosynthesis was not reduced. Compensatory increases in islet glucose metabolism above those observed in ZF-Sham rats were not seen in ZF-Px rats. Triacylglycerol content was not increased in ZF-Px islets, possibly due to lipodetoxification by enhanced lipolysis and fatty acid oxidation. Fatty acid accumulation into monoacylglycerol and diacylglycerol was increased in ZF-Px islets together with a 4.5-fold elevation in stearoyl-CoA desaturase mRNA expression. CONCLUSIONS/INTERPRETATION Falling beta cell mass, reduced proinsulin biosynthesis and islet steatosis are not implicated in early beta cell failure and glucolipotoxicity in ZF-Px rats. Rather, severe beta cell dysfunction with a specific reduction in GSIS and marked depletion of beta cell insulin stores with altered lipid partitioning underlie beta cell failure in this animal model of type 2 diabetes.
Collapse
Affiliation(s)
- V Delghingaro-Augusto
- Molecular Nutrition Unit and the Montreal Diabetes Research Center, CR-CHUM, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Matveyenko AV, Gurlo T, Daval M, Butler AE, Butler PC. Successful versus failed adaptation to high-fat diet-induced insulin resistance: the role of IAPP-induced beta-cell endoplasmic reticulum stress. Diabetes 2009; 58:906-16. [PMID: 19151199 PMCID: PMC2661593 DOI: 10.2337/db08-1464] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Obesity is a known risk factor for type 2 diabetes. However, most obese individuals do not develop diabetes because they adapt to insulin resistance by increasing beta-cell mass and insulin secretion. Islet pathology in type 2 diabetes is characterized by beta-cell loss, islet amyloid derived from islet amyloid polypeptide (IAPP), and increased beta-cell apoptosis characterized by endoplasmic reticulum (ER) stress. We hypothesized that IAPP-induced ER stress distinguishes successful versus unsuccessful islet adaptation to insulin resistance. RESEARCH DESIGN AND METHODS To address this, we fed wild-type (WT) and human IAPP transgenic (HIP) rats either 10 weeks of regular chow or a high-fat diet and prospectively examined the relations among beta-cell mass and turnover, beta-cell ER stress, insulin secretion, and insulin sensitivity. RESULTS A high-fat diet led to comparable insulin resistance in WT and HIP rats. WT rats compensated with increased insulin secretion and beta-cell mass. In HIP rats, in contrast, neither beta-cell function nor mass compensated for the increased insulin demand, leading to diabetes. The failure to increase beta-cell mass in HIP rats was the result of ER stress-induced beta-cell apoptosis that increased in proportion to diet-induced insulin resistance. CONCLUSIONS IAPP-induced ER stress distinguishes the successful versus unsuccessful islet adaptation to a high-fat diet in rats. These studies are consistent with the hypothesis that IAPP oligomers contribute to increased beta-cell apoptosis and beta-cell failure in humans with type 2 diabetes.
Collapse
Affiliation(s)
- Aleksey V Matveyenko
- Larry Hillblom Islet Research Center, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA.
| | | | | | | | | |
Collapse
|
48
|
Menge BA, Schrader H, Breuer TGK, Dabrowski Y, Uhl W, Schmidt WE, Meier JJ. Metabolic consequences of a 50% partial pancreatectomy in humans. Diabetologia 2009; 52:306-17. [PMID: 19037627 DOI: 10.1007/s00125-008-1219-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2008] [Accepted: 10/05/2008] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Partial pancreatectomy is frequently performed in patients with pancreatic tumours or chronic pancreatitis, but little is known about the metabolic impact of this intervention. We examined the effects of approximately 50% partial pancreatectomy on glucose homeostasis and insulin secretion. METHODS Fourteen patients with chronic pancreatitis, ten patients with pancreatic carcinoma and 13 patients with benign pancreatic tumours or extra-pancreatic masses (control group) underwent 240 min oral glucose tolerance tests before and after pancreatic tail-resection (n = 12), duodenopancreatectomy (n = 19) or duodenum-preserving pancreatic-head resection (n = 6). RESULTS Partial pancreatectomy led to a reduction in post-challenge insulin excursions by 49% in chronic pancreatitis patients, 52% in carcinoma patients and 55% in controls (p < 0.05). Nevertheless, post-challenge glucose concentrations were transiently ameliorated after surgery (p < 0.001). In the control participants, pancreatic-head resection caused a transient reduction of post-challenge glycaemia, whereas pancreatic-tail resection increased both fasting and post-challenge glycaemia (p < 0.05). Insulin sensitivity was highest in chronic pancreatitis patients before surgery (p < 0.01), but remained unchanged by the partial pancreatectomy. High pre-operative body weight and elevated fasting glucose levels were associated with poor glycaemic control after surgery. CONCLUSIONS/INTERPRETATION Insulin secretion is diminished after pancreatic-head and -tail resection, but post-challenge glucose concentrations can be ameliorated after pancreatic-head resection. These data highlight the unequal impact of different surgical procedures on glucose control and suggest that obesity and high pre-operative glucose levels should be considered as risk factors for the development of hyperglycaemia after pancreatic surgery.
Collapse
Affiliation(s)
- B A Menge
- Department of Medicine I, St Josef-Hospital, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Regulation of blood glucose concentrations requires an adequate number of beta-cells that respond appropriately to blood glucose levels. beta-Cell mass cannot yet be measured in humans in vivo, necessitating autopsy studies, although both pre- and postmorbid changes may confound this approach. Autopsy studies report deficits in beta-cell mass ranging from 0 to 65% in type 2 diabetes (T2DM), and approximately 70-100% in type 1 diabetes (T1DM), and, when evaluated, increased beta-cell apoptosis in both T1DM and T2DM. A deficit of beta-cell mass of approximately 50% in animal studies leads to impaired insulin secretion (when evaluated directly in the portal vein) and induction of insulin resistance. We postulate three phases for diabetes progression. Phase 1: selective beta-cell cytotoxicity (autoimmune in T1DM, unknown in T2DM) leading to impaired beta-cell function and gradual loss of beta-cell mass through apoptosis. Phase 2: decompensation of glucose control when the pattern of portal vein insulin secretion is sufficiently impaired to cause hepatic insulin resistance. Phase 3: adverse consequences of glucose toxicity accelerate beta-cell dysfunction and insulin resistance. The relative contribution of beta-cell loss versus beta-cell dysfunction to diabetes onset remains an area of controversy. However, because cytotoxicity sufficient to induce beta-cell apoptosis predictably disturbs beta-cell function, it is naive to attempt to distinguish the relative contributions of these linked processes to diabetes onset.
Collapse
Affiliation(s)
- A V Matveyenko
- Larry Hillblom Islet Research Center, UCLA David Geffen School of Medicine, Los Angeles, CA 90095-7073, USA.
| | | |
Collapse
|
50
|
Affiliation(s)
- Jack L Leahy
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Vermont, Burlington, Vermont, USA.
| |
Collapse
|