1
|
Moon JH, Choe HJ, Lim S. Pancreatic beta-cell mass and function and therapeutic implications of using antidiabetic medications in type 2 diabetes. J Diabetes Investig 2024; 15:669-683. [PMID: 38676410 PMCID: PMC11143426 DOI: 10.1111/jdi.14221] [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: 01/21/2024] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Nowadays, the focus of diabetes treatment has switched from lowering the glucose level to preserving glycemic homeostasis and slowing the disease progression. The main pathophysiology of both type 1 diabetes and long-standing type 2 diabetes is pancreatic β-cell mass loss and dysfunction. According to recent research, human pancreatic β-cells possess the ability to proliferate in response to elevated insulin demands. It has been demonstrated that in insulin-resistant conditions in humans, such as obesity or pregnancy, the β-cell mass increases. This ability could be helpful in developing novel treatment approaches to restore a functional β-cell mass. Treatment strategies aimed at boosting β-cell function and mass may be a useful tool for managing diabetes mellitus and stopping its progression. This review outlines the processes of β-cell failure and detail the many β-cell abnormalities that manifest in people with diabetes mellitus. We also go over standard techniques for determining the mass and function of β-cells. Lastly, we provide the therapeutic implications of utilizing antidiabetic drugs in controlling the mass and function of pancreatic β-cells.
Collapse
Affiliation(s)
- Joon Ho Moon
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| | - Hun Jee Choe
- Department of Internal MedicineHallym University Dongtan Sacred Heart HospitalHwaseongSouth Korea
| | - Soo Lim
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| |
Collapse
|
2
|
Chaari A, Saikia N, Paul P, Yousef M, Ding F, Ladjimi M. Experimental and computational investigation of the effect of Hsc70 structural variants on inhibiting amylin aggregation. Biophys Chem 2024; 309:107235. [PMID: 38608617 DOI: 10.1016/j.bpc.2024.107235] [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: 01/06/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
The misfolding and aggregation of human islet amyloid polypeptide (hIAPP), also known as amylin, have been implicated in the pathogenesis of type 2 diabetes (T2D). Heat shock proteins, specifically, heat shock cognate 70 (Hsc70), are molecular chaperones that protect against hIAPP misfolding and inhibits its aggregation. Nevertheless, there is an incomplete understanding of the mechanistic interactions between Hsc70 domains and hIAPP, thus limiting their potential therapeutic role in diabetes. This study investigates the inhibitory capacities of different Hsc70 variants, aiming to identify the structural determinants that strike a balance between efficacy and cytotoxicity. Our experimental findings demonstrate that the ATPase activity of Hsc70 is not a pivotal factor for inhibiting hIAPP misfolding. We underscore the significance of the C-terminal substrate-binding domain of Hsc70 in inhibiting hIAPP aggregation, emphasizing that the removal of the lid subdomain diminishes the inhibitory effect of Hsc70. Additionally, we employed atomistic discrete molecular dynamics simulations to gain deeper insights into the interaction between Hsc70 variants and hIAPP. Integrating both experimental and computational findings, we propose a mechanism by which Hsc70's interaction with hIAPP monomers disrupts protein-protein connections, primarily by shielding the β-sheet edges of the Hsc70-β-sandwich. The distinctive conformational dynamics of the alpha helices of Hsc70 potentially enhance hIAPP binding by obstructing the exposed edges of the β-sandwich, particularly at the β5-β8 region along the alpha helix interface. This, in turn, inhibits fibril growth, and similar results were observed following hIAPP dimerization. Overall, this study elucidates the structural intricacies of Hsc70 crucial for impeding hIAPP aggregation, improving our understanding of the potential anti-aggregative properties of molecular chaperones in diabetes treatment.
Collapse
Affiliation(s)
- Ali Chaari
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar.
| | - Nabanita Saikia
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Pradipta Paul
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Mohammad Yousef
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Moncef Ladjimi
- Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, P.O. Box 24144, Doha, Qatar
| |
Collapse
|
3
|
Wirth F, Heitz FD, Seeger C, Combaluzier I, Breu K, Denroche HC, Thevenet J, Osto M, Arosio P, Kerr-Conte J, Verchere CB, Pattou F, Lutz TA, Donath MY, Hock C, Nitsch RM, Grimm J. A human antibody against pathologic IAPP aggregates protects beta cells in type 2 diabetes models. Nat Commun 2023; 14:6294. [PMID: 37813862 PMCID: PMC10562398 DOI: 10.1038/s41467-023-41986-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023] Open
Abstract
In patients with type 2 diabetes, pancreatic beta cells progressively degenerate and gradually lose their ability to produce insulin and regulate blood glucose. Beta cell dysfunction and loss is associated with an accumulation of aggregated forms of islet amyloid polypeptide (IAPP) consisting of soluble prefibrillar IAPP oligomers as well as insoluble IAPP fibrils in pancreatic islets. Here, we describe a human monoclonal antibody selectively targeting IAPP oligomers and neutralizing IAPP aggregate toxicity by preventing membrane disruption and apoptosis in vitro. Antibody treatment in male rats and mice transgenic for human IAPP, and human islet-engrafted mouse models of type 2 diabetes triggers clearance of IAPP oligomers resulting in beta cell protection and improved glucose control. These results provide new evidence for the pathological role of IAPP oligomers and suggest that antibody-mediated removal of IAPP oligomers could be a pharmaceutical strategy to support beta cell function in type 2 diabetes.
Collapse
Affiliation(s)
- Fabian Wirth
- Neurimmune AG, Wagistrasse 18, 8952, Schlieren, Switzerland
| | | | | | | | - Karin Breu
- Neurimmune AG, Wagistrasse 18, 8952, Schlieren, Switzerland
| | - Heather C Denroche
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Departments of Surgery and Pathology & Laboratory Medicine, University of British Columbia, A4-151 950 W 28 Ave, Vancouver, BC, Canada
| | - Julien Thevenet
- Univ-Lille, Inserm, CHU Lille, U1190 - EGID, F-59000, Lille, France
| | - Melania Osto
- Institute of Veterinary Physiology, Vetsuisse Faculty of the University of Zürich, Winterthurerstrasse 260, 8057, Zürich, Switzerland
| | - Paolo Arosio
- Institute for Chemical and Bioengineering, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093, Zürich, Switzerland
| | - Julie Kerr-Conte
- Univ-Lille, Inserm, CHU Lille, U1190 - EGID, F-59000, Lille, France
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Departments of Surgery and Pathology & Laboratory Medicine, University of British Columbia, A4-151 950 W 28 Ave, Vancouver, BC, Canada
| | - François Pattou
- Univ-Lille, Inserm, CHU Lille, U1190 - EGID, F-59000, Lille, France
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty of the University of Zürich, Winterthurerstrasse 260, 8057, Zürich, Switzerland
| | - Marc Y Donath
- Clinic for Endocrinology, Diabetes & Metabolism, and Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Christoph Hock
- Neurimmune AG, Wagistrasse 18, 8952, Schlieren, Switzerland
- Institute for Regenerative Medicine-IREM, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Roger M Nitsch
- Neurimmune AG, Wagistrasse 18, 8952, Schlieren, Switzerland
- Institute for Regenerative Medicine-IREM, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland
| | - Jan Grimm
- Neurimmune AG, Wagistrasse 18, 8952, Schlieren, Switzerland.
| |
Collapse
|
4
|
Smith AA, Moore KBE, Ambs PM, Saraswati AP, Fortin JS. Recent Advances in the Discovery of Therapeutics to Curtail Islet Amyloid Polypeptide Aggregation for Type 2 Diabetes Treatment. Adv Biol (Weinh) 2022; 6:e2101301. [PMID: 35931462 DOI: 10.1002/adbi.202101301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 07/04/2022] [Indexed: 01/28/2023]
Abstract
In humans with type 2 diabetes, at least 70% of patients exhibit islet amyloid plaques formed by misfolding islet amyloid polypeptides (IAPP). The oligomeric conformation and accumulation of the IAPP plaques lead to a panoply of cytotoxic effects on the islet β-cells. Currently, no marketed therapies for the prevention or elimination of these amyloid deposits exist, and therefore significant efforts are required to address this gap. To date, most of the experimental treatments are limited to only in vitro stages of testing. In general, the proposed therapeutics use various targeting strategies, such as binding to the N-terminal region of islet amyloid polypeptide on residues 1-19 or the hydrophobic region of IAPP. Other strategies include targeting the peptide self-assembly through π-stacking. These methods are realized by using several different families of compounds, four of which are highlighted in this review: naturally occurring products, small molecules, organometallic compounds, and nanoparticles. Each of these categories holds immense potential to optimize and develop inhibitor(s) of pancreatic amyloidosis in the near future.
Collapse
Affiliation(s)
- Alyssa A Smith
- Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Kendall B E Moore
- Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | | | - Akella Prasanth Saraswati
- Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Jessica S Fortin
- Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
5
|
Velander P, Wu L, Hildreth SB, Vogelaar NJ, Mukhopadhyay B, Helm RF, Zhang S, Xu B. Catechol-containing compounds are a broad class of protein aggregation inhibitors: Redox state is a key determinant of the inhibitory activities. Pharmacol Res 2022; 184:106409. [PMID: 35995346 PMCID: PMC10074477 DOI: 10.1016/j.phrs.2022.106409] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/20/2022] [Accepted: 08/17/2022] [Indexed: 12/30/2022]
Abstract
A range of neurodegenerative and related aging diseases, such as Alzheimer's disease and type 2 diabetes, are linked to toxic protein aggregation. Yet the mechanisms of protein aggregation inhibition by small molecule inhibitors remain poorly understood, in part because most protein targets of aggregation assembly are partially unfolded or intrinsically disordered, which hinders detailed structural characterization of protein-inhibitor complexes and structural-based inhibitor design. Herein we employed a parallel small molecule library-screening approach to identify inhibitors against three prototype amyloidogenic proteins in neurodegeneration and related proteinopathies: amylin, Aβ and tau. One remarkable class of inhibitors identified from these screens against different amyloidogenic proteins was catechol-containing compounds and redox-related quinones/anthraquinones. Secondary assays validated most of the identified inhibitors. In vivo efficacy evaluation of a selected catechol-containing compound, rosmarinic acid, demonstrated its strong mitigating effects of amylin amyloid deposition and related diabetic pathology in transgenic HIP rats. Further systematic investigation of selected class of inhibitors under aerobic and anaerobic conditions revealed that the redox state of the broad class of catechol-containing compounds is a key determinant of the amyloid inhibitor activities. The molecular insights we gained not only explain why a large number of catechol-containing polyphenolic natural compounds, often enriched in healthy diet, have anti-neurodegeneration and anti-aging activities, but also could guide the rational design of therapeutic or nutraceutical strategies to target a broad range of neurodegenerative and related aging diseases.
Collapse
Affiliation(s)
- Paul Velander
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Ling Wu
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; BRITE Research Institute and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA; Affiliated Faculty, Duke/UNC Alzheimer's Disease Research Center, Durham, NC 27710, USA
| | - Sherry B Hildreth
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Nancy J Vogelaar
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; Center for Drug Discovery, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Biswarup Mukhopadhyay
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Richard F Helm
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Bin Xu
- Department of Biochemistry, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; Center for Drug Discovery, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; BRITE Research Institute and Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA; Affiliated Faculty, Duke/UNC Alzheimer's Disease Research Center, Durham, NC 27710, USA.
| |
Collapse
|
6
|
Linking hIAPP misfolding and aggregation with type 2 diabetes mellitus: a structural perspective. Biosci Rep 2022; 42:231205. [PMID: 35475576 PMCID: PMC9118370 DOI: 10.1042/bsr20211297] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
There are over 40 identified human disorders that involve certain proteins folding incorrectly, accumulating in the body causing damage to cells and organs and causing disease. Type 2 Diabetes Mellitus (T2DM) is one of these protein misfolding disorders (PMDs) and involves human islet amyloid polypeptide (hIAPP) misfolding and accumulating in parts of the body, primarily in the pancreas, causing damage to islet cells and affecting glucose regulation. In this review, we have summarised our current understanding of what causes hIAPP to misfold, what conformations are found in different parts of the body with a particular focus on what is known about the structure of hIAPP and how this links to T2DM. Understanding the molecular basis behind these misfolding events is essential for understanding the role of hIAPP to develop better therapeutics since type 2 diabetes currently affects over 4.9 million people in the United Kingdom alone and is predicted to increase as our population ages.
Collapse
|
7
|
Sevcuka A, White K, Terry C. Factors That Contribute to hIAPP Amyloidosis in Type 2 Diabetes Mellitus. Life (Basel) 2022; 12:life12040583. [PMID: 35455074 PMCID: PMC9025880 DOI: 10.3390/life12040583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022] Open
Abstract
Cases of Type 2 Diabetes Mellitus (T2DM) are increasing at an alarming rate due to the rise in obesity, sedentary lifestyles, glucose-rich diets and other factors. Numerous studies have increasingly illustrated the pivotal role that human islet amyloid polypeptide (hIAPP) plays in the pathology of T2DM through damage and subsequent loss of pancreatic β-cell mass. HIAPP can misfold and form amyloid fibrils which are preceded by pre-fibrillar oligomers and monomers, all of which have been linked, to a certain extent, to β-cell cytotoxicity through a range of proposed mechanisms. This review provides an up-to-date summary of recent progress in the field, highlighting factors that contribute to hIAPP misfolding and aggregation such as hIAPP protein concentration, cell stress, molecular chaperones, the immune system response and cross-seeding with other amyloidogenic proteins. Understanding the structure of hIAPP and how these factors affect amyloid formation will help us better understand how hIAPP misfolds and aggregates and, importantly, help identify potential therapeutic targets for inhibiting amyloidosis so alternate and more effective treatments for T2DM can be developed.
Collapse
|
8
|
Min J, Ma F, Seyran B, Pellegrini M, Greeff O, Moncada S, Tudzarova S. β-cell-specific deletion of PFKFB3 restores cell fitness competition and physiological replication under diabetogenic stress. Commun Biol 2022; 5:248. [PMID: 35318430 PMCID: PMC8941137 DOI: 10.1038/s42003-022-03209-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/02/2022] [Indexed: 11/13/2022] Open
Abstract
HIF1α and PFKFB3 play a critical role in the survival of damaged β-cells in type–2 diabetes while rendering β-cells non-responsive to glucose stimulation. To discriminate the role of PFKFB3 from HIF1α in vivo, we generated mice with conditional β-cell specific disruption of the Pfkfb3 gene on a human islet pancreatic polypeptide (hIAPP+/−) background and a high-fat diet (HFD) [PFKFB3βKO + diabetogenic stress (DS)]. PFKFB3 disruption in β-cells under DS led to selective purging of hIAPP-damaged β-cells and the disappearance of insulin- and glucagon positive bihormonal cells. PFKFB3 disruption induced a three-fold increase in β-cell replication as evidenced by minichromosome maintenance 2 protein (MCM2) expression. Unlike high-, lower DS or switch to restricted chow diet abolished HIF1α levels and reversed glucose intolerance of PFKFB3βKO DS mice. Our data suggest that replication and functional recovery of β-cells under DS depend on β-cell competitive and selective purification of HIF1α and PFKFB3-positive β-cells. β-cell specific deletion of PFKFB3 results in removal of bihormonal cells and increase in β-cell replication, suggesting that this could lead to β-cell replenishment in type–2 diabetes.
Collapse
Affiliation(s)
- Jie Min
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.,Department of Endocrinology, Union Hospital of Tongji Medical College Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feiyang Ma
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Berfin Seyran
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Oppel Greeff
- Department of Pharmacology, University of Pretoria, Pretoria, South Africa
| | | | - Slavica Tudzarova
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Lutz TA. Creating the amylin story. Appetite 2022; 172:105965. [DOI: 10.1016/j.appet.2022.105965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023]
|
10
|
Rodriguez Camargo DC, Chia S, Menzies J, Mannini B, Meisl G, Lundqvist M, Pohl C, Bernfur K, Lattanzi V, Habchi J, Cohen SI, Knowles TPJ, Vendruscolo M, Linse S. Surface-Catalyzed Secondary Nucleation Dominates the Generation of Toxic IAPP Aggregates. Front Mol Biosci 2021; 8:757425. [PMID: 34790701 PMCID: PMC8591229 DOI: 10.3389/fmolb.2021.757425] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/12/2021] [Indexed: 01/22/2023] Open
Abstract
The aggregation of the human islet amyloid polypeptide (IAPP) is associated with diabetes type II. A quantitative understanding of this connection at the molecular level requires that the aggregation mechanism of IAPP is resolved in terms of the underlying microscopic steps. Here we have systematically studied recombinant IAPP, with amidated C-terminus in oxidised form with a disulphide bond between residues 3 and 7, using thioflavin T fluorescence to monitor the formation of amyloid fibrils as a function of time and IAPP concentration. We used global kinetic analyses to connect the macroscopic measurements of aggregation to the microscopic mechanisms, and show that the generation of new aggregates is dominated by the secondary nucleation of monomers on the fibril surface. We then exposed insulinoma cells to aliquots extracted from different time points of the aggregation process, finding the highest toxicity at the midpoint of the reaction, when the secondary nucleation rate reaches its maximum. These results identify IAPP oligomers as the most cytotoxic species generated during IAPP aggregation, and suggest that compounds that target secondary nucleation of IAPP could be most effective as therapeutic candidates for diabetes type II.
Collapse
Affiliation(s)
- Diana C Rodriguez Camargo
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden.,Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom
| | - Sean Chia
- Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom.,Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Menzies
- Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom
| | - Benedetta Mannini
- Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom.,Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Martin Lundqvist
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Christin Pohl
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Katja Bernfur
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Veronica Lattanzi
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Johnny Habchi
- Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom.,Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Samuel Ia Cohen
- Wren Therapeutics Limited, Clarendon House, Cambridge, United Kingdom
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.,Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Verma N, Srodulski S, Velmurugan S, Hoskins A, Pandey VK, Despa F, Despa S. Gestational diabetes triggers postpartum cardiac hypertrophy via activation of calcineurin/NFAT signaling. Sci Rep 2021; 11:20926. [PMID: 34686739 PMCID: PMC8536766 DOI: 10.1038/s41598-021-00422-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/12/2021] [Indexed: 01/11/2023] Open
Abstract
Population-based studies identified an association between a prior pregnancy complicated by gestational diabetes mellitus (GDM) and cardiac hypertrophy and dysfunction later in life. It is however unclear whether GDM initiates this phenotype and what are the underlying mechanisms. We addressed these questions by using female rats that express human amylin (HIP rats) as a GDM model and their wild-type (WT) littermates as the normal pregnancy model. Pregnant and two months postpartum HIP females had increased left-ventricular mass and wall thickness compared to non-pregnant HIP females, which indicates the presence of concentric hypertrophy. These parameters were unchanged in WT females during both pregnancy and postpartum periods. Hypertrophic Ca2+-dependent calcineurin/NFAT signaling was stimulated two months after giving birth in HIP females but not in the WT. In contrast, the CaMKII/HDAC hypertrophy pathway was active immediately after giving birth and returned to the baseline by two months postpartum in both WT and HIP females. Myocytes from two months postpartum HIP females exhibited slower Ca2+ transient relaxation and higher diastolic Ca2+ levels, which may explain calcineurin activation. No such effects occurred in the WT. These results suggest that a GDM-complicated pregnancy accelerates the development of pathological cardiac remodeling likely through activation of calcineurin/NFAT signaling.
Collapse
Affiliation(s)
- Nirmal Verma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Sarah Srodulski
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Sathya Velmurugan
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Amanda Hoskins
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Vivek K Pandey
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA
| | - Sanda Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 S Limestone, CTW 465, Lexington, KY, 40536, USA.
| |
Collapse
|
12
|
Wu L, Velander P, Brown AM, Wang Y, Liu D, Bevan DR, Zhang S, Xu B. Rosmarinic Acid Potently Detoxifies Amylin Amyloid and Ameliorates Diabetic Pathology in a Transgenic Rat Model of Type 2 Diabetes. ACS Pharmacol Transl Sci 2021; 4:1322-1337. [PMID: 34423269 PMCID: PMC8369672 DOI: 10.1021/acsptsci.1c00028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Indexed: 11/30/2022]
Abstract
Protein aggregation is associated with a large number of human protein-misfolding diseases, yet FDA-approved drugs are currently not available. Amylin amyloid and plaque depositions in the pancreas are hallmark features of type 2 diabetes. Moreover, these amyloid deposits are implicated in the pathogenesis of diabetic complications such as neurodegeneration. We recently discovered that catechols and redox-related quinones/anthraquinones represent a broad class of protein aggregation inhibitors. Further screening of a targeted library of natural compounds in complementary medicine that were enriched with catechol-containing compounds identified rosmarinic acid (RA) as a potent inhibitor of amylin aggregation (estimated inhibitory concentration IC50 = 200-300 nM). Structure-function relationship analysis of RA showed the additive effects of the two catechol-containing components of the RA molecule. We further showed that RA does not reverse fibrillation back to monomeric amylin but rather lead to nontoxic, remodeled protein aggregates. RA has significant ex vivo efficacy in reducing human amylin oligomer levels in HIP rat sera as well as in sera from diabetic patients. In vivo efficacy studies of RA treatment with the diabetic HIP rat model demonstrated significant reduction in amyloid islet deposition and strong mitigation of diabetic pathology. Our work provides new in vitro molecular mechanisms and in vivo efficacy insights for a model nutraceutical agent against type 2 diabetes and other aging-related protein-misfolding diseases.
Collapse
Affiliation(s)
- Ling Wu
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
- Biomanufacturing
Research Institute & Technology Enterprise (BRITE) and Department
of Pharmaceutical Sciences, North Carolina
Central University, Durham, North Carolina 27707, United States
| | - Paul Velander
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
| | - Anne M. Brown
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
| | - Yao Wang
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
| | - Dongmin Liu
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
| | - David R. Bevan
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
| | - Shijun Zhang
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Bin Xu
- Department
of Biochemistry, Center for Drug Discovery, Department of Human Nutrition, Foods,
and Exercise, and School of Neuroscience, Virginia Polytechnic
Institute and State University, Blacksburg, Virginia 24061, United States
- Biomanufacturing
Research Institute & Technology Enterprise (BRITE) and Department
of Pharmaceutical Sciences, North Carolina
Central University, Durham, North Carolina 27707, United States
- Affiliated
Program Faculty, Duke Comprehensive Stroke
Center, Durham, North Carolina 27710, United States
| |
Collapse
|
13
|
Burgos JI, Vallier L, Rodríguez-Seguí SA. Monogenic Diabetes Modeling: In Vitro Pancreatic Differentiation From Human Pluripotent Stem Cells Gains Momentum. Front Endocrinol (Lausanne) 2021; 12:692596. [PMID: 34295307 PMCID: PMC8290520 DOI: 10.3389/fendo.2021.692596] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
The occurrence of diabetes mellitus is characterized by pancreatic β cell loss and chronic hyperglycemia. While Type 1 and Type 2 diabetes are the most common types, rarer forms involve mutations affecting a single gene. This characteristic has made monogenic diabetes an interesting disease group to model in vitro using human pluripotent stem cells (hPSCs). By altering the genotype of the original hPSCs or by deriving human induced pluripotent stem cells (hiPSCs) from patients with monogenic diabetes, changes in the outcome of the in vitro differentiation protocol can be analyzed in detail to infer the regulatory mechanisms affected by the disease-associated genes. This approach has been so far applied to a diversity of genes/diseases and uncovered new mechanisms. The focus of the present review is to discuss the latest findings obtained by modeling monogenic diabetes using hPSC-derived pancreatic cells generated in vitro. We will specifically focus on the interpretation of these studies, the advantages and limitations of the models used, and the future perspectives for improvement.
Collapse
Affiliation(s)
- Juan Ignacio Burgos
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Ludovic Vallier
- Wellcome-Medical Research Council Cambridge Stem Cell Institute and Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Santiago A. Rodríguez-Seguí
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET-Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| |
Collapse
|
14
|
Wang Z, Gurlo T, Matveyenko AV, Elashoff D, Wang P, Rosenberger M, Junge JA, Stevens RC, White KL, Fraser SE, Butler PC. Live-cell imaging of glucose-induced metabolic coupling of β and α cell metabolism in health and type 2 diabetes. Commun Biol 2021; 4:594. [PMID: 34012065 PMCID: PMC8134470 DOI: 10.1038/s42003-021-02113-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 04/15/2021] [Indexed: 12/18/2022] Open
Abstract
AbstractType 2 diabetes is characterized by β and α cell dysfunction. We used phasor-FLIM (Fluorescence Lifetime Imaging Microscopy) to monitor oxidative phosphorylation and glycolysis in living islet cells before and after glucose stimulation. In healthy cells, glucose enhanced oxidative phosphorylation in β cells and suppressed oxidative phosphorylation in α cells. In Type 2 diabetes, glucose increased glycolysis in β cells, and only partially suppressed oxidative phosphorylation in α cells. FLIM uncovers key perturbations in glucose induced metabolism in living islet cells and provides a sensitive tool for drug discovery in diabetes.
Collapse
|
15
|
Liu M, Li N, Qu C, Gao Y, Wu L, Hu LG. Amylin deposition activates HIF1α and 6-phosphofructo-2-kinase/fructose-2, 6-biphosphatase 3 (PFKFB3) signaling in failing hearts of non-human primates. Commun Biol 2021; 4:188. [PMID: 33580152 PMCID: PMC7881154 DOI: 10.1038/s42003-021-01676-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023] Open
Abstract
Hyperamylinemia induces amylin aggregation and toxicity in the pancreas and contributes to the development of type-2 diabetes (T2D). Cardiac amylin deposition in patients with obesity and T2D was found to accelerate heart dysfunction. Non-human primates (NHPs) have similar genetic, metabolic, and cardiovascular processes as humans. However, the underlying mechanisms of cardiac amylin in NHPs, particularly related to the hypoxia inducible factor (HIF)1α and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) signaling pathways, are unknown. Here, we demonstrate that in NHPs, amylin deposition in heart failure (HF) contributes to cardiac dysfunction via activation of HIF1α and PFKFB3 signaling. This was confirmed in two in vitro cardiomyocyte models. Furthermore, alterations of intracellular Ca2+, reactive oxygen species, mitochondrial function, and lactate levels were observed in amylin-treated cells. Our study demonstrates a pathological role for amylin in the activation of HIF1α and PFKFB3 signaling in NHPs with HF, establishing amylin as a promising target for heart disease patients.
Collapse
Affiliation(s)
- Miao Liu
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China
| | - Nan Li
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China
| | - Chun Qu
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China
| | - Yilin Gao
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China
| | - Lijie Wu
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China
| | - Liangbiao George Hu
- Department of Translational Safety and Bioanalytical Sciences, Amgen R&D (Shanghai) Co. Ltd., Shanghai, China.
| |
Collapse
|
16
|
Campbell JE, Newgard CB. Mechanisms controlling pancreatic islet cell function in insulin secretion. Nat Rev Mol Cell Biol 2021; 22:142-158. [PMID: 33398164 PMCID: PMC8115730 DOI: 10.1038/s41580-020-00317-7] [Citation(s) in RCA: 348] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 02/07/2023]
Abstract
Metabolic homeostasis in mammals is tightly regulated by the complementary actions of insulin and glucagon. The secretion of these hormones from pancreatic β-cells and α-cells, respectively, is controlled by metabolic, endocrine, and paracrine regulatory mechanisms and is essential for the control of blood levels of glucose. The deregulation of these mechanisms leads to various pathologies, most notably type 2 diabetes, which is driven by the combined lesions of impaired insulin action and a loss of the normal insulin secretion response to glucose. Glucose stimulates insulin secretion from β-cells in a bi-modal fashion, and new insights about the underlying mechanisms, particularly relating to the second or amplifying phase of this secretory response, have been recently gained. Other recent work highlights the importance of α-cell-produced proglucagon-derived peptides, incretin hormones from the gastrointestinal tract and other dietary components, including certain amino acids and fatty acids, in priming and potentiation of the β-cell glucose response. These advances provide a new perspective for the understanding of the β-cell failure that triggers type 2 diabetes.
Collapse
Affiliation(s)
- Jonathan E Campbell
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA.
- Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, NC, USA.
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
17
|
Štambuk T, Gornik O. Protein Glycosylation in Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:285-305. [PMID: 34495541 DOI: 10.1007/978-3-030-70115-4_14] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is a group of metabolic disorders characterized by the presence of hyperglycaemia. Due to its high prevalence and substantial heterogeneity, many studies have been investigating markers that could identify predisposition for the disease development, differentiate between the various subtypes, establish early diagnosis, predict complications or represent novel therapeutic targets. N-glycans, complex oligosaccharide molecules covalently linked to proteins, emerged as potential markers and functional effectors of various diabetes subtypes, appearing to have the capacity to meet these requirements. For instance, it has been shown that N-glycome changes in patients with type 2 diabetes and that N-glycans can even identify individuals with an increased risk for its development. Moreover, genome-wide association studies identified glycosyltransferase genes as candidate causal genes for both type 1 and type 2 diabetes. N-glycans have also been suggested to have a major role in preventing the impairment of glucose-stimulated insulin secretion by modulating cell surface expression of glucose transporters. In this chapter we aimed to describe four major diabetes subtypes: type 1, type 2, gestational and monogenic diabetes, giving an overview of suggested role for N-glycosylation in their development, diagnosis and management.
Collapse
Affiliation(s)
- Tamara Štambuk
- Genos, Glycoscience Research Laboratory, Zagreb, Croatia.
| | - Olga Gornik
- University of Zagreb Faculty of Pharmacy and Biochemistry, Zagreb, Croatia
| |
Collapse
|
18
|
N-glycans as functional effectors of genetic and epigenetic disease risk. Mol Aspects Med 2020; 79:100891. [PMID: 32861467 DOI: 10.1016/j.mam.2020.100891] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/19/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022]
Abstract
N-glycosylation is a frequent modification of proteins, essential for all domains of life. N-glycan biosynthesis is a dynamic, complex, non-templated process, wherein specific glycoforms are modulated by various microenvironmental cues, cellular signals and local availability of dedicated enzymes and sugar precursors. This intricate regulatory network comprises hundreds of proteins, whose activity is dependent on both sequence of implicated genes and the regulation of their expression. In this regard, variation in N-glycosylation patterns stems from either gene polymorphisms or from stable epigenetic regulation of gene expression in different individuals. Moreover, epigenome alters in response to various environmental factors, representing a direct link between environmental exposure and changes in gene expression, that are subsequently reflected through altered N-glycosylation. N-glycosylation itself has a fundamental role in numerous biological processes, ranging from protein folding, cellular homeostasis, adhesion and immune regulation, to the effector functions in multiple diseases. Moreover, specific modification of the glycan structure can modulate glycoprotein's biological function or direct the faith of the entire cell, as seen on the examples of antibodies and T cells, respectively. Since immunoglobulin G is one of the most profoundly studied glycoproteins in general, the focus of this review will be on its N-glycosylation changes and their functional implications. By deepening the knowledge on the mechanistic roles that certain glycoforms exert in differential pathological processes, valuable insight into molecular perturbations occurring during disease development could be obtained. The prospect of resolving the exact biological pathways involved offers a potential for the development of new therapeutic interventions and molecular tools that would aid in prognosis, early referral and timely treatment of multiple disease conditions.
Collapse
|
19
|
Ueberberg S, Nauck MA, Uhl W, Montemurro C, Tannapfel A, Clark A, Meier JJ. Islet Amyloid in Patients With Diabetes Due to Exocrine Pancreatic Disorders, Type 2 Diabetes, and Nondiabetic Patients. J Clin Endocrinol Metab 2020; 105:5818378. [PMID: 32271378 DOI: 10.1210/clinem/dgaa176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/08/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Amyloid deposits are a typical finding in pancreatic islets from patients with type 2 diabetes. Whether this is linked to the pathogenesis of type 2 diabetes is currently unknown. Therefore, we compared the occurrence of islet amyloid in patients with type 2 diabetes, diabetes secondary to pancreatic disorders, and nondiabetic individuals. PATIENTS AND METHODS Pancreatic tissue from 15 nondiabetic patients, 22 patients with type 2 diabetes, and 11 patients with diabetes due to exocrine pancreatic disorders (chronic pancreatitis, pancreatic carcinoma) were stained for insulin, amyloid, and apoptosis. β-cell area, amyloid deposits, and β-cell apoptosis were quantified by morphometric analysis. RESULTS The proportion of islets containing amyloid deposits was significantly higher in both type 2 diabetes and diabetes due to exocrine pancreatic disorders than in healthy subjects. Islets with both amyloid and apoptosis were observed more frequently in type 2 diabetes and significantly more so in diabetes due to exocrine pancreatic disorders. In both diabetic groups, apoptotic ß-cells were found significantly more frequently in islets with more prominent amyloid deposits. CONCLUSIONS The occurrence of amyloid deposits in both type 2 diabetes and diabetes secondary to exocrine pancreatic disorders suggests that islet amyloid formation is a common feature of diabetes mellitus of different etiologies and may be associated with a loss of pancreatic ß-cells.
Collapse
Affiliation(s)
- Sandra Ueberberg
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michael A Nauck
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Waldemar Uhl
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| | - Chiara Montemurro
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
- David Geffen School of Medicine, University of California, Los Angeles, CA, US
| | | | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Juris J Meier
- Diabetes Division St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
20
|
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
|
21
|
Lai D, Huang M, Zhao L, Tian Y, Li Y, Liu D, Wu Y, Deng F. Delphinidin-induced autophagy protects pancreatic β cells against apoptosis resulting from high-glucose stress via AMPK signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1242-1249. [PMID: 31781740 DOI: 10.1093/abbs/gmz126] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 09/30/2019] [Accepted: 07/26/2019] [Indexed: 01/11/2023] Open
Abstract
Hyperglycemia, a diagnostic characteristic of diabetes mellitus, is detrimental to pancreatic β cells. Delphinidin, a member of the anthocyanin family, inhibits glucose absorption, increases glucagon-like peptide-1 (GLP-1) secretion, and improves insulin secretion in diabetes. However, whether delphinidin plays a protective role in pancreatic β-cell mass and function is not clear. In this study, delphinidin was found to decrease the high-glucose-induced apoptosis of RIN-m5F pancreatic β cells. In addition, delphinidin induced autophagy in RIN-m5F cells under the normal and high-glucose conditions, while 3-methyladenine (3-MA) inhibition of autophagy significantly diminished the protective role of delphinidin against high-glucose-induced apoptosis of pancreatic β cells. Delphinidin also decreased the level of cleaved caspase 3 and increased the phosphorylation level of AMP-activated protein kinase α (AMPKα) Thr172. Compound C, an AMPK inhibitor, was found to decrease the ratio of LC3-II/LC3-I, and the apoptotic rate of high-glucose-injured cells was increased after treatment with delphinidin, indicating that delphinidin attenuated the negative effects of high-glucose stress to cells. In conclusion, our data demonstrate that delphinidin protects pancreatic β cells against high-glucose-induced injury by autophagy regulation via the AMPK signaling pathway. These findings might shed light on the underlying mechanisms of diabetes and help improve the prevention and therapy of this common disease.
Collapse
Affiliation(s)
- Dengni Lai
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mingyong Huang
- Tianxiang Biotechnology Co., Ltd of Hunan, Shaoyang 422000, China
| | - Lingyan Zhao
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yan Tian
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yong Li
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Dongpo Liu
- State Key Laboratory of Subhealth Intervention Technology, Changsha 410128, China
| | - Yanyang Wu
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Fangming Deng
- Key Laboratory for Food Science and Biotechnology of Hunan Province, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
22
|
Vilchis-Flores LH, Barajas-Medina GA, Villa-Martínez AK, Salazar López SS, Luna-Patiño GA, Quiroz-Hernández ME, Guzmán-Vanegas MA, Rodríguez-Cortes R, Angulo-Romero F, Reyes-Escogido MDL, Aguilar-García A, Jiménez-Ceja LM, Folli F, Guardado-Mendoza R. Pancreatic β-cell dysfunction in normoglycemic patients and risk factors. Acta Diabetol 2019; 56:1305-1314. [PMID: 31468201 DOI: 10.1007/s00592-019-01411-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/21/2019] [Indexed: 01/21/2023]
Abstract
AIMS To evaluate pancreatic β-cell function (βf) in patients with normoglycemia (NG) and normal glucose tolerance (NGT) and related risk factors. METHODS An observational and comparative study in 527 patients with NG and NGT that were divided by quartiles of βf according to the disposition index derived from OGTT. Anthropometrical, clinical, nutritional, and biochemical variables were measured and associated with βf. RESULTS Quartiles of βf were Q1 = DI < 1.93 n = 131, Q2 = DI 1.93-2.45 n = 134, Q3 = DI 2.46-3.1 n = 133, and Q4 = DI > 3.1 n = 129. There was a progressive reduction in pancreatic β-cell function and it is negatively correlated with age, weight, BMI, total body fat and visceral fat, waist circumference, total cholesterol, LDL, and triglycerides (p < 0.01). Glucose levels during OGTT had a negative correlation with βf; the product of fasting glucose by 1-h glucose had the best correlation with βf (r = 0.611, p < 0.001) and was the best predictor of βdf (AUC 0.816, CI 95% 0.774-0.857), even better than 1-h glucose (r = 0.581, p < 0.001). Energy, fat, and carbohydrate intake were negatively correlated with βf (p < 0.05). Glucose levels at 1-h OGTT > 110 mg/dl were positively associated with pancreatic βdf (OR 6.85, CI 95% 3.86-12.4). In the multivariate analysis, glucose levels during OGTT, fasting insulin, and BMI were the main factors associated with βf. CONCLUSIONS A subgroup of patients with NG and NGT may have a loss of 40% of their βf. Factors related to this βdf were age, adiposity, glucose during OGTT, and the product of fasting and 1-h glucose, as well as food intake.
Collapse
Affiliation(s)
- Luis H Vilchis-Flores
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Georgina A Barajas-Medina
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Ana Karen Villa-Martínez
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Sara S Salazar López
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Gabriela A Luna-Patiño
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - María Elena Quiroz-Hernández
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | | | - Rafael Rodríguez-Cortes
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Fabiola Angulo-Romero
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Ma De Lourdes Reyes-Escogido
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | | | - Lilia M Jiménez-Ceja
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Guanajuato, Mexico
| | - Franco Folli
- Dipartimento di Scienze della Salute, Universita' degli Studi di Milano, Milan, Italy
| | - Rodolfo Guardado-Mendoza
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato / Hospital Regional de Alta Especialidad del Bajío, Col. San Carlos la Roncha, 37660, León, Guanajuato, Mexico.
| |
Collapse
|
23
|
Nie T, Zhang S, Vazhoor Amarsingh G, Liu H, McCann MJ, Cooper GJS. Altered metabolic gene expression in the brain of a triprolyl-human amylin transgenic mouse model of type 2 diabetes. Sci Rep 2019; 9:14588. [PMID: 31601900 PMCID: PMC6787337 DOI: 10.1038/s41598-019-51088-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a major health concern worldwide; however, the molecular mechanism underlying its development is poorly understood. The hormone amylin is postulated to be involved, as human amylin forms amyloid in the pancreases of diabetic patients, and oligomers have been shown to be cytotoxic to β-cells. As rodent amylin is non-amyloidogenic, mice expressing human amylin have been developed to investigate this hypothesis. However, it is not possible to differentiate the effects of amylin overexpression from β-cell loss in these models. We have developed transgenic mice that overexpress [25, 28, 29 triprolyl]human amylin, a non-amyloidogenic variant of amylin, designated the Line 44 model. This model allows us to investigate the effects of chronic overexpression of non-cytotoxic amylin. We characterised this model and found it developed obesity, hyperglycaemia and hyperinsulinaemia. This phenotype was associated with alterations in the expression of genes involved in the amylin, insulin and leptin signalling pathways within the brain. This included genes such as c-Fos (a marker of amylin activation); Socs3 (a leptin inhibitor); and Cart, Pomc and Npy (neuropeptides that control appetite). We also examined Socs3 protein expression and phosphorylated Stat3 to determine if changes at the mRNA level would be reflected at the protein level.
Collapse
Affiliation(s)
- Tina Nie
- School of Biological Sciences, Faculty of Science, the University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Shaoping Zhang
- School of Biological Sciences, Faculty of Science, the University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,The Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, the University of Auckland, Auckland, New Zealand
| | - Greeshma Vazhoor Amarsingh
- School of Biological Sciences, Faculty of Science, the University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Hong Liu
- School of Biological Sciences, Faculty of Science, the University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mark J McCann
- Food Nutrition & Health Team, AgResearch Ltd, Grasslands Research Centre, Palmerston North, 4442, New Zealand
| | - Garth J S Cooper
- School of Biological Sciences, Faculty of Science, the University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand. .,The Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, the University of Auckland, Auckland, New Zealand. .,Centre for Advanced Discovery and Experimental Therapeutics, Division of Cardiovascular Sciences, Faculty of Biology Medicine & Health, School of Medical Sciences, the University of Manchester, Manchester, M13 9NT, United Kingdom.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW To discuss the current understanding of "β cell identity" and factors underlying altered identity of pancreatic β cells in diabetes, especially in humans. RECENT FINDINGS Altered identity of β cells due to dedifferentiation and/or transdifferentiation has been proposed as a mechanism of loss of β cells in diabetes. In dedifferentiation, β cells do not undergo apoptosis; rather, they lose their identity and function. Dedifferentiation is well characterized by the decrease in expression of key β cell markers such as genes encoding major transcription factors, e.g., MafA, NeuroD1, Nkx6.1, and Foxo1, and an increase in atypical or "disallowed" genes for β cells such as lactate dehydrogenase, monocarboxylate transporter MCT1, or progenitor cell genes (Neurog3, Pax4, or Sox9). Moreover, altered identity of mature β cells in diabetes also involves transdifferentiation of β cells into other islet hormone producing cells. For example, overexpression of α cell specific transcription factor Arx or ablation of Pdx1 resulted in an increase of α cell numbers and a decrease in β cell numbers in rodents. The frequency of α-β double-positive cells was also prominent in human subjects with T2D. These altered identities of β cells likely serve as a compensatory response to enhance function/expand cell numbers and may also camouflage/protect cells from ongoing stress. However, it is equally likely that this may be a reflection of new cell formation as a frank regenerative response to ongoing tissue injury. Physiologically, all these responses are complementary. In diabetes, (1) endocrine identity recapitulates the less mature/less-differentiated fetal/neonatal cell type, possibly representing an adaptive mechanism; (2) residual β cells may be altered in their subtype proportions or other molecular features; (3) in humans, "altered identity" is a preferable term to dedifferentiation as their cellular fate (differentiated cells losing identity or progenitors becoming more differentiated) is unclear as yet.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, PO Box 34110 Doha, Qatar
| | - Alexandra E. Butler
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, PO Box 34110 Doha, Qatar
| |
Collapse
|
25
|
Popescu I, Yin G, Velmurugan S, Erickson JR, Despa F, Despa S. Lower sarcoplasmic reticulum Ca 2+ threshold for triggering afterdepolarizations in diabetic rat hearts. Heart Rhythm 2019; 16:765-772. [PMID: 30414461 PMCID: PMC6491240 DOI: 10.1016/j.hrthm.2018.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Type 2 diabetes (T2D) increases arrhythmia risk through incompletely elucidated mechanisms. Ventricular arrhythmias could be initiated by delayed afterdepolarizations (DADs) resulting from elevated spontaneous sarcoplasmic reticulum (SR) Ca2+ release (SR Ca2+ leak). OBJECTIVE The purpose of this study was to test the role of DADs and SR Ca2+ leak in triggering arrhythmias in T2D hearts. METHODS We compared rats with late-onset T2D that display pancreatic and cardiac phenotypes similar to those in humans with T2D (HIP rats) and their nondiabetic littermates (wild type [WT]). RESULTS HIP rats showed higher propensity for premature ventricular complexes and ventricular tachyarrhythmias, whereas HIP myocytes displayed more frequent DADs and had lower SR Ca2+ content than WT. However, the threshold SR Ca2+ at which depolarizing transient inward currents (Itis) are generated was also significantly decreased in HIP myocytes and was below the actual SR Ca2+ load, which explains the increased DAD incidence despite reduced Ca2+ in SR. In agreement with these findings, Ca2+ spark frequency was augmented in myocytes from HIP vs WT rats, which suggests activation of ryanodine receptors (RyRs) in HIP hearts. Indeed, RyR phosphorylation (by CaMKII and protein kinase A) and oxidation are enhanced in HIP hearts, whereas there is no RyR O-GlcNAcylation in either HIP or control hearts. CaMKII inhibition dissipated the difference in Ca2+ spark frequency between HIP and WT myocytes. CONCLUSION The threshold SR Ca2+ for generating depolarizing Itis is lower in T2D because of RyR activation after hyperphosphorylation and oxidation, which favors the occurrence of DADs despite low SR Ca2+ loads.
Collapse
Affiliation(s)
- Iuliana Popescu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Guo Yin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Sathya Velmurugan
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Jeffrey R Erickson
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Sanda Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
26
|
Butler AE, Kirakossian D, Gurlo T, Gao F, Coppola G, Butler PC. In the setting of β-cell stress, the pancreatic duct gland transcriptome shows characteristics of an activated regenerative response. Am J Physiol Gastrointest Liver Physiol 2018; 315:G848-G854. [PMID: 30095296 PMCID: PMC6293255 DOI: 10.1152/ajpgi.00177.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The pancreatic duct gland (PDG) compartment has been proposed as a potential stem cell niche based on its coiled tubular structure embedded in mesenchyme, its proliferation and expansion in response to pancreatic injury, and the fact that it contains endocrine and exocrine epithelial cells. Little is known of the molecular signature of the PDG compartment in either a quiescent state or the potentially activated state during β-cell stress characteristic of diabetes. To address this, we performed RNA sequencing on RNA obtained from PDGs of wild-type vs. prediabetic HIP rats, a model of type 2 diabetes. The transcriptome of the PDG compartment, compared with a library of 84 tissue types, placed PDGs midpoint between the exocrine and endocrine pancreas and closely related to seminiferous tubules, consistent with a role as a stem cell niche for the exocrine and endocrine pancreas. Standard differential expression analysis (permissive threshold P < 0.005) identified 245 genes differentially expressed in PDGs from HIP rats vs. WT rats, with overrepresentation of transcripts involved in acute inflammatory responses, regulation of cell proliferation, and tissue development, while pathway analysis pointed to enrichment of cell movement-related pathways. In conclusion, the transcriptome of the PDG compartment is consistent with a pancreatic stem cell niche that is activated by ongoing β-cell stress signals. The documented PDG transcriptome provides potential candidates to be exploited for lineage tracing studies of this as yet little investigated compartment. NEW & NOTEWORTHY The pancreatic duct gland (PDG) compartment has been proposed as a potential stem cell niche. Transcriptome analysis of the PDG gland placed it midpoint between exocrine and endocrine tissues with adaptation toward response to inflammation and increased cell movement in a model of type 2 diabetes with ongoing β-cell apoptosis. These findings support the proposal that PDGs may act as a pancreatic stem cell niche.
Collapse
Affiliation(s)
- Alexandra E. Butler
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - David Kirakossian
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tatyana Gurlo
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Fuying Gao
- 2Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Giovanni Coppola
- 2Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Peter C. Butler
- 1Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
27
|
Wu X, Li Z, Chen K, Yin P, Zheng L, Sun S, Chen X. Egr-1 transactivates WNT5A gene expression to inhibit glucose-induced β-cell proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2018; 1861:S1874-9399(18)30218-9. [PMID: 30025875 DOI: 10.1016/j.bbagrm.2018.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023]
Abstract
Selective β-cell loss is a characteristic of type 2 diabetes mellitus (T2DM). Inhibition of glucose-stimulated β-cell proliferation is one of the in vivo results of the lipotoxicity of saturated fatty acids (SFAs). However, the mechanism by which lipotoxicity inhibits β-cell proliferation is still unclear. In this study, we found palmitate, a saturated fatty acid, inhibited the β-cell proliferation induced by high glucose through the induction of Wnt5a expression in vitro and in vivo. We also found that Wnt5a was both sufficient and necessary for inhibition of β-cell proliferation. Additionally, Egr-1, but not NF-κB, FOXO1, Smad2, Smad3, SP1 or SP3 mediated the expression of Wnt5a. Deletion and site-directed mutagenesis of the WNT5A promoter revealed that activation of WNT5A gene transcription depends primarily on a putative Egr-binding sequence between nucleotides -52 to -44, upstream of the transcription start site. Furthermore, Egr-1 bound directly to this sequence in response to palmitate treatment, both in vitro and in vivo. Moreover, after mice islets were treated with Egr inhibitors, the expression of Wnt5a decreased significantly and the glucose-induced β-cell proliferation inhibited by palmitate was resumed. These findings establish Wnt5a as an Egr-1 target gene in β-cells, uncovering a novel Egr-1/Wnt5a pathway by which saturated free fatty acids block glucose-induced β-cell proliferation. Our study lends support for the potential of Egr-1 inhibitors or Wnt5a antibodies as therapeutics for the treatment of T2DM.
Collapse
Affiliation(s)
- XingEr Wu
- The Molecular Diagnostic Center, Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China; Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - ZeHong Li
- Guzhen Sub-bureau, Zhongshan Public Security Bureau, Zhongshan 528400, Guangdong, China
| | - Kang Chen
- Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China
| | - PeiHong Yin
- Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China.
| | - ShiJun Sun
- The Molecular Diagnostic Center, Zhongshan City People's Hospital, Zhongshan 528403, Guangdong, China.
| | - XiaoYu Chen
- The Eighth Affiliated Hospital of Sun Yat-Sen University, Futian, 518000 Shenzhen, China.
| |
Collapse
|
28
|
Abedini A, Derk J, Schmidt AM. The receptor for advanced glycation endproducts is a mediator of toxicity by IAPP and other proteotoxic aggregates: Establishing and exploiting common ground for novel amyloidosis therapies. Protein Sci 2018; 27:1166-1180. [PMID: 29664151 PMCID: PMC6032365 DOI: 10.1002/pro.3425] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/23/2022]
Abstract
Proteotoxicity plays a key role in many devastating human disorders, including Alzheimer's, Huntington's and Parkinson's diseases; type 2 diabetes; systemic amyloidosis; and cardiac dysfunction, to name a few. The cellular mechanisms of proteotoxicity in these disorders have been the focus of considerable research, but their role in prevalent and morbid disorders, such as diabetes, is less appreciated. There is a large body of literature on the impact of glucotoxicity and lipotoxicity on insulin-producing pancreatic β-cells, and there is increasing recognition that proteotoxicty plays a key role. Pancreatic islet amyloidosis by the hormone IAPP, the production of advanced glycation endproducts (AGE), and insulin misprocessing into cytotoxic aggregates are all sources of β-cell proteotoxicity in diabetes. AGE, produced by the reaction of reducing sugars with proteins and lipids are ligands for the receptor for AGE (RAGE), as are the toxic pre-fibrillar aggregates of IAPP produced during amyloid formation. The mechanisms of amyloid formation by IAPP in vivo or in vitro are not well understood, and the cellular mechanisms of IAPP-induced β-cell death are not fully defined. Here, we review recent findings that illuminate the factors and mechanisms involved in β-cell proteotoxicity in diabetes. Together, these new insights have far-reaching implications for the establishment of unifying mechanisms by which pathological amyloidoses imbue their injurious effects in vivo.
Collapse
Affiliation(s)
- Andisheh Abedini
- Diabetes Research Program, Division of Endocrinology, Department of MedicineNew York University Medical Center, 550 First Avenue, Smilow 906New YorkNew York10016
| | - Julia Derk
- Diabetes Research Program, Division of Endocrinology, Department of MedicineNew York University Medical Center, 550 First Avenue, Smilow 906New YorkNew York10016
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of MedicineNew York University Medical Center, 550 First Avenue, Smilow 906New YorkNew York10016
| |
Collapse
|
29
|
Marasco MR, Linnemann AK. β-Cell Autophagy in Diabetes Pathogenesis. Endocrinology 2018; 159:2127-2141. [PMID: 29617763 PMCID: PMC5913620 DOI: 10.1210/en.2017-03273] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/27/2018] [Indexed: 12/25/2022]
Abstract
Nearly 100 years have passed since Frederick Banting and Charles Best first discovered and purified insulin. Their discovery and subsequent improvements revolutionized the treatment of diabetes, and the field continues to move at an ever-faster pace with respect to unique treatments for both type 1 and type 2 diabetes. Despite these advances, we still do not fully understand how apoptosis of the insulin-producing β-cells is triggered, presenting a challenge in the development of preventative measures. In recent years, the process of autophagy has generated substantial interest in this realm due to discoveries highlighting its clear role in the maintenance of cellular homeostasis. As a result, the number of studies focused on islet and β-cell autophagy has increased substantially in recent years. In this review, we will discuss what is currently known regarding the role of β-cell autophagy in type 1 and type 2 diabetes pathogenesis, with an emphasis on new and exciting developments over the past 5 years. Further, we will discuss how these discoveries might be translated into unique treatments in the coming years.
Collapse
Affiliation(s)
- Michelle R Marasco
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Amelia K Linnemann
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
30
|
Sasahara K. Membrane-mediated amyloid deposition of human islet amyloid polypeptide. Biophys Rev 2018; 10:453-462. [PMID: 29204886 PMCID: PMC5899711 DOI: 10.1007/s12551-017-0351-x] [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: 09/28/2017] [Accepted: 11/14/2017] [Indexed: 01/01/2023] Open
Abstract
Amyloid deposition of human islet amyloid polypeptide (hIAPP) within the islet of Langerhans is closely associated with type II diabetes mellitus. Accumulating evidence indicates that the membrane-mediated aggregation and subsequent deposition of hIAPP are linked to the dysfunction and death of insulin-producing pancreatic β-cells, but the molecular process of hIAPP deposition is poorly understood. In this review, I focus on recent in vitro studies utilizing model membranes to observe the membrane-mediated aggregation/deposition of hIAPP. Membrane surfaces can serve as templates for both hIAPP adsorption and aggregation. Using high-sensitivity surface analyzing/imaging techniques that can characterize the processes of hIAPP aggregation and deposition at the membrane surface, these studies provide valuable insights into the mechanism of membrane damage caused by amyloid deposition of the peptide.
Collapse
Affiliation(s)
- Kenji Sasahara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
31
|
Abedini A, Cao P, Plesner A, Zhang J, He M, Derk J, Patil SA, Rosario R, Lonier J, Song F, Koh H, Li H, Raleigh DP, Schmidt AM. RAGE binds preamyloid IAPP intermediates and mediates pancreatic β cell proteotoxicity. J Clin Invest 2018; 128:682-698. [PMID: 29337308 PMCID: PMC5785261 DOI: 10.1172/jci85210] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/17/2017] [Indexed: 01/04/2023] Open
Abstract
Islet amyloidosis is characterized by the aberrant accumulation of islet amyloid polypeptide (IAPP) in pancreatic islets, resulting in β cell toxicity, which exacerbates type 2 diabetes and islet transplant failure. It is not fully clear how IAPP induces cellular stress or how IAPP-induced toxicity can be prevented or treated. We recently defined the properties of toxic IAPP species. Here, we have identified a receptor-mediated mechanism of islet amyloidosis-induced proteotoxicity. In human diabetic pancreas and in cellular and mouse models of islet amyloidosis, increased expression of the receptor for advanced glycation endproducts (RAGE) correlated with human IAPP-induced (h-IAPP-induced) β cell and islet inflammation, toxicity, and apoptosis. RAGE selectively bound toxic intermediates, but not nontoxic forms of h-IAPP, including amyloid fibrils. The isolated extracellular ligand-binding domains of soluble RAGE (sRAGE) blocked both h-IAPP toxicity and amyloid formation. Inhibition of the interaction between h-IAPP and RAGE by sRAGE, RAGE-blocking antibodies, or genetic RAGE deletion protected pancreatic islets, β cells, and smooth muscle cells from h-IAPP-induced inflammation and metabolic dysfunction. sRAGE-treated h-IAPP Tg mice were protected from amyloid deposition, loss of β cell area, β cell inflammation, stress, apoptosis, and glucose intolerance. These findings establish RAGE as a mediator of IAPP-induced toxicity and suggest that targeting the IAPP/RAGE axis is a potential strategy to mitigate this source of β cell dysfunction in metabolic disease.
Collapse
Affiliation(s)
- Andisheh Abedini
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Ping Cao
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | | | - Jinghua Zhang
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Meilun He
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Julia Derk
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Sachi A. Patil
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Rosa Rosario
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Jacqueline Lonier
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Fei Song
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| | - Hyunwook Koh
- Division of Biostatistics, Department of Population Health, NYU School of Medicine, New York, New York, USA
| | - Huilin Li
- Division of Biostatistics, Department of Population Health, NYU School of Medicine, New York, New York, USA
| | - Daniel P. Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, NYU School of Medicine, New York, New York, USA
| |
Collapse
|
32
|
Jurczak MJ, Saini S, Ioja S, Costa DK, Udeh N, Zhao X, Whaley JM, Kibbey RG. SGLT2 knockout prevents hyperglycemia and is associated with reduced pancreatic β-cell death in genetically obese mice. Islets 2018; 10:181-189. [PMID: 30118626 PMCID: PMC6284495 DOI: 10.1080/19382014.2018.1503027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Inhibition of the sodium-glucose co-transporter type 2 (SGLT2) has received growing acceptance as a novel, safe and effective means to improve glycemic control in patients with type 2 diabetes. Inhibition of SGLT2 lowers the renal glucose threshold and reduces plasma glucose by promoting glucose excretion in urine. Both animal studies and clinical trials in man suggest that SGLT2 inhibition has the potential to improve pancreatic β-cell function by reducing glucose toxicity. However, there is limited data exploring how reducing glucotoxicity via SGLT2 inhibition affects rates of β-cell proliferation and death throughout life in the context of insulin resistance and type 2 diabetes. SGLT2-/- mice were backcrossed to the db/db strain to produce littermate control db/db-SGLT2+/+ and experimental db/db-SGLT2-/- mice. Mice were euthanized at 5, 12 and 20 weeks of age to collect plasma for glucose, insulin, lipid and cytokine measures, and pancreata for histological analysis including determination of β-cell mass and rates of proliferation and death. SGLT2 deletion in db/db mice reduced plasma glucose as early as 5 weeks of age and continued throughout life without changes in plasma lipids or cytokines. Reduced plasma glucose levels occurred in parallel with an increase in the relative β-cell volume and reduced frequency of β-cell death, and no apparent change in rates of β-cell proliferation. These data add to a growing body of evidence demonstrating that improved glycemic control achieved through SGLT2 inhibition can preserve β-cell function and endogenous insulin secretion by reducing glucose toxicity and rates of β-cell death.
Collapse
Affiliation(s)
- Michael J. Jurczak
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Saumya Saini
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Simona Ioja
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Diana K. Costa
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Nnamdi Udeh
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaojian Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jean M. Whaley
- Metabolic Diseases Biology, Bristol-Myers Squibb Research and Development, Princeton, NJ, USA
| | - Richard G. Kibbey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Departments of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- CONTACT Richard G. Kibbey Internal Medicine-Endocrinology, Yale University School of Medicine, TAC S269, P.O. Box 9812, New Haven, CT 06536-8012; Michael J. Jurczak Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1060, Pittsburgh, PA 15261
| |
Collapse
|
33
|
Amylin and diabetic cardiomyopathy - amylin-induced sarcolemmal Ca 2+ leak is independent of diabetic remodeling of myocardium. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1923-1930. [PMID: 29066284 DOI: 10.1016/j.bbadis.2017.10.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/06/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023]
Abstract
Amylin is a pancreatic β-cell hormone co-secreted with insulin, plays a role in normal glucose homeostasis, and forms amyloid in the pancreatic islets of individuals with type-2 diabetes. Aggregated amylin is also found in blood and extra-pancreatic tissues, including myocardium. Myocardial amylin accumulation is associated with myocyte Ca2+ dysregulation in diabetic rats expressing human amylin. Whether deposition of amylin in the heart is a consequence of or a contributor to diabetic cardiomyopathy remains unknown. We used amylin knockout (AKO) mice intravenously infused with either human amylin (i.e, the aggregated form) or non-amyloidogenic (i.e., monomeric) rodent amylin to test the hypothesis that aggregated amylin accumulates in the heart in the absence of diabetes. AKO mice infused with human amylin, but not rodent amylin, showed amylin deposits in the myocardium. Cardiac amylin level was larger in males compared to females. Sarcolemmal Ca2+ leak and Ca2+ transients were increased in myocytes isolated from males infused with human amylin while no significant changes occurred in either females injected with human amylin or in rat amylin-infused mice. In isolated cardiac myocytes, the amylin receptor antagonist AC-187 did not effectively block the interaction of amylin with the sarcolemma. In conclusion, circulating aggregated amylin accumulates preferentially in male vs. female hearts and its effects on myocyte Ca2+ cycling do not require diabetic remodeling of the myocardium. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
|
34
|
Protective effects of a G. lucidum proteoglycan on INS-1 cells against IAPP-induced apoptosis via attenuating endoplasmic reticulum stress and modulating CHOP/JNK pathways. Int J Biol Macromol 2017; 106:893-900. [PMID: 28893685 DOI: 10.1016/j.ijbiomac.2017.08.089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 08/03/2017] [Accepted: 08/14/2017] [Indexed: 01/09/2023]
Abstract
Fudan-Yueyang-G. lucidum (FYGL) is a water-soluble macromolecular proteoglycan extracted from Ganoderma lucidum which has been used for health promotion for a long time in China. The aim of this study was to investigate the protective effects of FYGL on INS-1 rat insulinoma beta cells against IAPP-induced cell apoptosis, as well as the underlying mechanisms. The results showed that apoptotic cells were significantly increased when incubated with islet amyloid polypeptide (IAPP). However, cytotoxicity of IAPP was significantly attenuated by co-incubation of the cells with FYGL. The results of RT-PCR showed that mRNA expression of caspase-3, caspase-12 and C/EBP homologous protein (CHOP) in IAPP-treated cells were inhibited by FYGL. Moreover, FYGL significantly prevented the IAPP-induced abnormal expression of inositol-requiring protein-1α (IRE1α), protein kinase RNA (PKR)-like ER kinase (PERK), activating transcription factor 6 (ATF6), as well as suppressed the activation of CHOP and c-Jun N-terminal kinase (JNK). Taken together, our results suggest that FYGL protects INS-1 pancreatic beta cells against IAPP-induced apoptosis through attenuating endoplasmic reticulum stress (ERS) and modulating CHOP/JNK pathways.
Collapse
|
35
|
Qian J, Thomas AP, Schroeder AM, Rakshit K, Colwell CS, Matveyenko AV. Development of diabetes does not alter behavioral and molecular circadian rhythms in a transgenic rat model of type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 2017; 313:E213-E221. [PMID: 28465284 PMCID: PMC5582890 DOI: 10.1152/ajpendo.00406.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 01/09/2023]
Abstract
Metabolic state and circadian clock function exhibit a complex bidirectional relationship. Circadian disruption increases propensity for metabolic dysfunction, whereas common metabolic disorders such as obesity and type 2 diabetes (T2DM) are associated with impaired circadian rhythms. Specifically, alterations in glucose availability and glucose metabolism have been shown to modulate clock gene expression and function in vitro; however, to date, it is unknown whether development of diabetes imparts deleterious effects on the suprachiasmatic nucleus (SCN) circadian clock and SCN-driven outputs in vivo. To address this question, we undertook studies in aged diabetic rats transgenic for human islet amyloid polypeptide, an established nonobese model of T2DM (HIP rat), which develops metabolic defects closely recapitulating those present in patients with T2DM. HIP rats were also cross-bred with a clock gene reporter rat model (Per1:luciferase transgenic rat) to permit assessment of the SCN and the peripheral molecular clock function ex vivo. Utilizing these animal models, we examined effects of diabetes on 1) behavioral circadian rhythms, 2) photic entrainment of circadian activity, 3) SCN and peripheral tissue molecular clock function, and 4) melatonin secretion. We report that circadian activity, light-induced entrainment, molecular clockwork, as well as melatonin secretion are preserved in the HIP rat model of T2DM. These results suggest that despite the well-characterized ability of glucose to modulate circadian clock gene expression acutely in vitro, SCN clock function and key behavioral and physiological outputs appear to be preserved under chronic diabetic conditions characteristic of nonobese T2DM.
Collapse
MESH Headings
- Animals
- Behavior, Animal/physiology
- Circadian Rhythm/genetics
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Disease Models, Animal
- Disease Progression
- Islet Amyloid Polypeptide/genetics
- Islet Amyloid Polypeptide/metabolism
- Light
- Male
- Period Circadian Proteins/metabolism
- Rats
- Rats, Sprague-Dawley
- Rats, Transgenic
- Suprachiasmatic Nucleus/metabolism
- Suprachiasmatic Nucleus/pathology
Collapse
Affiliation(s)
- Jingyi Qian
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Anthony P Thomas
- Department of Medicine, University of California Los Angeles, Los Angeles, California; and
| | - Analyne M Schroeder
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Kuntol Rakshit
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Christopher S Colwell
- Departments of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, Minnesota;
| |
Collapse
|
36
|
Schludi B, Moin ASM, Montemurro C, Gurlo T, Matveyenko AV, Kirakossian D, Dawson DW, Dry SM, Butler PC, Butler AE. Islet inflammation and ductal proliferation may be linked to increased pancreatitis risk in type 2 diabetes. JCI Insight 2017; 2:92282. [PMID: 28679961 DOI: 10.1172/jci.insight.92282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/23/2017] [Indexed: 12/26/2022] Open
Abstract
Pancreatitis is more frequent in type 2 diabetes mellitus (T2DM), although the underlying cause is unknown. We tested the hypothesis that ongoing β cell stress and apoptosis in T2DM induces ductal tree proliferation, particularly the pancreatic duct gland (PDG) compartment, and thus potentially obstructs exocrine outflow, a well-established cause of pancreatitis. PDG replication was increased 2-fold in human pancreas from individuals with T2DM, and was associated with increased pancreatic intraepithelial neoplasia (PanIN), lesions associated with pancreatic inflammation and with the potential to obstruct pancreatic outflow. Increased PDG replication in the prediabetic human-IAPP-transgenic (HIP) rat model of T2DM was concordant with increased β cell stress but preceded metabolic derangement. Moreover, the most abundantly expressed chemokines released by the islets in response to β cell stress in T2DM, CXCL1, -4, and -10, induced proliferation in human pancreatic ductal epithelium. Also, the diabetes medications reported as potential modifiers for the risk of pancreatitis in T2DM modulated PDG proliferation accordingly. We conclude that chronic stimulation and proliferation of the PDG compartment in response to islet inflammation in T2DM is a potentially novel mechanism that serves as a link to the increased risk for pancreatitis in T2DM and may potentially be modified by currently available diabetes therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - David W Dawson
- Department of Pathology and Laboratory Medicine.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | - Sarah M Dry
- Department of Pathology and Laboratory Medicine.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | - Peter C Butler
- Larry L. Hillblom Islet Research Center.,Jonsson Comprehensive Cancer Center, UCLA, David Geffen School of Medicine, Los Angeles, California, USA
| | | |
Collapse
|
37
|
Courtade JA, Klimek-Abercrombie AM, Chen YC, Patel N, Lu PYT, Speake C, Orban PC, Najafian B, Meneilly G, Greenbaum CJ, Warnock GL, Panagiotopoulos C, Verchere CB. Measurement of Pro-Islet Amyloid Polypeptide (1-48) in Diabetes and Islet Transplants. J Clin Endocrinol Metab 2017; 102:2595-2603. [PMID: 28368485 DOI: 10.1210/jc.2016-2773] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 03/20/2017] [Indexed: 12/21/2022]
Abstract
CONTEXT Islet amyloid is a feature of β-cell failure in type 2 diabetes (T2D) and type 1 diabetes (T1D) recipients of islet transplants. Islet amyloid contains islet amyloid polypeptide (IAPP; amylin), a circulating peptide that is produced in β cells by processing of its precursor, proIAPP1-67, via an intermediate form, proIAPP1-48. Elevated proinsulin to C-peptide ratios in the plasma of persons with diabetes suggest defects in β-cell prohormone processing. OBJECTIVE Determine whether plasma levels of precursor forms of IAPP are elevated in diabetes. DESIGN, SETTING, AND PATIENTS We developed an immunoassay to detect proIAPP1-48 in human plasma, and we determined the ratio of proIAPP1-48 to mature IAPP in subjects with T1D, T2D, recipients of islet transplants, and healthy controls. RESULTS The proIAPP1-48 immunoassay had a limit of detection of 0.18 ± 0.06 pM and cross-reactivity with intact proIAPP1-67 <15%. Healthy individuals had plasma concentrations of proIAPP1-48 immunoreactivity of 1.5 ± 0.2 pM and a proIAPP1-48 to total IAPP ratio of 0.28 ± 0.03. Plasma concentrations of proIAPP1-48 immunoreactivity were not significantly different in subjects with T2D but were markedly increased in T1D recipients of islet transplants. Children and adults with T1D had reduced mature IAPP levels relative to age-matched controls but an elevated ratio of proIAPP1-48 to total IAPP. CONCLUSION The β cells in T1D and islet transplants have impaired processing of the proIAPP1-48 intermediate. The ratio of proIAPP1-48-to-IAPP immunoreactivity may have value as a biomarker of β-cell stress and dysfunction.
Collapse
Affiliation(s)
- Jaques A Courtade
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Agnieszka M Klimek-Abercrombie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Yi-Chun Chen
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Nirja Patel
- American Laboratory Products Company, Salem, New Hampshire 03079
| | - Phoebe Y T Lu
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Genetics, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Cate Speake
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington 98101
| | - Paul C Orban
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
| | - Behzad Najafian
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Graydon Meneilly
- Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Carla J Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute, Seattle, Washington 98101
| | - Garth L Warnock
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - Constadina Panagiotopoulos
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | - C Bruce Verchere
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
- Research Institute, BC Children's Hospital, Vancouver, British Columbia V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| |
Collapse
|
38
|
Li H, Li Y, Xiang L, Zhang J, Zhu B, Xiang L, Dong J, Liu M, Xiang G. GDF11 Attenuates Development of Type 2 Diabetes via Improvement of Islet β-Cell Function and Survival. Diabetes 2017; 66:1914-1927. [PMID: 28450417 DOI: 10.2337/db17-0086] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022]
Abstract
Growth differentiation factor 11 (GDF11) has been implicated in the regulation of islet development and a variety of aging conditions, but little is known about the physiological functions of GDF11 in adult pancreatic islets. Here, we showed that systematic replenishment of GDF11 not only preserved insulin secretion but also improved the survival and morphology of β-cells and improved glucose metabolism in both nongenetic and genetic mouse models of type 2 diabetes (T2D). Conversely, anti-GDF11 monoclonal antibody treatment caused β-cell failure and lethal T2D. In vitro treatment of isolated murine islets and MIN6 cells with recombinant GDF11 attenuated glucotoxicity-induced β-cell dysfunction and apoptosis. Mechanistically, the GDF11-mediated protective effects could be attributed to the activation of transforming growth factor-β/Smad2 and phosphatidylinositol-4,5-bisphosphate 3-kinase-AKT-FoxO1 signaling. These findings suggest that GDF11 repletion may improve β-cell function and mass and thus may lead to a new therapeutic approach for T2D.
Collapse
Affiliation(s)
- Huan Li
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Yixiang Li
- Radiation-Diagnostic/Oncology School of Medicine, Emory University, Atlanta, GA
| | - Lingwei Xiang
- Mathematics and Statistics Department, Georgia State University, Atlanta, GA
| | - JiaJia Zhang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Biao Zhu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| |
Collapse
|
39
|
Courtade JA, Wang EY, Yen P, Dai DL, Soukhatcheva G, Orban PC, Verchere CB. Loss of prohormone convertase 2 promotes beta cell dysfunction in a rodent transplant model expressing human pro-islet amyloid polypeptide. Diabetologia 2017; 60:453-463. [PMID: 27999871 DOI: 10.1007/s00125-016-4174-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/03/2016] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS A contributor to beta cell failure in type 2 diabetes and islet transplants is amyloid formation by aggregation of the beta cell peptide, islet amyloid polypeptide (IAPP). Similar to the proinsulin processing pathway that generates insulin, IAPP is derived from a prohormone precursor, proIAPP, which requires cleavage by prohormone convertase (PC) 1/3 and PC2 in rodent pancreatic beta cells. We hypothesised that loss of PC2 would promote beta cell death and dysfunction in a rodent model of human beta cell proIAPP overexpression. METHODS We generated an islet transplant model wherein immune-deficient mouse models of diabetes received islets expressing amyloidogenic human proIAPP and lacking PC2, leading to restoration of normoglycaemia accompanied by increased secretion of human proIAPP. Blood glucose levels were analysed for up to 16 weeks in transplant recipients and grafts were assessed for islet amyloid and beta cell number and death. RESULTS Hyperglycaemia (blood glucose >16.9 mmol/l) returned in 94% of recipients of islets expressing human proIAPP and lacking PC2, whereas recipients of islets that express human proIAPP and normal PC2 levels remained normoglycaemic for at least 16 weeks. Islet graft failure was accompanied by a ∼20% reduction in insulin-positive cells, yet the degree of amyloid deposition and beta cell apoptosis was similar to those of controls expressing human proIAPP with functional PC2 levels. CONCLUSIONS/INTERPRETATION PC2 deficiency in transplanted mouse islets expressing human proIAPP promotes beta cell loss and graft failure. Our data suggest that impaired NH2-terminal processing and increased secretion of human proIAPP promote beta cell failure.
Collapse
Affiliation(s)
- Jaques A Courtade
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Evan Y Wang
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul Yen
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Derek L Dai
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Galina Soukhatcheva
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul C Orban
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - C Bruce Verchere
- Research Institute, BC Children's Hospital, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
40
|
Zhang X, St Clair JR, London E, Raleigh DP. Islet Amyloid Polypeptide Membrane Interactions: Effects of Membrane Composition. Biochemistry 2017; 56:376-390. [PMID: 28054763 DOI: 10.1021/acs.biochem.6b01016] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amyloid formation by islet amyloid polypeptide (IAPP) contributes to β-cell dysfunction in type 2 diabetes. Perturbation of the β-cell membrane may contribute to IAPP-induced toxicity. We examine the effects of lipid composition, salt, and buffer on IAPP amyloid formation and on the ability of IAPP to induce leakage of model membranes. Even low levels of anionic lipids promote amyloid formation and membrane permeabilization. Increasing the percentage of the anionic lipids, 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-l-serine (POPS) or 1,2-dioleoyl-sn-glycero-3-phospho(1'-rac-glycerol), enhances the rate of amyloid formation and increases the level of membrane permeabilization. The choice of zwitterionic lipid has no noticeable effect on membrane-catalyzed amyloid formation but in most cases affects leakage, which tends to decrease in the following order: 1,2-dioleoyl-sn-glycero-3-phosphocholine > 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine > sphingomyelin. Uncharged lipids that increase the level of membrane order weaken the ability of IAPP to induce leakage. Leakage is due predominately to pore formation rather than complete disruption of the vesicles under the conditions used in these studies. Cholesterol at or below physiological levels significantly reduces the rate of vesicle-catalyzed IAPP amyloid formation and decreases the susceptibility to IAPP-induced leakage. The effects of cholesterol on amyloid formation are masked by 25 mol % POPS. Overall, there is a strong inverse correlation between the time to form amyloid and the extent of vesicle leakage. NaCl reduces the rate of membrane-catalyzed amyloid formation by anionic vesicles, but accelerates amyloid formation in solution. The implications for IAPP membrane interactions are discussed, as is the possibility that the loss of phosphatidylserine asymmetry enhances IAPP amyloid formation and membrane damage in vivo via a positive feedback loop.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States
| | - Johnna R St Clair
- Department of Biochemistry and Cell Biology, Stony Brook University , Stony Brook, New York 11794-5215, United States
| | - Erwin London
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States.,Department of Biochemistry and Cell Biology, Stony Brook University , Stony Brook, New York 11794-5215, United States
| | - Daniel P Raleigh
- Department of Chemistry, Stony Brook University , Stony Brook, New York 11794-3400, United States.,Graduate Program in Biochemistry and Structural Biology, Stony Brook University , Stony Brook, New York 11794-5215, United States
| |
Collapse
|
41
|
Haupt-Jorgensen M, Buschard K, Hansen AK, Josefsen K, Antvorskov JC. Gluten-free diet increases beta-cell volume and improves glucose tolerance in an animal model of type 2 diabetes. Diabetes Metab Res Rev 2016; 32:675-684. [PMID: 26991675 DOI: 10.1002/dmrr.2802] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 02/10/2016] [Accepted: 02/22/2016] [Indexed: 11/05/2022]
Abstract
BACKGROUND Gluten-free (GF) diet alleviates type 1 diabetes in animal models and possibly in humans. We recently showed that fatty acid-induced insulin secretion is enhanced by enzymatically digested gluten (gliadin) stimulation in INS-1E insulinoma cells. We therefore hypothesized that GF diet would induce beta-cell rest and ameliorate type 2 diabetes. METHODS C57BL/6JBomTac (B6) mice were fed a high-fat (HF), gluten-free high-fat (GF-HF), standard (STD) or gluten-free (GF) diet for 42 weeks. RESULTS Short-term (6-24 weeks) GF-HF versus HF feeding impaired glucose tolerance and increased fasting glucose. Long-term (36-42 weeks) GF-HF versus HF feeding improved glucose tolerance and decreased fasting leptin. Mice fed a GF-HF versus HF diet for 42 weeks showed higher volumes of beta cells, islets and pancreas. The beta-cell volume correlated with the islet- and pancreas volume as well as body weight. GF-HF versus HF diet did not influence toll-like receptor 4 (Tlr4), interleukin 1 (IL-1), interleukin 6 (IL-6) or tumour necrosis factor-alpha (TNF-alpha) mRNA expression in intestine. STD versus GF feeding did not affect any parameter studied. CONCLUSIONS Long-term feeding with GF-HF versus HF increases beta-cell volume and improves glucose tolerance in B6 mice. The mechanism may include beta-cell rest, but is unlikely to include TLR4 and proinflammatory cytokines in the intestine. Beta-cell volume correlates with pancreas volume and body weight, indicating that insulin secretion capacity controls pancreas volume. Thus, long-term GF diets may be beneficial for obese type 2 diabetes patients and trials should be performed. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
| | | | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
42
|
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
|
43
|
Abstract
Type two diabetes (T2D) is a challenging metabolic disorder for which a cure has not yet been found. Its etiology is associated with several phenomena, including significant loss of insulin-producing, beta cell (β cell) mass via progressive programmed cell death and disrupted cellular autophagy. In diabetes, the etiology of β cell death and the role of mitochondria are complex and involve several layers of mechanisms. Understanding the dynamics of those mechanisms could permit researchers to develop an intervention for the progressive loss of β cells. Currently, diabetes research has shifted toward rejuvenation and plasticity technology and away from the simplified approach of hormonal compensation. Diabetes research is currently challenged by questions such as how to enhance cell survival, decrease apoptosis and replenish β cell mass in diabetic patients. In this review, we discuss evidence that β cell development and mass formation are guided by specific signaling systems, particularly hormones, transcription factors, and growth factors, all of which could be manipulated to enhance mass growth. There is also strong evidence that β cells are dynamically active cells, which, under specific conditions such as obesity, can increase in size and subsequently increase insulin secretion. In certain cases of aggressive or advanced forms of T2D, β cells become markedly impaired, and the only alternatives for maintaining glucose homeostasis are through partial or complete cell grafting (the Edmonton protocol). In these cases, the harvesting of an enriched population of viable β cells is required for transplantation. This task necessitates a deep understanding of the pharmacological agents that affect β cell survival, mass, and function. The aim of this review is to initiate discussion about the important signals in pancreatic β cell development and mass formation and to highlight the process by which cell death occurs in diabetes. This review also examines the attempts that have been made to recover or increase cell mass in diabetic patients by using various pharmacological agents.
Collapse
Affiliation(s)
- Husnia I Marrif
- Department of Pharmacology, Faculty of Medicine, University of Benghazi Benghazi, Libya
| | - Salma I Al-Sunousi
- Department of Histology and Anatomy, Faculty of Medicine, University of Benghazi Benghazi, Libya
| |
Collapse
|
44
|
Hydroxychloroquine hindering of diabetic isletopathy carries its signature on the inflammatory cytokines. J Mol Histol 2016; 47:183-93. [PMID: 26872459 DOI: 10.1007/s10735-016-9664-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/11/2016] [Indexed: 01/07/2023]
Abstract
Hydroxychloroquine (HCQ) is supposed to have favorable effects in diabetes mellitus (DM). However no previous experimental studies had investigated its effect on the structure of the endocrine pancreas, islets of Langerhans (IOL), in DM. In addition, the mechanism by which HCQ acts in DM is not well understood. In this study, we hypothesized that the possible favorable effects of HCQ in DM at the structural as well as at metabolic levels could be accomplished, in part, by its anti-inflammatory action. A total of 45 rats were divided equally into; control, DM and HCQ + DM groups (received citrate buffer, 27.5 mg/kg single ip STZ and STZ + HCQ 200 mg/kg/w respectively). After 4 weeks, samples from pancreas were histologically studied for the resulting changes. The HCQ + DM group showed preservation of IOL structure, a significant increase (p < 0.05) in the β-cell area, %, mass, IOL proliferation and neogenesis as well as correction of the significantly increased (p < 0.05) α-cell area, %, disturbed glucose homeostasis and lipid profile compared with the DM group. The significantly elevated inflammatory cytokines in the latter were lowered in the HCQ + DM group. Therefore, HCQ showed definite favorable effects on the histological as well as the metabolic profiles in DM which may be partly attributed to its anti-inflammatory action. This notable improvement of DM by HCQ deserves further studies to distinctly approve HCQ as a promising oral hypoglycemic agent.
Collapse
|
45
|
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
|
46
|
Jeong HR, An SSA. Causative factors for formation of toxic islet amyloid polypeptide oligomer in type 2 diabetes mellitus. Clin Interv Aging 2015; 10:1873-9. [PMID: 26604727 PMCID: PMC4655906 DOI: 10.2147/cia.s95297] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human islet amyloid polypeptide (h-IAPP) is a peptide hormone that is synthesized and cosecreted with insulin from insulin-secreting pancreatic β-cells. Recently, h-IAPP was proposed to be the main component responsible for the cytotoxic pancreatic amyloid deposits in patients with type 2 diabetes mellitus (T2DM). Since the causative factors of IAPP (or amylin) oligomer aggregation are not fully understood, this review will discuss the various forms of h-IAPP aggregation. Not all forms of IAPP aggregates trigger the destruction of β-cell function and loss of β-cell mass; however, toxic oligomers do trigger these events. Once these toxic oligomers form under abnormal metabolic conditions in T2DM, they can lead to cell disruption by inducing cell membrane destabilization. In this review, the various factors that have been shown to induce toxic IAPP oligomer formation will be presented, as well as the potential mechanism of oligomer and fibril formation from pro-IAPPs. Initially, pro-IAPPs undergo enzymatic reactions to produce the IAPP monomers, which can then develop into oligomers and fibrils. By this mechanism, toxic oligomers could be generated by diverse pathway components. Thus, the interconnections between factors that influence amyloid aggregation (eg, absence of PC2 enzyme, deamidation, reduction of disulfide bonds, environmental factors in the cell, genetic mutations, copper metal ions, and heparin) will be presented. Hence, this review will aid in understanding the fundamental causative factors contributing to IAPP oligomer formation and support studies for investigating novel T2DM therapeutic approaches, such as the development of inhibitory agents for preventing oligomerization at the early stages of diabetic pathology.
Collapse
Affiliation(s)
- Hye Rin Jeong
- Department of Bionano Technology, Gachon Medical Research Institute, Gachon University, Gyeonggi-do, Republic of Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon Medical Research Institute, Gachon University, Gyeonggi-do, Republic of Korea
| |
Collapse
|
47
|
Akter R, Cao P, Noor H, Ridgway Z, Tu LH, Wang H, Wong AG, Zhang X, Abedini A, Schmidt AM, Raleigh DP. Islet Amyloid Polypeptide: Structure, Function, and Pathophysiology. J Diabetes Res 2015; 2016:2798269. [PMID: 26649319 PMCID: PMC4662979 DOI: 10.1155/2016/2798269] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/24/2015] [Indexed: 01/29/2023] Open
Abstract
The hormone islet amyloid polypeptide (IAPP, or amylin) plays a role in glucose homeostasis but aggregates to form islet amyloid in type-2 diabetes. Islet amyloid formation contributes to β-cell dysfunction and death in the disease and to the failure of islet transplants. Recent work suggests a role for IAPP aggregation in cardiovascular complications of type-2 diabetes and hints at a possible role in type-1 diabetes. The mechanisms of IAPP amyloid formation in vivo or in vitro are not understood and the mechanisms of IAPP induced β-cell death are not fully defined. Activation of the inflammasome, defects in autophagy, ER stress, generation of reactive oxygen species, membrane disruption, and receptor mediated mechanisms have all been proposed to play a role. Open questions in the field include the relative importance of the various mechanisms of β-cell death, the relevance of reductionist biophysical studies to the situation in vivo, the molecular mechanism of amyloid formation in vitro and in vivo, the factors which trigger amyloid formation in type-2 diabetes, the potential role of IAPP in type-1 diabetes, the development of clinically relevant inhibitors of islet amyloidosis toxicity, and the design of soluble, bioactive variants of IAPP for use as adjuncts to insulin therapy.
Collapse
Affiliation(s)
- Rehana Akter
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Ping Cao
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Harris Noor
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Zachary Ridgway
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Ling-Hsien Tu
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Hui Wang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Amy G. Wong
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Xiaoxue Zhang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Andisheh Abedini
- Diabetes Research Program, NYU School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, NYU School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Daniel P. Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
- Research Department of Structural and Molecule Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
48
|
Islet Neogenesis Associated Protein (INGAP) induces the differentiation of an adult human pancreatic ductal cell line into insulin-expressing cells through stepwise activation of key transcription factors for embryonic beta cell development. Differentiation 2015; 90:77-90. [DOI: 10.1016/j.diff.2015.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/13/2015] [Accepted: 10/22/2015] [Indexed: 01/13/2023]
|
49
|
Plasma amylin concentration in suckling goat neonates and its relationship with C-reactive protein, selected biochemical and hormonal indicators. ACTA VET BRNO 2015. [DOI: 10.2754/avb201584030237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Amylin is a recently discovered neuropeptide hormone that belongs to the calcitonin gene-related peptide family. It is co-secreted with insulin in response to feed intake. In goat kids, neonatal mortality and morbidity seems to be relatively higher than in other farm species. This high mortality and morbidity in goat kids may be associated with underdeveloped metabolism and immune system during the first week of life. The main objectives of this study were to determine amylin concentration and its relationship with some hormones, biochemical indicators and with a general inflammatory marker, CRP (C-reactive protein) in goat neonates. Blood samples were collected from 30 Saanen goat neonates at 20–35 days of age. Plasma amylin and other hormone concentrations were measured by ELISA, whereas serum biochemical indices were analysed by spectrophotometry. The mean values of plasma amylin concentrations were 9.07 ± 0.25 pmol/l. Plasma amylin concentrations were positively correlated with plasma non-esterified fatty acids, CRP, prolactin, cortisol, insulin; however, a negative correlation was determined between plasma amylin and serum triglyceride concentrations. The current study suggests that amylin contents are strongly associated with circulating concentrations of some hormones and with those of CRP in Saanen goat kids.
Collapse
|
50
|
Lambert R, Srodulski S, Peng X, Margulies KB, Despa F, Despa S. Intracellular Na+ Concentration ([Na+]i) Is Elevated in Diabetic Hearts Due to Enhanced Na+-Glucose Cotransport. J Am Heart Assoc 2015; 4:e002183. [PMID: 26316524 PMCID: PMC4599504 DOI: 10.1161/jaha.115.002183] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Intracellular Na+ concentration ([Na+]i) regulates Ca2+ cycling, contractility, metabolism, and electrical stability of the heart. [Na+]i is elevated in heart failure, leading to arrhythmias and oxidative stress. We hypothesized that myocyte [Na+]i is also increased in type 2 diabetes (T2D) due to enhanced activity of the Na+–glucose cotransporter. Methods and Results To test this hypothesis, we used myocardial tissue from humans with T2D and a rat model of late-onset T2D (HIP rat). Western blot analysis showed increased Na+–glucose cotransporter expression in failing hearts from T2D patients compared with nondiabetic persons (by 73±13%) and in HIP rat hearts versus wild-type (WT) littermates (by 61±8%). [Na+]i was elevated in HIP rat myocytes both at rest (14.7±0.9 versus 11.4±0.7 mmol/L in WT) and during electrical stimulation (17.3±0.8 versus 15.0±0.7 mmol/L); however, the Na+/K+-pump function was similar in HIP and WT cells, suggesting that higher [Na+]i is due to enhanced Na+ entry in diabetic hearts. Indeed, Na+ influx was significantly larger in myocytes from HIP versus WT rats (1.77±0.11 versus 1.29±0.06 mmol/L per minute). Na+–glucose cotransporter inhibition with phlorizin or glucose-free solution greatly reduced Na+ influx in HIP myocytes (to 1.20±0.16 mmol/L per minute), whereas it had no effect in WT cells. Phlorizin also significantly decreased glucose uptake in HIP myocytes (by 33±9%) but not in WT, indicating an increased reliance on the Na+–glucose cotransporter for glucose uptake in T2D hearts. Conclusions Myocyte Na+–glucose cotransport is enhanced in T2D, which increases Na+ influx and causes Na+ overload. Higher [Na+]i may contribute to arrhythmogenesis and oxidative stress in diabetic hearts.
Collapse
Affiliation(s)
- Rebekah Lambert
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY (R.L., S.S., X.P., F.D., S.D.)
| | - Sarah Srodulski
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY (R.L., S.S., X.P., F.D., S.D.)
| | - Xiaoli Peng
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY (R.L., S.S., X.P., F.D., S.D.)
| | - Kenneth B Margulies
- Cardiovascular Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA (K.B.M.)
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY (R.L., S.S., X.P., F.D., S.D.)
| | - Sanda Despa
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY (R.L., S.S., X.P., F.D., S.D.)
| |
Collapse
|