1
|
Esser N, Hogan MF, Templin AT, Akter R, Fountaine BS, Castillo JJ, El-Osta A, Manathunga L, Zhyvoloup A, Raleigh DP, Zraika S, Hull RL, Kahn SE. The islet tissue plasminogen activator/plasmin system is upregulated with human islet amyloid polypeptide aggregation and protects beta cells from aggregation-induced toxicity. Diabetologia 2024; 67:1897-1911. [PMID: 39245780 PMCID: PMC11410534 DOI: 10.1007/s00125-024-06161-0] [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: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 09/10/2024]
Abstract
AIMS/HYPOTHESIS Apart from its fibrinolytic activity, the tissue plasminogen activator (tPA)/plasmin system has been reported to cleave the peptide amyloid beta, attenuating brain amyloid deposition in Alzheimer's disease. As aggregation of human islet amyloid polypeptide (hIAPP) is toxic to beta cells, we sought to determine whether activation of the fibrinolytic system can also reduce islet amyloid deposition and its cytotoxic effects, which are both observed in type 2 diabetes. METHODS The expression of Plat (encoding tPA) and plasmin activity were measured in isolated islets from amyloid-prone hIAPP transgenic mice or non-transgenic control islets expressing non-amyloidogenic mouse islet amyloid polypeptide cultured in the absence or presence of the amyloid inhibitor Congo Red. Plat expression was also determined in hIAPP-treated primary islet endothelial cells, bone marrow-derived macrophages (BMDM) and INS-1 cells, in order to determine the islet cell type(s) producing tPA in response to hIAPP aggregation. Cell-free thioflavin-T assays and MS were used to respectively monitor hIAPP aggregation kinetics and investigate plasmin cleavage of hIAPP. Cell viability was assessed in INS-1 beta cells treated with hIAPP with or without plasmin. Finally, to confirm the findings in human samples, PLAT expression was measured in freshly isolated islets from donors with and without type 2 diabetes. RESULTS In isolated islets from transgenic mice, islet Plat expression and plasmin activity increased significantly with the process of amyloid deposition (p≤0.01, n=5); these effects were not observed in islets from non-transgenic mice and were blocked by Congo Red (p≤0.01, n=4). In response to hIAPP exposure, Plat expression increased in BMDM and INS-1 cells vs vehicle-treated cells (p≤0.05, n=4), but not in islet endothelial cells. Plasmin reduced hIAPP fibril formation in a dose-dependent manner in a cell-free system, and restored hIAPP-induced loss of cell viability in INS-1 beta cells (p≤0.01, n=5). Plasmin cleaved monomeric hIAPP, inducing a rapid decrease in the abundance of full-length hIAPP and the appearance of hIAPP 1-11 and 12-37 fragments. hIAPP 12-37, which contains the critical amyloidogenic region, was not toxic to INS-1 cells. Finally, PLAT expression was significantly increased by 2.4-fold in islets from donors with type 2 diabetes (n=4) vs islets from donors without type 2 diabetes (n=7) (p≤0.05). CONCLUSIONS/INTERPRETATION The fibrinolytic system is upregulated in islets with hIAPP aggregation. Plasmin rapidly degrades hIAPP, limiting its aggregation into amyloid and thus protecting beta cells from hIAPP-induced toxicity. Thus, increasing islet plasmin activity might be a strategy to limit beta cell loss in type 2 diabetes.
Collapse
Affiliation(s)
- Nathalie Esser
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
- Laboratory of Immunometabolism and Nutrition, GIGA, University of Liège, CHU of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU of Liège, Liège, Belgium
| | - Meghan F Hogan
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew T Templin
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rehana Akter
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Joseph J Castillo
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Lakshan Manathunga
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
| | - Alexander Zhyvoloup
- Research Department of Structural and Molecular Biology, University College London, London, UK
| | - Daniel P Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA.
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA.
- Research Department of Structural and Molecular Biology, University College London, London, UK.
| | - Sakeneh Zraika
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rebecca L Hull
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Steven E Kahn
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Mukherjee N, Contreras CJ, Lin L, Colglazier KA, Mather EG, Kalwat MA, Esser N, Kahn SE, Templin AT. RIPK3 promotes islet amyloid-induced β-cell loss and glucose intolerance in a humanized mouse model of type 2 diabetes. Mol Metab 2024; 80:101877. [PMID: 38218538 PMCID: PMC10830894 DOI: 10.1016/j.molmet.2024.101877] [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: 11/06/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVE Aggregation of human islet amyloid polypeptide (hIAPP), a β-cell secretory product, leads to islet amyloid deposition, islet inflammation and β-cell loss in type 2 diabetes (T2D), but the mechanisms that underlie this process are incompletely understood. Receptor interacting protein kinase 3 (RIPK3) is a pro-death signaling molecule that has recently been implicated in amyloid-associated brain pathology and β-cell cytotoxicity. Here, we evaluated the role of RIPK3 in amyloid-induced β-cell loss using a humanized mouse model of T2D that expresses hIAPP and is prone to islet amyloid formation. METHODS We quantified amyloid deposition, cell death and caspase 3/7 activity in islets isolated from WT, Ripk3-/-, hIAPP and hIAPP; Ripk3-/- mice in real time, and evaluated hIAPP-stimulated inflammation in WT and Ripk3-/- bone marrow derived macrophages (BMDMs) in vitro. We also characterized the role of RIPK3 in glucose stimulated insulin secretion (GSIS) in vitro and in vivo. Finally, we examined the role of RIPK3 in high fat diet (HFD)-induced islet amyloid deposition, β-cell loss and glucose homeostasis in vivo. RESULTS We found that amyloid-prone hIAPP mouse islets exhibited increased cell death and caspase 3/7 activity compared to amyloid-free WT islets in vitro, and this was associated with increased RIPK3 expression. hIAPP; Ripk3-/- islets were protected from amyloid-induced cell death compared to hIAPP islets in vitro, although amyloid deposition and caspase 3/7 activity were not different between genotypes. We observed that macrophages are a source of Ripk3 expression in isolated islets, and that Ripk3-/- BMDMs were protected from hIAPP-stimulated inflammatory gene expression (Tnf, Il1b, Nos2). Following 52 weeks of HFD feeding, islet amyloid-prone hIAPP mice exhibited impaired glucose tolerance and decreased β-cell area compared to WT mice in vivo, whereas hIAPP; Ripk3-/- mice were protected from these impairments. CONCLUSIONS In conclusion, loss of RIPK3 protects from amyloid-induced inflammation and islet cell death in vitro and amyloid-induced β-cell loss and glucose intolerance in vivo. We propose that therapies targeting RIPK3 may reduce islet inflammation and β-cell loss and improve glucose homeostasis in the pathogenesis of T2D.
Collapse
Affiliation(s)
- Noyonika Mukherjee
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher J Contreras
- Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Lin
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Kaitlyn A Colglazier
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Egan G Mather
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Michael A Kalwat
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and the University of Washington, Seattle, WA, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and the University of Washington, Seattle, WA, USA
| | - Andrew T Templin
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, IN, USA; Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
3
|
Hsu C, Templin AT, Prosswimmer T, Shea D, Li J, Brooks‐Worrell B, Kahn SE, Daggett V. Human islet amyloid polypeptide-induced β-cell cytotoxicity is linked to formation of α-sheet structure. Protein Sci 2024; 33:e4854. [PMID: 38062941 PMCID: PMC10823758 DOI: 10.1002/pro.4854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 01/30/2024]
Abstract
Type 2 diabetes (T2D) results from insulin secretory dysfunction arising in part from the loss of pancreatic islet β-cells. Several factors contribute to β-cell loss, including islet amyloid formation, which is observed in over 90% of individuals with T2D. The amyloid is comprised of human islet amyloid polypeptide (hIAPP). Here we provide evidence that early in aggregation, hIAPP forms toxic oligomers prior to formation of amyloid fibrils. The toxic oligomers contain α-sheet secondary structure, a nonstandard secondary structure associated with toxic oligomers in other amyloid diseases. De novo, synthetic α-sheet compounds designed to be nontoxic and complementary to the α-sheet structure in the toxic oligomers inhibit hIAPP aggregation and neutralize oligomer-mediated cytotoxicity in cell-based assays. In vivo administration of an α-sheet design to mice for 4 weeks revealed no evidence of toxicity nor did it elicit an immune response. Furthermore, the α-sheet designs reduced endogenous islet amyloid formation and mitigation of amyloid-associated β-cell loss in cultured islets isolated from an hIAPP transgenic mouse model of islet amyloidosis. Characterization of the involvement of α-sheet in early aggregation of hIAPP and oligomer toxicity contributes to elucidation of the molecular mechanisms underlying amyloid-associated β-cell loss.
Collapse
Affiliation(s)
- Cheng‐Chieh Hsu
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
- Molecular Engineering ProgramUniversity of WashingtonSeattleWashingtonUSA
| | - Andrew T. Templin
- Division of Metabolism, Endocrinology and Nutrition, Department of MedicineVA Puget Sound Health Care System and University of WashingtonSeattleWashingtonUSA
| | - Tatum Prosswimmer
- Molecular Engineering ProgramUniversity of WashingtonSeattleWashingtonUSA
| | - Dylan Shea
- Molecular Engineering ProgramUniversity of WashingtonSeattleWashingtonUSA
| | - Jinzheng Li
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Barbara Brooks‐Worrell
- Division of Metabolism, Endocrinology and Nutrition, Department of MedicineVA Puget Sound Health Care System and University of WashingtonSeattleWashingtonUSA
| | - Steven E. Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of MedicineVA Puget Sound Health Care System and University of WashingtonSeattleWashingtonUSA
| | - Valerie Daggett
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
- Molecular Engineering ProgramUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
4
|
Zammit NW, Wong YY, Walters SN, Warren J, Barry SC, Grey ST. RELA governs a network of islet-specific metabolic genes necessary for beta cell function. Diabetologia 2023; 66:1516-1531. [PMID: 37311878 PMCID: PMC10317895 DOI: 10.1007/s00125-023-05931-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/14/2023] [Indexed: 06/15/2023]
Abstract
AIMS/HYPOTHESIS NF-κB activation unites metabolic and inflammatory responses in many diseases yet less is known about the role that NF-κB plays in normal metabolism. In this study we investigated how RELA impacts the beta cell transcriptional landscape and provides network control over glucoregulation. METHODS We generated novel mouse lines harbouring beta cell-specific deletion of either the Rela gene, encoding the canonical NF-κB transcription factor p65 (βp65KO mice), or the Ikbkg gene, encoding the NF-κB essential modulator NEMO (βNEMOKO mice), as well as βA20Tg mice that carry beta cell-specific and forced transgenic expression of the NF-κB-negative regulator gene Tnfaip3, which encodes the A20 protein. Mouse studies were complemented by bioinformatics analysis of human islet chromatin accessibility (assay for transposase-accessible chromatin with sequencing [ATAC-seq]), promoter capture Hi-C (pcHi-C) and p65 binding (chromatin immunoprecipitation-sequencing [ChIP-seq]) data to investigate genome-wide control of the human beta cell metabolic programme. RESULTS Rela deficiency resulted in complete loss of stimulus-dependent inflammatory gene upregulation, consistent with its known role in governing inflammation. However, Rela deletion also rendered mice glucose intolerant because of functional loss of insulin secretion. Glucose intolerance was intrinsic to beta cells as βp65KO islets failed to secrete insulin ex vivo in response to a glucose challenge and were unable to restore metabolic control when transplanted into secondary chemical-induced hyperglycaemic recipients. Maintenance of glucose tolerance required Rela but was independent of classical NF-κB inflammatory cascades, as blocking NF-κB signalling in vivo by beta cell knockout of Ikbkg (NEMO), or beta cell overexpression of Tnfaip3 (A20), did not cause severe glucose intolerance. Thus, basal p65 activity has an essential and islet-intrinsic role in maintaining normal glucose homeostasis. Genome-wide bioinformatic mapping revealed the presence of p65 binding sites in the promoter regions of specific metabolic genes and in the majority of islet enhancer hubs (~70% of ~1300 hubs), which are responsible for shaping beta cell type-specific gene expression programmes. Indeed, the islet-specific metabolic genes Slc2a2, Capn9 and Pfkm identified within the large network of islet enhancer hub genes showed dysregulated expression in βp65KO islets. CONCLUSIONS/INTERPRETATION These data demonstrate an unappreciated role for RELA as a regulator of islet-specific transcriptional programmes necessary for the maintenance of healthy glucose metabolism. These findings have clinical implications for the use of anti-inflammatories, which influence NF-κB activation and are associated with diabetes.
Collapse
Affiliation(s)
- Nathan W Zammit
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Ying Ying Wong
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Stacey N Walters
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Joanna Warren
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Translation Science Pillar, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Simon C Barry
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Shane T Grey
- Transplantation Immunology Laboratory, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
- Translation Science Pillar, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
5
|
Sanders HM, Chalyavi F, Fields CR, Kostelic MM, Li MH, Raleigh DP, Zanni MT, Marty MT. Interspecies Variation Affects Islet Amyloid Polypeptide Membrane Binding. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:986-990. [PMID: 37126782 PMCID: PMC10330443 DOI: 10.1021/jasms.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The aggregation of islet amyloid polypeptide (IAPP) is associated with β-cell dysfunction in type 2 diabetes (T2D) in humans. One possible mechanism of toxicity is the interaction of IAPP oligomers with lipid membranes to disrupt the bilayer integrity and/or homeostasis of the cell. Amino acid sequence variations of IAPPs between species can greatly decrease their propensity for aggregation. For example, human IAPP is toxic to β-cells, but rat and pig IAPP are not. However, it is not clear how these differences affect membrane association. Using native mass spectrometry with lipid nanodiscs, we explored the differences in the association of human, rat, and pig IAPP with lipid bilayers. We discovered that human and rat IAPP bound nanodiscs with anionic dipalmitoyl-phosphatidylglycerol (DPPG) lipids, but pig IAPP did not. Furthermore, human and rat IAPP interacted differently with the membrane. Human IAPP show potential tetramer complexes, but rat IAPP associated with the membrane sequentially. Thus, overall IAPP-bilayer interactions are not necessarily related to disease, but small differences in oligomeric behavior at the membrane may instead play a role.
Collapse
Affiliation(s)
- Henry M. Sanders
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Farzaneh Chalyavi
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Caitlyn R. Fields
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Marius M. Kostelic
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Ming-Hao Li
- Department of Chemistry and Laufer Center for Quantitative Biology, Stony Brook University, 100 Nicolls Rd., Stony Brook, New York 11794, USA
| | - Daniel P. Raleigh
- Department of Chemistry and Laufer Center for Quantitative Biology, Stony Brook University, 100 Nicolls Rd., Stony Brook, New York 11794, USA
| | - Martin T. Zanni
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michael T. Marty
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
6
|
Brownrigg GP, Xia YH, Chu CMJ, Wang S, Chao C, Zhang JA, Skovsø S, Panzhinskiy E, Hu X, Johnson JD, Rideout EJ. Sex differences in islet stress responses support female β cell resilience. Mol Metab 2023; 69:101678. [PMID: 36690328 PMCID: PMC9971554 DOI: 10.1016/j.molmet.2023.101678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/07/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Pancreatic β cells play a key role in maintaining glucose homeostasis; dysfunction of this critical cell type causes type 2 diabetes (T2D). Emerging evidence points to sex differences in β cells, but few studies have examined male-female differences in β cell stress responses and resilience across multiple contexts, including diabetes. Here, we address the need for high-quality information on sex differences in β cell and islet gene expression and function using both human and rodent samples. METHODS In humans, we compared β cell gene expression and insulin secretion in donors with T2D to non-diabetic donors in both males and females. In mice, we generated a well-powered islet RNAseq dataset from 20-week-old male and female siblings with similar insulin sensitivity. Our unbiased gene expression analysis pointed to a sex difference in the endoplasmic reticulum (ER) stress response. Based on this analysis, we hypothesized female islets would be more resilient to ER stress than male islets. To test this, we subjected islets isolated from age-matched male and female mice to thapsigargin treatment and monitored protein synthesis, cell death, and β cell insulin production and secretion. Transcriptomic and proteomic analyses were used to characterize sex differences in islet responses to ER stress. RESULTS Our single-cell analysis of human β cells revealed sex-specific changes to gene expression and function in T2D, correlating with more robust insulin secretion in human islets isolated from female donors with T2D compared to male donors with T2D. In mice, RNA sequencing revealed differential enrichment of unfolded protein response pathway-associated genes, where female islets showed higher expression of genes linked with protein synthesis, folding, and processing. This differential expression was physiologically significant, as islets isolated from female mice were more resilient to ER stress induction with thapsigargin. Specifically, female islets showed a greater ability to maintain glucose-stimulated insulin production and secretion during ER stress compared with males. CONCLUSIONS Our data demonstrate sex differences in β cell gene expression in both humans and mice, and that female β cells show a greater ability to maintain glucose-stimulated insulin secretion across multiple physiological and pathological contexts.
Collapse
Affiliation(s)
- George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yi Han Xia
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chieh Min Jamie Chu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Su Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jiashuo Aaron Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Evgeniy Panzhinskiy
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
7
|
Szunerits S, Abderrahmani A, Boukherroub R. Nanoparticles and Nanocolloidal Carbon: Will They Be the Next Antidiabetic Class That Targets Fibrillation and Aggregation of Human Islet Amyloid Polypeptide in Type 2 Diabetes? Acc Chem Res 2022; 55:2869-2881. [PMID: 36174237 DOI: 10.1021/acs.accounts.2c00415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nanotechnology is revolutionizing human medicine. Nanoparticles (NPs) are currently used for treating various cancers, for developing vaccines, and for imaging, and other promises offered by NPs might come true soon. Due to the interplay between NPs and proteins, there is more and more evidence supporting the role of NPs for treating amyloid-based diseases. NPs can induce some conformational changes of the adsorbed protein molecules via various molecular interactions, leading to inhibition of aggregation and fibrillation of several and different amyloid proteins. Though an in depth understanding of such interactions between NPs and amyloid structures is still lacking, the inhibition of protein aggregation by NPs represents a new generation of innovative and effective medicines to combat metabolic diseases such as type 2 diabetes (T2D). Here, we lay out advances made in the field of T2D notably for optimizing protein aggregation inhibition strategies. This Account covers discussions about the current understanding of β-cells, the insulin producing cells within the pancreas, under diabetic conditions, notably increased glucose and fatty acid levels, and the implication of these conditions on the formation of human islet amyloid polypeptide (hIAPP) amylin oligomers and aggregates. Owing to the great potential of carbon nanostructures to interfere with protein aggregation, an important part of this Account will be devoted to the state of the art of therapeutic options in the form of emerging nanomaterials-based amyloidosis inhibitors. Our group has recently made some substantial progress in this regard by investigating the impact of glucose and fatty acid concentrations on hIAPP aggregation and β-cell toxicity. Furthermore, the great potential of carbon nanocolloids in reversing hIAPP aggregation under diabetic conditions will be highlighted as the approach has been validated on β-cell cultures from rats. We hope that this Account will evoke new ideas and concepts in this regard. We give some lead references below on pancreatic β-cell aspects and carbon quantum dots for managing diabetics and nanomedicine related aspects, a topic of interest in our laboratory.
Collapse
Affiliation(s)
- Sabine Szunerits
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France
| |
Collapse
|
8
|
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: 4] [Impact Index Per Article: 2.0] [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
|
9
|
Castillo JJ, Aplin AC, Hackney DJ, Hogan MF, Esser N, Templin AT, Akter R, Kahn SE, Raleigh DP, Zraika S, Hull RL. Islet amyloid polypeptide aggregation exerts cytotoxic and proinflammatory effects on the islet vasculature in mice. Diabetologia 2022; 65:1687-1700. [PMID: 35871651 PMCID: PMC10208275 DOI: 10.1007/s00125-022-05756-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/28/2022] [Indexed: 01/29/2023]
Abstract
AIMS/HYPOTHESIS The islet vasculature, including its constituent islet endothelial cells, is a key contributor to the microenvironment necessary for normal beta cell health and function. In type 2 diabetes, islet amyloid polypeptide (IAPP) aggregates, forming amyloid deposits that accumulate between beta cells and islet capillaries. This process is known to be toxic to beta cells but its impact on the islet vasculature has not previously been studied. Here, we report the first characterisation of the effects of IAPP aggregation on islet endothelial cells/capillaries using cell-based and animal models. METHODS Primary and immortalised islet endothelial cells were treated with amyloidogenic human IAPP (hIAPP) alone or in the presence of the amyloid blocker Congo Red or the Toll-like receptor (TLR) 2/4 antagonist OxPAPc. Cell viability was determined0 along with mRNA and protein levels of inflammatory markers. Islet capillary abundance, morphology and pericyte coverage were determined in pancreases from transgenic mice with beta cell expression of hIAPP using conventional and confocal microscopy. RESULTS Aggregated hIAPP decreased endothelial cell viability in immortalised and primary islet endothelial cells (by 78% and 60%, respectively) and significantly increased expression of inflammatory markers Il6, Vcam1 and Edn1 mRNA relative to vehicle treatment in both cell types (p<0.05; n=4). Both cytotoxicity and the proinflammatory response were ameliorated by Congo Red (p<0.05; n=4); whereas TLR2/4-inhibition blocked inflammatory gene expression (p<0.05; n=6) without improving viability. Islets from high-fat-diet-fed amyloid-laden hIAPP transgenic mice also exhibited significantly increased expression of most markers of endothelial inflammation (p<0.05; n=5) along with decreased capillary density compared with non-transgenic littermates fed the same diet (p<0.01). Moreover, a 16% increase in capillary diameter was observed in amyloid-adjacent capillaries (p<0.01), accompanied by a doubling in pericyte structures positive for neuron-glial antigen 2 (p<0.001). CONCLUSIONS/INTERPRETATION Islet endothelial cells are susceptible to hIAPP-induced cytotoxicity and exhibit a TLR2/4-dependent proinflammatory response to aggregated hIAPP. Additionally, we observed amyloid-selective effects that decreased islet capillary density, accompanied by increased capillary diameter and increased pericyte number. Together, these data demonstrate that the islet vasculature is a target of the cytotoxic and proinflammatory effects of aggregated hIAPP that likely contribute to the detrimental effects of hIAPP aggregation on beta cell function and survival in type 2 diabetes.
Collapse
Affiliation(s)
- Joseph J Castillo
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alfred C Aplin
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Daryl J Hackney
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - Meghan F Hogan
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nathalie Esser
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew T Templin
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rehana Akter
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Steven E Kahn
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Daniel P Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, NY, USA
- Research Department of Structural and Molecular Biology, University College London, London, UK
| | - Sakeneh Zraika
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rebecca L Hull
- Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
10
|
Oh SJ, Lee MS. Role of Autophagy in the Pathogenesis of Diabetes and Therapeutic Potential of Autophagy Modulators in the Treatment of Diabetes and Metabolic Syndrome. J Korean Med Sci 2022; 37:e276. [PMID: 36163475 PMCID: PMC9512677 DOI: 10.3346/jkms.2022.37.e276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/25/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is critically involved in the maintenance of intracellular nutrient homeostasis and organelle function. Dysregulated autophagy is likely to play a role in the development of metabolic disorders and diabetes because autophagy is critical in the rejuvenation of dysfunctional or stressed endoplasmic reticulum and mitochondria that play a crucial role in the development of diabetes. Indeed, systemic autophagy insufficiency led to the increased tissue lipid content, aggravated metabolic and finally more severe diabetes when metabolic stress was imposed, suggesting that autophagy insufficiency of dysfunction of lysosome, an effector organelle of autophagy, due to aging, genetic predisposition or environmental factors could be an underlying cause of diabetes. Conversely, autophagy enhancer could improve metabolic profile of obese mice by reducing tissue lipid content and ameliorating metabolic inflammation. Furthermore, clearance of human islet amyloid polypeptide (hIAPP) oligomer and amyloid that accumulate in pancreatic islets of > 90% of diabetes patients was also dependent on autophagy. Consistently, autophagy enhancer could improve glucose profile and β-cell function of transgenic mice expressing amyloidogenic hIAPP in pancreatic β-cells, which was accompanied by reduced accumulation of hIAPP oligomer or amyloid, ameliorated β-cell apoptosis and increased β-cell mass. These results suggest that autophagy enhancer could be a novel therapeutic modality against diabetes associated with lipid overload and human diabetes characterized by islet amyloid accumulation.
Collapse
Affiliation(s)
- Soo-Jin Oh
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Myung-Shik Lee
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Korea.
| |
Collapse
|
11
|
Abstract
Diabetes is a chronic metabolic disease affecting an increasing number of people. Although diabetes has negative health outcomes for diagnosed individuals, a population at particular risk are pregnant women, as diabetes impacts not only a pregnant woman's health but that of her child. In this review, we cover the current knowledge and unanswered questions on diabetes affecting an expectant mother, focusing on maternal and fetal outcomes.
Collapse
Affiliation(s)
- Cecilia González Corona
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA
| | - Ronald J. Parchem
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
SORLA mediates endocytic uptake of proIAPP and protects against islet amyloid deposition. Mol Metab 2022; 65:101585. [PMID: 36055578 PMCID: PMC9474563 DOI: 10.1016/j.molmet.2022.101585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/27/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
Objective Sorting-related receptor with type A repeats (SORLA) is a neuronal sorting receptor that prevents accumulation of amyloid-beta peptides, the main constituent of senile plaques in Alzheimer disease. Recent transcriptomic studies show that SORLA transcripts are also found in beta cells of pancreatic islets, yet the role of SORLA in islets is unknown. Based on its protective role in reducing the amyloid burden in the brain, we hypothesized that SORLA has a similar function in the pancreas via regulation of amyloid formation from islet amyloid polypeptide (IAPP). Methods We generated human IAPP transgenic mice lacking SORLA (hIAPP:SORLA KO) to assess the consequences of receptor deficiency for islet histopathology and function in vivo. Using both primary islet cells and cell lines, we further investigated the molecular mechanisms whereby SORLA controls the cellular metabolism and accumulation of IAPP. Results Loss of SORLA activity in hIAPP:SORLA KO resulted in a significant increase in islet amyloid deposits and associated islet cell death compared to hIAPP:SORLA WT animals. Aggravated islet amyloid deposition was observed in mice fed a normal chow diet, not requiring high-fat diet feeding typically needed to induce islet amyloidosis in mouse models. In vitro studies showed that SORLA binds to and mediates the endocytic uptake of proIAPP, but not mature IAPP, delivering the propeptide to an endolysosomal fate. Conclusions SORLA functions as a proIAPP-specific clearance receptor, protecting against islet amyloid deposition and associated cell death caused by IAPP. SORLA is an endocytic receptor for amyloidogenic peptides expressed in islet beta cells. SORLA mediates cellular clearance of proIAPP. Loss of SORLA activity in mouse models causes spontaneous islet amyloid deposition.
Collapse
|
13
|
Taneera J, Ali A, Hamad M. The Role of Estrogen Signaling in Cellular Iron Metabolism in Pancreatic β Cells. Pancreas 2022; 51:121-127. [PMID: 35404886 DOI: 10.1097/mpa.0000000000001978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT Several lines of evidence suggest that estrogen (17-β estradiol; E2) protects against diabetes mellitus and plays important roles in pancreatic β-cell survival and function. Mounting clinical and experimental evidence also suggest that E2 modulates cellular iron metabolism by regulating the expression of several iron regulatory genes, including hepcidin (HAMP), hypoxia-inducible factor 1-α, ferroportin (SLC40A1), and lipocalin (LCN2). However, whether E2 regulates cellular iron metabolism in pancreatic β cells and whether the antidiabetic effects of E2 can be, at least partially, attributed to its role in iron metabolism is not known. In this context, pancreatic β cells express considerable levels of conventional E2 receptors (ERs; mainly ER-α) and nonconventional G protein-coupled estrogen receptors and hence responsive to E2 signals. Moreover, pancreatic islet cells require significant amounts of iron for proper functioning, replication and survival and, hence, well equipped to manage cellular iron metabolism (acquisition, utilization, storage, and release). In this review, we examine the link between E2 and cellular iron metabolism in pancreatic β cells and discuss the bearing of such a link on β-cell survival and function.
Collapse
Affiliation(s)
| | - Amjad Ali
- From the Research Institute for Medical and Health Sciences
| | - Mawieh Hamad
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
14
|
Mukherjee N, Lin L, Contreras CJ, Templin AT. β-Cell Death in Diabetes: Past Discoveries, Present Understanding, and Potential Future Advances. Metabolites 2021; 11:796. [PMID: 34822454 PMCID: PMC8620854 DOI: 10.3390/metabo11110796] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022] Open
Abstract
β-cell death is regarded as a major event driving loss of insulin secretion and hyperglycemia in both type 1 and type 2 diabetes mellitus. In this review, we explore past, present, and potential future advances in our understanding of the mechanisms that promote β-cell death in diabetes, with a focus on the primary literature. We first review discoveries of insulin insufficiency, β-cell loss, and β-cell death in human diabetes. We discuss findings in humans and mouse models of diabetes related to autoimmune-associated β-cell loss and the roles of autoreactive T cells, B cells, and the β cell itself in this process. We review discoveries of the molecular mechanisms that underlie β-cell death-inducing stimuli, including proinflammatory cytokines, islet amyloid formation, ER stress, oxidative stress, glucotoxicity, and lipotoxicity. Finally, we explore recent perspectives on β-cell death in diabetes, including: (1) the role of the β cell in its own demise, (2) methods and terminology for identifying diverse mechanisms of β-cell death, and (3) whether non-canonical forms of β-cell death, such as regulated necrosis, contribute to islet inflammation and β-cell loss in diabetes. We believe new perspectives on the mechanisms of β-cell death in diabetes will provide a better understanding of this pathological process and may lead to new therapeutic strategies to protect β cells in the setting of diabetes.
Collapse
Affiliation(s)
- Noyonika Mukherjee
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA; (L.L.); (C.J.C.)
| | - Li Lin
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA; (L.L.); (C.J.C.)
| | - Christopher J. Contreras
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA; (L.L.); (C.J.C.)
- Department of Medicine, Roudebush Veterans Affairs Medical Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew T. Templin
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA; (L.L.); (C.J.C.)
- Department of Medicine, Roudebush Veterans Affairs Medical Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
15
|
Wong HY, Hui Q, Hao Z, Warnock GL, Woo M, Luciani DS, Marzban L. The role of mitochondrial apoptotic pathway in islet amyloid-induced β-cell death. Mol Cell Endocrinol 2021; 537:111424. [PMID: 34400259 DOI: 10.1016/j.mce.2021.111424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/24/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022]
Abstract
Islet amyloid, formed by aggregation of human islet amyloid polypeptide (hIAPP), contributes to β-cell death in type 2 diabetes. We previously showed that extracellular hIAPP aggregates promote Fas-mediated β-cell apoptosis. Here, we tested if hIAPP aggregates can trigger the mitochondrial apoptotic pathway (MAP). hIAPP aggregation in Ad-hIAPP transduced INS-1 and human islet β-cells promoted cytochrome c release, caspase-9 activation and apoptosis, which were reduced by Bax inhibitor. Amyloid formation in hIAPP-expressing mouse islets during culture increased caspase-9 activation in β-cells. Ad-hIAPP transduced islets from CytcKA/KA and BaxBak βDKO mice (models of blocked MAP), had lower caspase-9-positive and apoptotic β-cells than transduced wild-type islets, despite comparable amyloid formation. Blocking Fas (markedly) and Bax or caspase-9 (modestly) reduced β-cell death induced by extracellular hIAPP aggregates. These findings suggest a role for MAP in amyloid-induced β-cell death and a potential strategy to reduce intracellular amyloid β-cell toxicity by blocking cytochrome c apoptotic function.
Collapse
Affiliation(s)
- Helen Y Wong
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Queenie Hui
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Zhenyue Hao
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Garth L Warnock
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute and Division of Endocrinology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Dan S Luciani
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Lucy Marzban
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada; College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
16
|
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: 2.3] [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
|
17
|
4-Phenylbutyrate (PBA) treatment reduces hyperglycemia and islet amyloid in a mouse model of type 2 diabetes and obesity. Sci Rep 2021; 11:11878. [PMID: 34088954 PMCID: PMC8178353 DOI: 10.1038/s41598-021-91311-2] [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: 03/03/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
Amyloid deposits in pancreatic islets, mainly formed by human islet amyloid polypeptide (hIAPP) aggregation, have been associated with loss of β-cell mass and function, and are a pathological hallmark of type 2 diabetes (T2D). Treatment with chaperones has been associated with a decrease in endoplasmic reticulum stress leading to improved glucose metabolism. The aim of this work was to investigate whether the chemical chaperone 4-phenylbutyrate (PBA) prevents glucose metabolism abnormalities and amyloid deposition in obese agouti viable yellow (Avy) mice that overexpress hIAPP in β cells (Avy hIAPP mice), which exhibit overt diabetes. Oral PBA treatment started at 8 weeks of age, when Avy hIAPP mice already presented fasting hyperglycemia, glucose intolerance, and impaired insulin secretion. PBA treatment strongly reduced the severe hyperglycemia observed in obese Avy hIAPP mice in fasting and fed conditions throughout the study. This effect was paralleled by a decrease in hyperinsulinemia. Importantly, PBA treatment reduced the prevalence and the severity of islet amyloid deposition in Avy hIAPP mice. Collectively, these results show that PBA treatment elicits a marked reduction of hyperglycemia and reduces amyloid deposits in obese and diabetic mice, highlighting the potential of chaperones for T2D treatment.
Collapse
|
18
|
Van Hulle F, De Groot K, Stangé G, Suenens K, De Mesmaeker I, De Paep DL, Ling Z, Hilbrands R, Gillard P, Keymeulen B, Kroon E, Westermark GT, Jacobs-Tulleneers-Thevissen D, Pipeleers D. Formation of amyloid in encapsulated human pancreatic and human stem cell-generated beta cell implants. Am J Transplant 2021; 21:2090-2099. [PMID: 33206461 DOI: 10.1111/ajt.16398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 01/25/2023]
Abstract
Detection of amyloid in intraportal islet implants of type 1 diabetes patients has been proposed as cause in their functional decline. The present study uses cultured adult human islets devoid of amyloid to examine conditions of its formation. After intraportal injection in patients, amyloid deposits <15 µm diameter were identified in 5%-12% of beta cell containing aggregates, 3-76 months posttransplant. Such deposits also formed in glucose-controlling islet implants in the kidney of diabetic mice but not in failing implants. Alginate-encapsulated islets formed amyloid during culture when functional, and in all intraperitoneal implants that corrected diabetes in mice, exhibiting larger sizes than in functioning nonencapsulated implants. After intraperitoneal injection in a patient, retrieved single capsules presented amyloid near living beta cells, whereas no amyloid occurred in clustered capsules with dead cells. Amyloid was also demonstrated in functional human stem cell-generated beta cell implants in subcutaneous devices of mice. Deposits up to 35 µm diameter were localized in beta cell-enriched regions and related to an elevated IAPP over insulin ratio in the newly generated beta cells. Amyloid in device-encapsulated human stem cell-generated beta cell implants marks the formation of a functional beta cell mass but also an imbalance between its activated state and its microenvironment.
Collapse
Affiliation(s)
- Freya Van Hulle
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Internal Medicine, University Hospital Brussels - UZB, Brussels, Belgium
| | - Kaat De Groot
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Internal Medicine, University Hospital Brussels - UZB, Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium
| | - Krista Suenens
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium
| | - Ines De Mesmaeker
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium
| | - Diedert L De Paep
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Department Surgery, University Hospital Brussels - UZB, Brussels, Belgium.,Beta Cell Bank, University Hospital Brussels - UZB, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Beta Cell Bank, University Hospital Brussels - UZB, Brussels, Belgium.,Consortium, Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| | - Robert Hilbrands
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Diabetes Clinic, University Hospital Brussels - UZB, Brussels, Belgium
| | - Pieter Gillard
- Department Endocrinology, University Hospital Leuven - KUL, Leuven, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Consortium, Center for Beta Cell Therapy in Diabetes, Brussels, Belgium.,Diabetes Clinic, University Hospital Brussels - UZB, Brussels, Belgium
| | - Evert Kroon
- Consortium, Center for Beta Cell Therapy in Diabetes, Brussels, Belgium.,ViaCyte, Inc, San Diego, California, USA
| | | | - Daniel Jacobs-Tulleneers-Thevissen
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Department Surgery, University Hospital Brussels - UZB, Brussels, Belgium.,Consortium, Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Research Center, Free University Brussels - VUB, Brussels, Belgium.,Consortium, Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| |
Collapse
|
19
|
Burillo J, Marqués P, Jiménez B, González-Blanco C, Benito M, Guillén C. Insulin Resistance and Diabetes Mellitus in Alzheimer's Disease. Cells 2021; 10:1236. [PMID: 34069890 PMCID: PMC8157600 DOI: 10.3390/cells10051236] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus is a progressive disease that is characterized by the appearance of insulin resistance. The term insulin resistance is very wide and could affect different proteins involved in insulin signaling, as well as other mechanisms. In this review, we have analyzed the main molecular mechanisms that could be involved in the connection between type 2 diabetes and neurodegeneration, in general, and more specifically with the appearance of Alzheimer's disease. We have studied, in more detail, the different processes involved, such as inflammation, endoplasmic reticulum stress, autophagy, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jesús Burillo
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos González-Blanco
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, 28040 Madrid, Spain; (J.B.); (P.M.); (B.J.); (C.G.-B.); (M.B.)
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28040 Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), 28040 Madrid, Spain
| |
Collapse
|
20
|
Sexual hormones and diabetes: The impact of estradiol in pancreatic β cell. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021. [PMID: 33832654 DOI: 10.1016/bs.ircmb.2021.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
Diabetes is one of the most prevalent metabolic diseases and its incidence is increasing throughout the world. Data from World Health Organization (WHO) point-out that diabetes is a major cause of blindness, kidney failure, heart attacks, stroke and lower limb amputation and estimated 1.6 million deaths were directly caused by it in 2016. Population studies show that the incidence of this disease increases in women after menopause, when the production of estrogen is decreasing in them. Knowing the impact that estrogenic signaling has on insulin-secreting β cells is key to prevention and design of new therapeutic targets. This chapter explores the role of estrogen and their receptors in the regulation of insulin secretion and biosynthesis, proliferation, regeneration and survival in pancreatic β cells. In addition, delves into the genetic animal models developed and its application for the specific study of the different estrogen signaling pathways. Finally, discusses the impact of menopause and hormone replacement therapy on pancreatic β cell function.
Collapse
|
21
|
Tang Y, Zhang D, Zhang Y, Liu Y, Gong X, Chang Y, Ren B, Zheng J. Introduction and Fundamentals of Human Islet Amyloid Polypeptide Inhibitors. ACS APPLIED BIO MATERIALS 2020; 3:8286-8308. [DOI: 10.1021/acsabm.0c01234] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yijing Tang
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| | - Dong Zhang
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| | - Yanxian Zhang
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| | - Yonglan Liu
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| | - Xiong Gong
- Department of Polymer Engineering, The University of Akron, Akron, Ohio 44325-0301, United States
| | - Yung Chang
- Department of Chemical Engineering, R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan
| | - Baiping Ren
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| | - Jie Zheng
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio 44325-3906, United States
| |
Collapse
|
22
|
Araújo AR, Reis RL, Pires RA. Natural Polyphenols as Modulators of the Fibrillization of Islet Amyloid Polypeptide. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1250:159-176. [PMID: 32601944 DOI: 10.1007/978-981-15-3262-7_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Diabetes mellitus type 2 (type-2 diabetes) is a metabolic disorder characterized by the increased blood glucose concentration and insulin resistance in peripheral tissues (e.g., muscles and adipose tissue). The initiation of the pathological cascade of events that lead to type-2 diabetes has been subject of debate; however, it has been commonly accepted that the oversecretion of human islet amyloid polypeptide (hIAPP, a hormone co-secreted with insulin) by the pancreatic 𝛽-cells is the main trigger of type-2 diabetes. In fact, 90% of the type-2 diabetes patients present hIAPP deposits in the extracellular space of the 𝛽-cells. These hIAPP supramolecular arrangements (both fibrillar and oligomeric) have been reported to be the origin of cytotoxicity, which leads to 𝛽-cell dysfunction through a series of different mechanisms, including the interaction of hIAPP oligomers with the cell membrane that leads to the influx of Ca2+ and increase in the cellular oxidative stress, among others. This overview shows the importance of developing type-2 diabetes treatment strategies able to (1) remodel of the secondary structure of cytotoxic hIAPP oligomers entrapping them into off-pathway nontoxic species and (2) reestablish physiological levels of oxidative stress. Natural polyphenols are a class of antioxidant compounds that are able to perform both functions. Herein we review the published literature of the most studied polyphenols, in particular for their ability to remodel the hIAPP aggregation pathway, to rescue the in vitro pancreatic 𝛽-cell viability and function, as well as to perform under a complex biological environment, i.e., in vivo animal models and clinical trials. Overall, natural polyphenols are able to control the cytotoxic hIAPP aggregation and minimize hIAPP-mediated cellular dysfunction and can be considered as important lead compounds for the treatment of type-2 diabetes.
Collapse
Affiliation(s)
- Ana R Araújo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal
| | - Ricardo A Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal. .,The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Guimarães, Portugal.
| |
Collapse
|
23
|
Pandey P, Nguyen N, Hansmann UHE. d-Retro Inverso Amylin and the Stability of Amylin Fibrils. J Chem Theory Comput 2020; 16:5358-5368. [PMID: 32667784 DOI: 10.1021/acs.jctc.0c00523] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Motivated by the role that amylin aggregates play in type-II diabetes, we compare the stability of regular amylin fibrils with the stability of fibrils where l-amino acid chains are replaced by d-retro inverso (DRI) amylin, that is, peptides where the sequence of amino acids is reversed, and at the same time, the l-amino acids are replaced by their mirror images. Our molecular dynamics simulations show that despite leading to only a marginal difference in the fibril structure and stability, aggregating DRI-amylin peptides have different patterns of contacts and hydrogen bonding. Because of these differences, DRI-amylin, when interacting with regular (l) amylin, alters the elongation process and lowers the stability of hybrid amylin fibrils. Our results not only suggest the potential use of DRI-amylin as an inhibitor of amylin fibril formation but also point to the possibility of using the insertion of DRI proteins in l-assemblies as a way to probe the role of certain kinds of hydrogen bonds in supramolecular assemblies or aggregates.
Collapse
Affiliation(s)
- Preeti Pandey
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Natalie Nguyen
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Ulrich H E Hansmann
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
24
|
Cao Q, Boyer DR, Sawaya MR, Ge P, Eisenberg DS. Cryo-EM structure and inhibitor design of human IAPP (amylin) fibrils. Nat Struct Mol Biol 2020; 27:653-659. [PMID: 32541896 PMCID: PMC8579859 DOI: 10.1038/s41594-020-0435-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Human islet amyloid polypeptide (hIAPP) functions as a glucose-regulating hormone but deposits as amyloid fibrils in more than 90% of patients with type II diabetes (T2D). Here we report the cryo-EM structure of recombinant full-length hIAPP fibrils. The fibril is composed of two symmetrically related protofilaments with ordered residues 14-37. Our hIAPP fibril structure (i) supports the previous hypothesis that residues 20-29 constitute the core of the hIAPP amyloid; (ii) suggests a molecular mechanism for the action of the hIAPP hereditary mutation S20G; (iii) explains why the six residue substitutions in rodent IAPP prevent aggregation; and (iv) suggests regions responsible for the observed hIAPP cross-seeding with β-amyloid. Furthermore, we performed structure-based inhibitor design to generate potential hIAPP aggregation inhibitors. Four of the designed peptides delay hIAPP aggregation in vitro, providing a starting point for the development of T2D therapeutics and proof of concept that the capping strategy can be used on full-length cryo-EM fibril structures.
Collapse
Affiliation(s)
- Qin Cao
- Department of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - David R Boyer
- Department of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael R Sawaya
- Department of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peng Ge
- California NanoSystem Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - David S Eisenberg
- Department of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute, Molecular Biology Institute, and Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
25
|
Noh D, Bower RL, Hay DL, Zhyvoloup A, Raleigh DP. Analysis of Amylin Consensus Sequences Suggests That Human Amylin Is Not Optimized to Minimize Amyloid Formation and Provides Clues to Factors That Modulate Amyloidogenicity. ACS Chem Biol 2020; 15:1408-1416. [PMID: 32364695 DOI: 10.1021/acschembio.9b01050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The neuropancreatic polypeptide hormone amylin forms pancreatic islet amyloid in type-2 diabetes. Islet amyloid formation contributes to β-cell death in the disease and to the failure of islet transplants, but the features which influence amylin amyloidogenicity are not understood. We constructed an amino acid sequence alignment of 202 sequences of amylin and used the alignment to design consensus sequences of vertebrate amylins, mammalian amylins, and primate amylins. Amylin is highly conserved, but there are differences between human amylin and each consensus sequence, ranging from one to six substitutions. Biophysical analysis shows that all of the consensus sequences form amyloid but do so more slowly than human amylin in vitro. The rate of amyloid formation by the primate consensus sequence is 3- to 4-fold slower than human amylin; the mammalian consensus sequence is approximately 20- to 25-fold slower, and the vertebrate consensus sequence is approximately 6-fold slower. All of the consensus sequences are moderately less toxic than human amylin toward a cultured β-cell line, with the vertebrate consensus sequence displaying the largest reduction in toxicity of 3- to 4-fold. All of the consensus sequences activate a human amylin receptor and exhibit only modest reductions in activity, ranging from 3- to 4-fold as judged by a cAMP production assay. The analysis argues that there is no strong selective evolutionary pressure to avoid the formation of islet amyloid and provides information relevant to the design of less amyloidogenic amylin variants.
Collapse
Affiliation(s)
- Daeun Noh
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, New York 11790, United States
| | - Rebekah L. Bower
- School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Debbie L. Hay
- School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland 1142, New Zealand
| | - Alexander Zhyvoloup
- Institute of Structural and Molecular Biology, University College London, Gower Street, London, WC1E 6BT United Kingdom
| | - Daniel P. Raleigh
- Institute of Structural and Molecular Biology, University College London, Gower Street, London, WC1E 6BT United Kingdom
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11790, United States
- Laufer Center for Quantitative Biology, Stony Brook University, Stony Brook, New York 11790, United States
| |
Collapse
|
26
|
Hogan MF, Ziemann M, K N H, Rodriguez H, Kaspi A, Esser N, Templin AT, El-Osta A, Kahn SE. RNA-seq-based identification of Star upregulation by islet amyloid formation. Protein Eng Des Sel 2020; 32:67-76. [PMID: 31504890 DOI: 10.1093/protein/gzz022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 06/24/2019] [Accepted: 07/05/2019] [Indexed: 12/26/2022] Open
Abstract
Aggregation of islet amyloid polypeptide (IAPP) into islet amyloid results in β-cell toxicity in human type 2 diabetes. To determine the effect of islet amyloid formation on gene expression, we performed ribonucleic acid (RNA) sequencing (RNA-seq) analysis using cultured islets from either wild-type mice (mIAPP), which are not amyloid prone, or mice that express human IAPP (hIAPP), which develop amyloid. Comparing mIAPP and hIAPP islets, 5025 genes were differentially regulated (2439 upregulated and 2586 downregulated). When considering gene sets (reactomes), 248 and 52 pathways were up- and downregulated, respectively. Of the top 100 genes upregulated under two conditions of amyloid formation, seven were common. Of these seven genes, only steroidogenic acute regulatory protein (Star) demonstrated no effect of glucose per se to modify its expression. We confirmed this differential gene expression using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and also demonstrated the presence of STAR protein in islets containing amyloid. Furthermore, Star is a part of reactomes representing metabolism, metabolism of lipids, metabolism of steroid hormones, metabolism of steroids and pregnenolone biosynthesis. Thus, examining gene expression that is differentially regulated by islet amyloid has the ability to identify new molecules involved in islet physiology and pathology applicable to type 2 diabetes.
Collapse
Affiliation(s)
- Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA 98018, USA
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC 3004, Australia
| | - Harikrishnan K N
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC 3004, Australia
| | - Hanah Rodriguez
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC 3004, Australia
| | - Antony Kaspi
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC 3004, Australia
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA 98018, USA
| | - Andrew T Templin
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA 98018, USA
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC 3004, Australia.,Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR.,University College Copenhagen, Faculty of Health, Department of Technology, Biomedical Laboratory Science, Copenhagen, Denmark
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, WA 98018, USA
| |
Collapse
|
27
|
Templin AT, Mellati M, Soininen R, Hogan MF, Esser N, Castillo JJ, Zraika S, Kahn SE, Hull RL. Loss of perlecan heparan sulfate glycosaminoglycans lowers body weight and decreases islet amyloid deposition in human islet amyloid polypeptide transgenic mice. Protein Eng Des Sel 2020; 32:95-102. [PMID: 31769491 DOI: 10.1093/protein/gzz041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/24/2019] [Accepted: 10/01/2019] [Indexed: 11/14/2022] Open
Abstract
Islet amyloid is a pathologic feature of type 2 diabetes (T2D) that is associated with β-cell loss and dysfunction. These amyloid deposits form via aggregation of the β-cell secretory product islet amyloid polypeptide (IAPP) and contain other molecules including the heparan sulfate proteoglycan perlecan. Perlecan has been shown to bind amyloidogenic human IAPP (hIAPP) via its heparan sulfate glycosaminoglycan (HS GAG) chains and to enhance hIAPP aggregation in vitro. We postulated that reducing the HS GAG content of perlecan would also decrease islet amyloid deposition in vivo. hIAPP transgenic mice were crossed with Hspg2Δ3/Δ3 mice harboring a perlecan mutation that prevents HS GAG attachment (hIAPP;Hspg2Δ3/Δ3), and male offspring from this cross were fed a high fat diet for 12 months to induce islet amyloid deposition. At the end of the study body weight, islet amyloid area, β-cell area, glucose tolerance and insulin secretion were analyzed. hIAPP;Hspg2Δ3/Δ3 mice exhibited significantly less islet amyloid deposition and greater β-cell area compared to hIAPP mice expressing wild type perlecan. hIAPP;Hspg2Δ3/Δ3 mice also gained significantly less weight than other genotypes. When adjusted for differences in body weight using multiple linear regression modeling, we found no differences in islet amyloid deposition or β-cell area between hIAPP transgenic and hIAPP;Hspg2Δ3/Δ3 mice. We conclude that loss of perlecan exon 3 reduces islet amyloid deposition in vivo through indirect effects on body weight and possibly also through direct effects on hIAPP aggregation. Both of these mechanisms may promote maintenance of glucose homeostasis in the setting of T2D.
Collapse
Affiliation(s)
- Andrew T Templin
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Mahnaz Mellati
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Raija Soininen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Pentti Kaiteran Katu 1, Linnanmaa, Oulu, Finland
| | - Meghan F Hogan
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - J Josh Castillo
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| | - Rebecca L Hull
- Division of Metabolism, Endocrinology and Nutrition, Veterans Affairs Puget Sound Health Care System and University of Washington, 1660 South Columbian Way, Seattle, 98108, Washington, USA
| |
Collapse
|
28
|
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: 0.4] [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
|
29
|
Influence of methionine–ruthenium complex on the fibril formation of human islet amyloid polypeptide. J Biol Inorg Chem 2019; 24:179-189. [DOI: 10.1007/s00775-019-01637-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/14/2019] [Indexed: 02/07/2023]
|
30
|
Morikawa S, Kaneko N, Okumura C, Taguchi H, Kurata M, Yamamoto T, Osawa H, Nakanishi A, Zako T, Masumoto J. IAPP/amylin deposition, which is correlated with expressions of ASC and IL-1β in β-cells of Langerhans' islets, directly initiates NLRP3 inflammasome activation. Int J Immunopathol Pharmacol 2018; 32:2058738418788749. [PMID: 30014749 PMCID: PMC6050799 DOI: 10.1177/2058738418788749] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recent findings revealed that type 2 diabetes mellitus (T2D) is a chronic inflammatory disease and an islet amyloid polypeptide (IAPP)/amylin, is deposited within pancreatic islets. IAPP/amylin has been reported to activate NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in infiltrated macrophages. NLRP3, an intracellular pattern recognition receptor, has been shown to recognize pathogens and/or metabolites and complexes with the adopter protein apoptosis-associated speck-like protein containing a caspase-recruitment domain ASC to form a huge complex, called an inflammasome, an interleukin (IL)-1β-processing platform. Although reactive oxygen species (ROS) were reported to be involved in activation of NLRP3 inflammasome, we were hypothesized that IAPP could directly activate NLRP3 inflammasome, leading to islets β-cell death. We analyzed expression of the inflammasome components ASC, NLRP3, caspase-1, IL-1β, IAPP/amylin, and insulin immunohistochemically in Langerhans' islets of autopsy cases. The initial event of NLRP3 inflammasome activation was assessed using a cell-free system consisting of NLRP3 and ASC with the amplified luminescent proximity homogeneous assay. IAPP/amylin deposition in Langerhans' islets was detected and significantly correlated with expressions of IL-1β and ASC. IAPP/amylin directly interacted with NLRP3 and initiated an interaction between NLRP3 and ASC in a cell-free system. The deposition of IAPP/amylin in β-cells of Langerhans' islets may act together with the expression level of an inflammasome component, ASC, to regulate IL-1β processing, and directly lead to the dysfunction of β-cells. The interaction between IAPP/amylin and NLRP3 could be an attractive drug target to avoid both inflammation and β-cell death for T2D therapy.
Collapse
Affiliation(s)
- Shinnosuke Morikawa
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Naoe Kaneko
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Chikara Okumura
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Haruka Taguchi
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Mie Kurata
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Toshihiro Yamamoto
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| | - Haruhiko Osawa
- 2 Department of Diabetes and Molecular Genetics, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Ayaka Nakanishi
- 3 Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Matsuyama, Japan
| | - Tamotsu Zako
- 3 Department of Chemistry and Biology, Graduate School of Science and Engineering, Ehime University, Matsuyama, Japan
| | - Junya Masumoto
- 1 Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Japan
| |
Collapse
|
31
|
Templin AT, Meier DT, Willard JR, Wolden-Hanson T, Conway K, Lin YG, Gillespie PJ, Bokvist KB, Attardo G, Kahn SE, Scheuner D, Hull RL. Use of the PET ligand florbetapir for in vivo imaging of pancreatic islet amyloid deposits in hIAPP transgenic mice. Diabetologia 2018; 61:2215-2224. [PMID: 30046852 PMCID: PMC6119478 DOI: 10.1007/s00125-018-4695-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/19/2018] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Islet amyloid deposits contribute to beta cell dysfunction and death in most individuals with type 2 diabetes but non-invasive methods to determine the presence of these pathological protein aggregates are currently not available. Therefore, we examined whether florbetapir, a radiopharmaceutical agent used for detection of amyloid-β deposits in the brain, also allows identification of islet amyloid in the pancreas. METHODS Saturation binding assays were used to determine the affinity of florbetapir for human islet amyloid polypeptide (hIAPP) aggregates in vitro. Islet amyloid-prone transgenic mice that express hIAPP in their beta cells and amyloid-free non-transgenic control mice were used to examine the ability of florbetapir to detect islet amyloid deposits in vitro, in vivo and ex vivo. Mice or mouse pancreases were subjected to autoradiographic, histochemical and/or positron emission tomography (PET) analyses to assess the utility of florbetapir in identifying islet amyloid. RESULTS In vitro, florbetapir bound synthetic hIAPP fibrils with a dissociation constant of 7.9 nmol/l. Additionally, florbetapir bound preferentially to amyloid-containing hIAPP transgenic vs amyloid-free non-transgenic mouse pancreas sections in vitro, as determined by autoradiography (16,475 ± 5581 vs 5762 ± 575 density/unit area, p < 0.05). In hIAPP transgenic and non-transgenic mice fed a high-fat diet for 1 year, intravenous administration of florbetapir followed by PET scanning showed that the florbetapir signal was significantly higher in amyloid-laden hIAPP transgenic vs amyloid-free non-transgenic pancreases in vivo during the first 5 min of the scan (36.83 ± 2.22 vs 29.34 ± 2.03 standardised uptake value × min, p < 0.05). Following PET, pancreases were excised and florbetapir uptake was determined ex vivo by γ counting. Pancreatic uptake of florbetapir was significantly correlated with the degree of islet amyloid deposition, the latter assessed by histochemistry (r = 0.74, p < 0.001). CONCLUSIONS/INTERPRETATION Florbetapir binds to islet amyloid deposits in a specific and quantitative manner. In the future, florbetapir may be useful as a non-invasive tool to identify islet amyloid deposits in humans.
Collapse
Affiliation(s)
- Andrew T Templin
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
- University of Washington, Seattle, WA, USA.
| | - Daniel T Meier
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | - Joshua R Willard
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | - Tami Wolden-Hanson
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Kelly Conway
- Avid Radiopharmaceuticals, Inc., Philadelphia, PA, USA
| | - Yin-Guo Lin
- Avid Radiopharmaceuticals, Inc., Philadelphia, PA, USA
| | | | | | | | - Steven E Kahn
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| | | | - Rebecca L Hull
- Department of Medicine, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- University of Washington, Seattle, WA, USA
| |
Collapse
|
32
|
Abstract
β cell replacement with either pancreas or islet transplantation has progressed immensely over the last decades with current 1- and 5-year insulin independence rates of approximately 85% and 50%, respectively. Recent advances are largely attributed to improvements in immunosuppressive regimen, donor selection, and surgical technique. However, both strategies are compromised by a scarce donor source. Xenotransplantation offers a potential solution by providing a theoretically unlimited supply of islets, but clinical application has been limited by concerns for a potent immune response against xenogeneic tissue. β cell clusters derived from embryonic or induced pluripotent stem cells represent another promising unlimited source of insulin producing cells, but clinical application is pending further advances in the function of the β cell like clusters. Exciting developments and rapid progress in all areas of β cell replacement prompted a lively debate by members of the young investigator committee of the International Pancreas and Islet Transplant Association at the 15th International Pancreas and Islet Transplant Association Congress in Melbourne and at the 26th international congress of The Transplant Society in Hong Kong. This international group of young investigators debated which modality of β cell replacement would predominate the landscape in 10 years, and their arguments are summarized here.
Collapse
|
33
|
Rodriguez Camargo DC, Garg D, Buday K, Franko A, Rodriguez Camargo A, Schmidt F, Cox SJ, Suladze S, Haslbeck M, Mideksa YG, Gemmecker G, Aichler M, Mettenleiter G, Schulz M, Walch AK, Hrabě de Angelis M, Feige MJ, Sierra CA, Conrad M, Tripsianes K, Ramamoorthy A, Reif B. hIAPP forms toxic oligomers in plasma. Chem Commun (Camb) 2018; 54:5426-5429. [PMID: 29745410 PMCID: PMC5970100 DOI: 10.1039/c8cc03097a] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In diabetes, hyperamylinemia contributes to cardiac dysfunction. The interplay between hIAPP, blood glucose and other plasma components is, however, not understood. We show that glucose and LDL interact with hIAPP, resulting in β-sheet rich oligomers with increased β-cell toxicity and hemolytic activity, providing mechanistic insights for a direct link between diabetes and cardiovascular diseases.
Collapse
|
34
|
Abstract
The oligomerization and fibrillation of human islet amyloid polypeptide (hIAPP) play a central role in the pathogenesis of type 2 diabetes. Strategies for remodelling the formation of hIAPP oligomers and fibrils have promising application potential in type 2 diabetes therapy. Herein, we demonstrated that PEG-PE micelle could inhibit hIAPP oligomerization and fibrillation through blocking the hydrophobic interaction and the conformational change from random coil to β-sheet structures of hIAPP. In addition, we also found that PEG-PE micelle could remodel the preformed hIAPP fibrils allowing the formation of short fibrils and co-aggregates. Taken together, PEG-PE micelle could rescue hIAPP-induced cytotoxicity by decreasing the content of hIAPP oligomers and fibrils that are related to the oxidative stress and cell membrane permeability. This study could be beneficial for the design and development of antiamyloidogenic agents.
Collapse
|
35
|
Mo XD, Gao LP, Wang QJ, Yin J, Jing YH. Lipid accelerating the fibril of islet amyloid polypeptide aggravated the pancreatic islet injury in vitro and in vivo. Lipids Health Dis 2018. [PMID: 29523142 PMCID: PMC5845206 DOI: 10.1186/s12944-018-0694-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background The fibrillation of islet amyloid polypeptide (IAPP) triggered the amyloid deposition, then enhanced the loss of the pancreatic islet mass. However, it is not clear what factor is the determinant in development of the fibril formation. The aim of this study is to investigate the effects of lipid on IAPP fibril and its injury on pancreatic islet. Methods The fibril form of human IAPP (hIAPP) was tested using thioflavin-T fluorescence assay and transmission electron microscope technology after incubated with palmitate for 5 h at 25 °C. The cytotoxicity of fibril hIAPP was evaluated in INS-1 cells through analyzing the leakage of cell membrane and cell apoptosis. Type 2 diabetes mellitus (T2DM) animal model was induced with low dose streptozotocin combined the high-fat diet feeding for two months in rats. Plasma biochemistry parameters were measured before sacrificed. Pancreatic islet was isolated to evaluate their function. Results The results showed that co-incubation of hIAPP and palmitate induced more fibril form. Fibril hIAPP induced cell lesions including cell membrane leakage and cell apoptosis accompanied insulin mRNA decrease in INS-1 cell lines. In vivo, Plasma glucose, triglyceride, rIAPP and insulin increased in T2DM rats compared with the control group. In addition, IAPP and insulin mRNA increased in pancreatic islet of T2DM rats. Furthermore, T2DM induced the reduction of insulin receptor expression and cleaved caspase-3 overexpression in pancreatic islet. Conclusions Results in vivo and in vitro suggested that lipid and IAPP plays a synergistic effect on pancreatic islet cell damage, which implicated in enhancing the IAPP expression and accelerating the fibril formation of IAPP.
Collapse
Affiliation(s)
- Xiao-Dan Mo
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China
| | - Qing-Jun Wang
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China. .,Key Laboratory of Preclinical Study for New Drugs of Gansu province, Lanzhou University, No. 199 of Donggang West Road, Lanzhou City, Gansu Province, 730000, People's Republic of China.
| |
Collapse
|
36
|
Zhang Y, Warnock GL, Ao Z, Park YJ, Safikhan N, Ghahary A, Marzban L. Amyloid formation reduces protein kinase B phosphorylation in primary islet β-cells which is improved by blocking IL-1β signaling. PLoS One 2018; 13:e0193184. [PMID: 29474443 PMCID: PMC5825069 DOI: 10.1371/journal.pone.0193184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 02/02/2018] [Indexed: 12/31/2022] Open
Abstract
Amyloid formation in the pancreatic islets due to aggregation of human islet amyloid polypeptide (hIAPP) contributes to reduced β-cell mass and function in type 2 diabetes (T2D) and islet transplantation. Protein kinase B (PKB) signaling plays a key role in the regulation of β-cell survival, function and proliferation. In this study, we used human and hIAPP-expressing transgenic mouse islets in culture as two ex vivo models of human islet amyloid formation to: 1. Investigate the effects of amyloid formation on PKB phosphorylation in primary islet β-cells; 2. Test if inhibition of amyloid formation and/or interleukin-1β (IL-1β) signaling in islets can restore the changes in β-cell phospho-PKB levels mediated by amyloid formation. Human and hIAPP-expressing mouse islets were cultured in elevated glucose with an amyloid inhibitor (Congo red) or embedded within collagen matrix to prevent amyloid formation. To block the IL-1β signaling, human islets were treated with an IL-1 receptor antagonist (anakinra) or a glucagon-like peptide-1 agonist (exenatide). β-cell phospho-PKB levels, proliferation, apoptosis, islet IL-1β levels and amyloid formation were assessed. Amyloid formation in both cultured human and hIAPP-expressing mouse islets reduced β-cell phospho-PKB levels and increased islet IL-1β levels, both of which were restored by prevention of amyloid formation either by the amyloid inhibitor or embedding islets in collagen matrix, resulting in improved β-cell survival. Furthermore, inhibition of IL-1β signaling by treatment with anakinra or exenatide increased β-cell phospho-PKB levels, enhanced proliferation and reduced apoptosis in amyloid forming human islets during 7-day culture. These data suggest that amyloid formation leads to reduced PKB phosphorylation in β-cells which is associated with elevated islet IL-1β levels. Inhibitors of amyloid or amyloid-induced IL-1β production may provide a new approach to restore phospho-PKB levels thereby enhance β-cell survival and proliferation in conditions associated with islet amyloid formation such as T2D and clinical islet transplantation.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Garth L. Warnock
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ziliang Ao
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yoo Jin Park
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nooshin Safikhan
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Aziz Ghahary
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lucy Marzban
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- * E-mail:
| |
Collapse
|
37
|
Denroche HC, Verchere CB. IAPP and type 1 diabetes: implications for immunity, metabolism and islet transplants. J Mol Endocrinol 2018; 60:R57-R75. [PMID: 29378867 DOI: 10.1530/jme-17-0138] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 12/06/2017] [Indexed: 01/12/2023]
Abstract
Islet amyloid polypeptide (IAPP), the main component of islet amyloid in type 2 diabetes and islet transplants, is now recognized as a contributor to beta cell dysfunction. Increasingly, evidence warrants its investigation in type 1 diabetes owing to both its immunomodulatory and metabolic actions. Autoreactive T cells to IAPP-derived epitopes have been described in humans, suggesting that IAPP is an islet autoantigen in type 1 diabetes. In addition, although aggregates of IAPP have not been implicated in type 1 diabetes, they are potent pro-inflammatory stimuli to innate immune cells, and thus, could influence autoimmunity. IAPP aggregates also occur rapidly in transplanted islets and likely contribute to islet transplant failure in type 1 diabetes through sterile inflammation. In addition, since type 1 diabetes is a disease of both insulin and IAPP deficiency, clinical trials have examined the potential benefits of IAPP replacement in type 1 diabetes with the injectable IAPP analogue, pramlintide. Pramlintide limits postprandial hyperglycemia by delaying gastric emptying and suppressing hyperglucagonemia, underlining the possible role of IAPP in postprandial glucose metabolism. Here, we review IAPP in the context of type 1 diabetes: from its potential involvement in type 1 diabetes pathogenesis, through its role in glucose metabolism and use of IAPP analogues as therapeutics, to its potential role in clinical islet transplant failure and considerations in this regard for future beta cell replacement strategies.
Collapse
Affiliation(s)
- Heather C Denroche
- Department of Surgery, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Bruce Verchere
- Department of Surgery, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
38
|
Kim J, Lim YM, Lee MS. The Role of Autophagy in Systemic Metabolism and Human-Type Diabetes. Mol Cells 2018; 41:11-17. [PMID: 29370692 PMCID: PMC5792707 DOI: 10.14348/molcells.2018.2228] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/11/2017] [Accepted: 10/12/2017] [Indexed: 12/18/2022] Open
Abstract
Autophagy is critical for the maintenance of organelle function and intracellular nutrient environment. Autophagy is also involved in systemic metabolic homeostasis, and its dysregulation can lead to or accelerate the development of metabolic disorders. While the role of autophagy in the global metabolism of model organisms has been investigated mostly using site-specific genetic knockout technology, the impact of dysregulated autophagy on systemic metabolism has been unclear. Here, we review recent papers showing the role of autophagy in systemic metabolism and in the development of metabolic disorders. Also included are data suggesting the role of autophagy in human-type diabetes, which are different in several key aspects from murine models of diabetes. The results shown here support the view that autophagy modulation could be a new modality for the treatment of metabolic syndrome associated with lipid overload and human-type diabetes.
Collapse
Affiliation(s)
- Jinyoung Kim
- Severance Biomedical Science Institute & Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Yu-Mi Lim
- Severance Biomedical Science Institute & Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722,
Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute & Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722,
Korea
| |
Collapse
|
39
|
Epigallocatechin gallate (EGCG) reduces the intensity of pancreatic amyloid fibrils in human islet amyloid polypeptide (hIAPP) transgenic mice. Sci Rep 2018; 8:1116. [PMID: 29348618 PMCID: PMC5773570 DOI: 10.1038/s41598-017-18807-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/15/2017] [Indexed: 01/27/2023] Open
Abstract
The formation of amyloid fibrils by human islet amyloid polypeptide protein (hIAPP) has been implicated in pancreas dysfunction and diabetes. However, efficient treatment options to reduce amyloid fibrils in vivo are still lacking. Therefore, we tested the effect of epigallocatechin gallate (EGCG) on fibril formation in vitro and in vivo. To determine the binding of hIAPP and EGCG, in vitro interaction studies were performed. To inhibit amyloid plaque formation in vivo, homozygous (tg/tg), hemizygous (wt/tg), and control mice (wt/wt) were treated with EGCG. EGCG bound to hIAPP in vitro and induced formation of amorphous aggregates instead of amyloid fibrils. Amyloid fibrils were detected in the pancreatic islets of tg/tg mice, which was associated with disrupted islet structure and diabetes. Although pancreatic amyloid fibrils could be detected in wt/tg mice, these animals were non-diabetic. EGCG application decreased amyloid fibril intensity in wt/tg mice, however it was ineffective in tg/tg animals. Our data indicate that EGCG inhibits amyloid fibril formation in vitro and reduces fibril intensity in non-diabetic wt/tg mice. These results demonstrate a possible in vivo effectiveness of EGCG on amyloid formation and suggest an early therapeutical application.
Collapse
|
40
|
Ullsten S, Bohman S, Oskarsson ME, Nilsson KPR, Westermark GT, Carlsson PO. Islet amyloid deposits preferentially in the highly functional and most blood-perfused islets. Endocr Connect 2017; 6:458-468. [PMID: 28790139 PMCID: PMC5574281 DOI: 10.1530/ec-17-0148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/08/2017] [Indexed: 01/13/2023]
Abstract
Islet amyloid and beta cell death in type 2 diabetes are heterogeneous events, where some islets are affected early in the disease process, whereas others remain visibly unaffected. This study investigated the possibility that inter-islet functional and vascular differences may explain the propensity for amyloid accumulation in certain islets. Highly blood-perfused islets were identified by microspheres in human islet amyloid polypeptide expressing mice fed a high-fat diet for three or 10 months. These highly blood-perfused islets had better glucose-stimulated insulin secretion capacity than other islets and developed more amyloid deposits after 10 months of high-fat diet. Similarly, human islets with a superior release capacity formed more amyloid in high glucose culture than islets with a lower release capacity. The amyloid formation in mouse islets was associated with a higher amount of prohormone convertase 1/3 and with a decreased expression of its inhibitor proSAAS when compared to islets with less amyloid. In contrast, levels of prohormone convertase 2 and expression of its inhibitor neuroendocrine protein 7B2 were unaltered. A misbalance in prohormone convertase levels may interrupt the normal processing of islet amyloid polypeptide and induce amyloid formation. Preferential amyloid load in the most blood-perfused and functional islets may accelerate the progression of type 2 diabetes.
Collapse
Affiliation(s)
- Sara Ullsten
- Department of Medical Cell BiologyUppsala University, Uppsala, Sweden
| | - Sara Bohman
- Department of Medical Cell BiologyUppsala University, Uppsala, Sweden
| | - Marie E Oskarsson
- Department of Medical Cell BiologyUppsala University, Uppsala, Sweden
| | | | | | - Per-Ola Carlsson
- Department of Medical Cell BiologyUppsala University, Uppsala, Sweden
- Department of Medical SciencesUppsala University, Uppsala, Sweden
| |
Collapse
|
41
|
Templin AT, Samarasekera T, Meier DT, Hogan MF, Mellati M, Crow MT, Kitsis RN, Zraika S, Hull RL, Kahn SE. Apoptosis Repressor With Caspase Recruitment Domain Ameliorates Amyloid-Induced β-Cell Apoptosis and JNK Pathway Activation. Diabetes 2017; 66:2636-2645. [PMID: 28729244 PMCID: PMC5606321 DOI: 10.2337/db16-1352] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 07/13/2017] [Indexed: 12/28/2022]
Abstract
Islet amyloid is present in more than 90% of individuals with type 2 diabetes, where it contributes to β-cell apoptosis and insufficient insulin secretion. Apoptosis repressor with caspase recruitment domain (ARC) binds and inactivates components of the intrinsic and extrinsic apoptosis pathways and was recently found to be expressed in islet β-cells. Using a human islet amyloid polypeptide transgenic mouse model of islet amyloidosis, we show ARC knockdown increases amyloid-induced β-cell apoptosis and loss, while ARC overexpression decreases amyloid-induced apoptosis, thus preserving β-cells. These effects occurred in the absence of changes in islet amyloid deposition, indicating ARC acts downstream of amyloid formation. Because islet amyloid increases c-Jun N-terminal kinase (JNK) pathway activation, we investigated whether ARC affects JNK signaling in amyloid-forming islets. We found ARC knockdown enhances JNK pathway activation, whereas ARC overexpression reduces JNK, c-Jun phosphorylation, and c-Jun target gene expression (Jun and Tnf). Immunoprecipitation of ARC from mouse islet lysates showed ARC binds JNK, suggesting interaction between JNK and ARC decreases amyloid-induced JNK phosphorylation and downstream signaling. These data indicate that ARC overexpression diminishes amyloid-induced JNK pathway activation and apoptosis in the β-cell, a strategy that may reduce β-cell loss in type 2 diabetes.
Collapse
Affiliation(s)
- Andrew T Templin
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Tanya Samarasekera
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Daniel T Meier
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Meghan F Hogan
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Mahnaz Mellati
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Michael T Crow
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY
| | - Sakeneh Zraika
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Rebecca L Hull
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| | - Steven E Kahn
- VA Puget Sound Health Care System and Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
42
|
Montane J, de Pablo S, Castaño C, Rodríguez-Comas J, Cadavez L, Obach M, Visa M, Alcarraz-Vizán G, Sanchez-Martinez M, Nonell-Canals A, Parrizas M, Servitja JM, Novials A. Amyloid-induced β-cell dysfunction and islet inflammation are ameliorated by 4-phenylbutyrate (PBA) treatment. FASEB J 2017; 31:5296-5306. [PMID: 28821639 DOI: 10.1096/fj.201700236r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/25/2017] [Indexed: 12/28/2022]
Abstract
Human islet amyloid polypeptide (hIAPP) aggregation is associated with β-cell dysfunction and death in type 2 diabetes (T2D). we aimed to determine whether in vivo treatment with chemical chaperone 4-phenylbutyrate (PBA) ameliorates hIAPP-induced β-cell dysfunction and islet amyloid formation. Oral administration of PBA in hIAPP transgenic (hIAPP Tg) mice expressing hIAPP in pancreatic β cells counteracted impaired glucose homeostasis and restored glucose-stimulated insulin secretion. Moreover, PBA treatment almost completely prevented the transcriptomic alterations observed in hIAPP Tg islets, including the induction of genes related to inflammation. PBA also increased β-cell viability and improved insulin secretion in hIAPP Tg islets cultured under glucolipotoxic conditions. Strikingly, PBA not only prevented but even reversed islet amyloid deposition, pointing to a direct effect of PBA on hIAPP. This was supported by in silico calculations uncovering potential binding sites of PBA to monomeric, dimeric, and pentameric fibrillar structures, and by in vitro assays showing inhibition of hIAPP fibril formation by PBA. Collectively, these results uncover a novel beneficial effect of PBA on glucose homeostasis by restoring β-cell function and preventing amyloid formation in mice expressing hIAPP in β cells, highlighting the therapeutic potential of PBA for the treatment of T2D.-Montane, J., de Pablo, S., Castaño, C., Rodríguez-Comas, J., Cadavez, L., Obach, M., Visa, M., Alcarraz-Vizán, G., Sanchez-Martinez, M., Nonell-Canals, A., Parrizas, M., Servitja, J.-M., Novials, A. Amyloid-induced β-cell dysfunction and islet inflammation are ameliorated by 4-phenylbutyrate (PBA) treatment.
Collapse
Affiliation(s)
- Joel Montane
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain; .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Sara de Pablo
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Carlos Castaño
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Júlia Rodríguez-Comas
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Lisa Cadavez
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mercè Obach
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Montse Visa
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Gema Alcarraz-Vizán
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | | | - Marcelina Parrizas
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Joan-Marc Servitja
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Barcelona, Spain; .,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
43
|
Akter R, Abedini A, Ridgway Z, Zhang X, Kleinberg J, Schmidt AM, Raleigh DP. Evolutionary Adaptation and Amyloid Formation: Does the Reduced Amyloidogenicity and Cytotoxicity of Ursine Amylin Contribute to the Metabolic Adaption of Bears and Polar Bears? Isr J Chem 2017; 57:750-761. [PMID: 29955200 PMCID: PMC6018008 DOI: 10.1002/ijch.201600081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Much of our knowledge of diabetes is derived from studies of rodent models. An alternative approach explores evolutionary solutions to physiological stress by studying organisms that face challenging metabolic environments. Polar bears eat an enormously lipid-rich diet without deleterious metabolic consequences. In contrast, transgenic rodents expressing the human neuropancreatic polypeptide hormone amylin develop hyperglycemia and extensive pancreatic islet amyloid when fed a high fat diet. The process of islet amyloid formation by human amylin contributes to β-cell dysfunction and loss of β-cell mass in type-2 diabetes. We show that ursine amylin is considerably less amyloidogenic and less toxic to β-cells than human amylin, consistent with the hypothesis that part of the adaptation of bears to metabolic challenges might include protection from islet amyloidosis-induced β-cell toxicity. Ursine and human amylin differ at four locations: H18R, S20G, F23L, and S29P. These are interesting from a biophysical perspective since the S20G mutation accelerates amyloid formation but the H18R slows it. An H18RS20G double mutant of human amylin behaves similarly to the H18R mutant, indicating that the substitution at position 18 dominates the S20G replacement. These data suggest one possible mechanism underpinning the protection of bears against metabolic challenges and provide insight into the design of soluble analogs of human amylin.
Collapse
Affiliation(s)
- Rehana Akter
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400
| | - Andisheh Abedini
- Diabetes Research Program, NYU School of Medicine, 522 First Avenue, New York, NY 10016
| | - Zachary Ridgway
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400
| | - Xiaoxue Zhang
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400
| | - Joel Kleinberg
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400
| | - Ann Marie Schmidt
- Diabetes Research Program, NYU School of Medicine, 522 First Avenue, New York, NY 10016
| | - Daniel P. Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400
| |
Collapse
|
44
|
Zhang Y, Song W. Islet amyloid polypeptide: Another key molecule in Alzheimer's pathogenesis? Prog Neurobiol 2017; 153:100-120. [PMID: 28274676 DOI: 10.1016/j.pneurobio.2017.03.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 02/17/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
Recent epidemiological evidence reveals that patients suffering from type 2 diabetes mellitus (T2DM) often experience a significant decline in cognitive function, and approximately 70% of those cases eventually develop Alzheimer's disease (AD). Although several pathological processes are shared by AD and T2DM, the exact molecular mechanisms connecting these two diseases are poorly understood. Aggregation of human islet amyloid polypeptide (hIAPP), the pathological hallmark of T2DM, has also been detected in brain tissue and is associated with cognitive decline and AD development. In addition, hIAPP and amyloid β protein (Aβ) share many biophysical and physiological properties as well as exert similar cytotoxic mechanisms. Therefore, it is important to examine the possible role of hIAPP in the pathogenesis of AD. In this article, we introduce the basics on this amyloidogenic protein. More importantly, we discuss the potential mechanisms of hIAPP-induced AD development, which will be beneficial for proposing novel and feasible strategies to optimize AD prevention and/or treatment in diabetics.
Collapse
Affiliation(s)
- Yun Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
45
|
Krotee P, Rodriguez JA, Sawaya MR, Cascio D, Reyes FE, Shi D, Hattne J, Nannenga BL, Oskarsson ME, Philipp S, Griner S, Jiang L, Glabe CG, Westermark GT, Gonen T, Eisenberg DS. Atomic structures of fibrillar segments of hIAPP suggest tightly mated β-sheets are important for cytotoxicity. eLife 2017; 6. [PMID: 28045370 PMCID: PMC5207774 DOI: 10.7554/elife.19273] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/01/2016] [Indexed: 01/09/2023] Open
Abstract
hIAPP fibrils are associated with Type-II Diabetes, but the link of hIAPP structure to islet cell death remains elusive. Here we observe that hIAPP fibrils are cytotoxic to cultured pancreatic β-cells, leading us to determine the structure and cytotoxicity of protein segments composing the amyloid spine of hIAPP. Using the cryoEM method MicroED, we discover that one segment, 19-29 S20G, forms pairs of β-sheets mated by a dry interface that share structural features with and are similarly cytotoxic to full-length hIAPP fibrils. In contrast, a second segment, 15-25 WT, forms non-toxic labile β-sheets. These segments possess different structures and cytotoxic effects, however, both can seed full-length hIAPP, and cause hIAPP to take on the cytotoxic and structural features of that segment. These results suggest that protein segment structures represent polymorphs of their parent protein and that segment 19-29 S20G may serve as a model for the toxic spine of hIAPP.
Collapse
Affiliation(s)
- Pascal Krotee
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| | - Jose A Rodriguez
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| | - Michael R Sawaya
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| | - Duilio Cascio
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| | - Francis E Reyes
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Dan Shi
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Johan Hattne
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Brent L Nannenga
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Marie E Oskarsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Stephan Philipp
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, United States
| | - Sarah Griner
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| | - Lin Jiang
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Brain Research Institute (BRI), University of California, Los Angeles, Los Angeles, United States
| | - Charles G Glabe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, United States.,Biochemistry Department, Faculty of Science and Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Tamir Gonen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - David S Eisenberg
- Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, United States.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, United States.,UCLA-DOE Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
46
|
Grizzanti J, Lee HG, Camins A, Pallas M, Casadesus G. The therapeutic potential of metabolic hormones in the treatment of age-related cognitive decline and Alzheimer's disease. Nutr Res 2016; 36:1305-1315. [PMID: 27923524 DOI: 10.1016/j.nutres.2016.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/17/2016] [Accepted: 11/03/2016] [Indexed: 01/04/2023]
Abstract
Aging leads to a number of physiological alterations, specifically changes in circulating hormone levels, increases in fat deposition, decreases in metabolism, changes in inflammatory responses, and reductions in growth factors. These progressive changes in physiology and metabolism are exacerbated by modern culture and Western diet and give rise to diseases such as obesity, metabolic syndrome, and type 2 (non-insulin-dependent) diabetes (T2D). These age and lifestyle-related metabolic diseases are often accompanied by insulin and leptin resistance, as well as aberrant amylin production and signaling. Many of these alterations in hormone production and signaling are directly influenced by an increase in both oxidative stress and inflammation. Importantly, changes in hormone production and signaling have direct effects on brain function and the development of age-related neurologic disorders. Therefore, this review aims to present evidence on the effects that diet and metabolic disease have on age-related cognitive decline and the development of cognitive diseases, particularly Alzheimer disease. This review will focus on the metabolic hormones insulin, leptin, and amylin and their role in cognitive decline, as well as the therapeutic potential of these hormones in treating cognitive disease. Future investigations targeting the long-term effects of insulin and leptin treatment may reveal evidence to reduce risk of cognitive decline and Alzheimer disease.
Collapse
Affiliation(s)
- John Grizzanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Hyoung-Gon Lee
- Department of Biology, University of Texas, San Antonio, TX, USA
| | - Antoni Camins
- Department of Pharmacology and Therapeutic Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Merce Pallas
- Department of Pharmacology and Therapeutic Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Casadesus
- School of Biomedical Sciences, Kent State University, Kent, OH, USA; Department of Biological Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
47
|
Hogan MF, Meier DT, Zraika S, Templin AT, Mellati M, Hull RL, Leissring MA, Kahn SE. Inhibition of Insulin-Degrading Enzyme Does Not Increase Islet Amyloid Deposition in Vitro. Endocrinology 2016; 157:3462-8. [PMID: 27404391 PMCID: PMC5007890 DOI: 10.1210/en.2016-1410] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Islet amyloid deposition in human type 2 diabetes results in β-cell loss. These amyloid deposits contain the unique amyloidogenic peptide human islet amyloid polypeptide (hIAPP), which is also a known substrate of the protease insulin-degrading enzyme (IDE). Whereas IDE inhibition has recently been demonstrated to improve glucose metabolism in mice, inhibiting it has also been shown to increase cell death when synthetic hIAPP is applied exogenously to a β-cell line. Thus, we wanted to determine whether a similar deleterious effect is observed when hIAPP is endogenously produced and secreted from islets. To address this issue, we cultured hIAPP transgenic mouse islets that have the propensity to form amyloid for 48 and 144 hours in 16.7 mM glucose in the presence and absence of the IDE inhibitor 1. At neither time interval did IDE inhibition increase amyloid formation or β-cell loss. Thus, the inhibition of IDE may represent an approach to improve glucose metabolism in human type 2 diabetes, without inducing amyloid deposition and its deleterious effects.
Collapse
Affiliation(s)
- Meghan F Hogan
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Daniel T Meier
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Andrew T Templin
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Mahnaz Mellati
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Rebecca L Hull
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Malcolm A Leissring
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| | - Steven E Kahn
- Division of Metabolism, Endocrinology, and Nutrition (M.F.H., D.T.M., S.Z., A.T.T., M.M., R.L.H., S.E.K.), Department of Medicine, Veterans Affairs Puget Sound Health Care System and University of Washington, Seattle, Washington 98108; and Institute for Memory Impairments and Neurological Disorders (M.A.L.), University of California, Irvine, Irvine, California 92697
| |
Collapse
|
48
|
Proteasome regulates turnover of toxic human amylin in pancreatic cells. Biochem J 2016; 473:2655-70. [PMID: 27340132 DOI: 10.1042/bcj20160026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/22/2016] [Indexed: 12/18/2022]
Abstract
Toxic human amylin (hA) oligomers and aggregates are implicated in the pathogenesis of type 2 diabetes mellitus (T2DM). Although recent studies demonstrated a causal connection between hA uptake and toxicity in pancreatic cells, the mechanism of amylin's clearance following its internalization and its relationship to toxicity is yet to be determined, and hence was investigated here. Using pancreatic rat insulinoma β-cells and human islets as model systems, we show that hA, following its internalization, first accumulates in the cytosol followed by its translocation into nucleus, and to a lesser extent lysosomes, keeping the net cytosolic amylin content low. An increase in hA accumulation in the nucleus of pancreatic cells correlated with its cytotoxicity, suggesting that its excessive accumulation in the nucleus is detrimental. hA interacted with 20S core and 19S lid subunits of the β-cell proteasomal complex, as suggested by immunoprecipitation and confocal microscopy studies, which subsequently resulted in a decrease in the proteasome's proteolytic activity in these cells. In vitro binding and activity assays confirmed an intrinsic and potent ability of amylin to interact with the 20S core complex thereby modulating its proteolytic activity. Interestingly, less toxic and aggregation incapable rat amylin (rA) showed a comparable inhibitory effect on proteasome activity and protein ubiquitination, decoupling amylin aggregation/ toxicity and amylin-induced protein stress. In agreement with these studies, inhibition of proteasomal proteolytic activity significantly increased intracellular amylin content and toxicity. Taken together, our results suggest a pivotal role of proteasomes in amylin's turnover and detoxification in pancreatic cells.
Collapse
|
49
|
Ribarič S. The Rationale for Insulin Therapy in Alzheimer's Disease. Molecules 2016; 21:molecules21060689. [PMID: 27240327 PMCID: PMC6273626 DOI: 10.3390/molecules21060689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a prevalence that increases with age. By 2050, the worldwide number of patients with AD is projected to reach more than 140 million. The prominent signs of AD are progressive memory loss, accompanied by a gradual decline in cognitive function and premature death. AD is the clinical manifestation of altered proteostasis. The initiating step of altered proteostasis in most AD patients is not known. The progression of AD is accelerated by several chronic disorders, among which the contribution of diabetes to AD is well understood at the cell biology level. The pathological mechanisms of AD and diabetes interact and tend to reinforce each other, thus accelerating cognitive impairment. At present, only symptomatic interventions are available for treating AD. To optimise symptomatic treatment, a personalised therapy approach has been suggested. Intranasal insulin administration seems to open the possibility for a safe, and at least in the short term, effective symptomatic intervention that delays loss of cognition in AD patients. This review summarizes the interactions of AD and diabetes from the cell biology to the patient level and the clinical results of intranasal insulin treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
50
|
Yoshimura M, Ono M, Watanabe H, Kimura H, Saji H. Development of 99mTc-Labeled Pyridyl Benzofuran Derivatives To Detect Pancreatic Amylin in Islet Amyloid Model Mice. Bioconjug Chem 2016; 27:1532-9. [DOI: 10.1021/acs.bioconjchem.6b00174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Masashi Yoshimura
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Ono
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Kimura
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hideo Saji
- Department
of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|