1
|
Kemeh MM, Lazo ND. Modulation of the Activity of the Insulin-Degrading Enzyme by Aβ Peptides. ACS Chem Neurosci 2023; 14:2935-2943. [PMID: 37498802 DOI: 10.1021/acschemneuro.3c00384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
The insulin-degrading enzyme (IDE) is an evolutionarily conserved protease implicated in the degradation of insulin and amyloidogenic peptides. Most of the biochemical and biophysical characterization of IDE's catalytic activity has been conducted using solutions containing a single substrate, i.e., insulin or Aβ(1-40). IDE's activity toward a particular substrate, however, is likely to be influenced by the presence of other substrates. Here, we show by a kinetic assay based on insulin's helical circular dichroic signal and MALDI TOF mass spectrometry that Aβ peptides modulate IDE's activity toward insulin in opposing ways. Aβ(1-40) enhances IDE-dependent degradation of insulin, whereas Aβ(pyroE3-42), the most pathogenic pyroglutamate-modified Aβ peptide in AD, inhibits IDE's activity. Intriguingly, Aβ(pyroE3-42) also inhibits IDE's ability to degrade Aβ(1-40). Together, our results implicate Aβ peptides in the abnormal catabolism of IDE's key substrates.
Collapse
Affiliation(s)
- Merc M Kemeh
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Noel D Lazo
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| |
Collapse
|
2
|
Zheng Q, Lee B, Kebede MT, Ivancic VA, Kemeh MM, Brito HL, Spratt DE, Lazo ND. Exchange Broadening Underlies the Enhancement of IDE-Dependent Degradation of Insulin by Anionic Membranes. ACS OMEGA 2022; 7:24757-24765. [PMID: 35874268 PMCID: PMC9301717 DOI: 10.1021/acsomega.2c02747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Insulin-degrading enzyme (IDE) is an evolutionarily conserved ubiquitous zinc metalloprotease implicated in the efficient degradation of insulin monomer. However, IDE also degrades monomers of amyloidogenic peptides associated with disease, complicating the development of IDE inhibitors. In this work, we investigated the effects of the lipid composition of membranes on the IDE-dependent degradation of insulin. Kinetic analysis based on chromatography and insulin's helical circular dichroic signal showed that the presence of anionic lipids in membranes enhances IDE's activity toward insulin. Using NMR spectroscopy, we discovered that exchange broadening underlies the enhancement of IDE's activity. These findings, together with the adverse effects of anionic membranes in the self-assembly of IDE's amyloidogenic substrates, suggest that the lipid composition of membranes is a key determinant of IDE's ability to balance the levels of its physiologically and pathologically relevant substrates and achieve proteostasis.
Collapse
Affiliation(s)
| | | | | | - Valerie A. Ivancic
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Merc M. Kemeh
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Henrique Lemos Brito
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Donald E. Spratt
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| | - Noel D. Lazo
- Gustaf H. Carlson School
of Chemistry and Biochemistry, Clark University, Worcester, Massachusetts 01610, United States
| |
Collapse
|
3
|
Lesire L, Leroux F, Deprez-Poulain R, Deprez B. Insulin-Degrading Enzyme, an Under-Estimated Potential Target to Treat Cancer? Cells 2022; 11:1228. [PMID: 35406791 PMCID: PMC8998118 DOI: 10.3390/cells11071228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
Insulin-degrading enzyme (IDE) is a multifunctional protease due to the variety of its substrates, its various cellular locations, its conservation between species and its many non-proteolytic functions. Numerous studies have successfully demonstrated its implication in two main therapeutic areas: metabolic and neuronal diseases. In recent years, several reports have underlined the overexpression of this enzyme in different cancers. Still, the exact role of IDE in the physiopathology of cancer remains to be elucidated. Known as the main enzyme responsible for the degradation of insulin, an essential growth factor for healthy cells and cancer cells, IDE has also been shown to behave like a chaperone and interact with the proteasome. The pharmacological modulation of IDE (siRNA, chemical compounds, etc.) has demonstrated interesting results in cancer models. All these results point towards IDE as a potential target in cancer. In this review, we will discuss evidence of links between IDE and cancer development or resistance, IDE's functions, catalytic or non-catalytic, in the context of cell proliferation, cancer development and the impact of the pharmacomodulation of IDE via cancer therapeutics.
Collapse
Affiliation(s)
| | | | - Rebecca Deprez-Poulain
- INSERM U1177 Drugs and Molecules for Living Systems, Institut Pasteur de Lille, European Genomic Institute for Diabetes, University of Lille, F-59000 Lille, France; (L.L.); (F.L.); (B.D.)
| | | |
Collapse
|
4
|
Ghoula M, Janel N, Camproux AC, Moroy G. Exploring the Structural Rearrangements of the Human Insulin-Degrading Enzyme through Molecular Dynamics Simulations. Int J Mol Sci 2022; 23:ijms23031746. [PMID: 35163673 PMCID: PMC8836115 DOI: 10.3390/ijms23031746] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/24/2022] [Accepted: 01/29/2022] [Indexed: 11/16/2022] Open
Abstract
Insulin-degrading enzyme (IDE) is a ubiquitously expressed metallopeptidase that degrades insulin and a large panel of amyloidogenic peptides. IDE is thought to be a potential therapeutic target for type-2 diabetes and neurodegenerative diseases, such as Alzheimer’s disease. IDE catalytic chamber, known as a crypt, is formed, so that peptides can be enclosed and degraded. However, the molecular mechanism of the IDE function and peptide recognition, as well as its conformation changes, remains elusive. Our study elucidates IDE structural changes and explains how IDE conformational dynamics is important to modulate the catalytic cycle of IDE. In this aim, a free-substrate IDE crystallographic structure (PDB ID: 2JG4) was used to model a complete structure of IDE. IDE stability and flexibility were studied through molecular dynamics (MD) simulations to witness IDE conformational dynamics switching from a closed to an open state. The description of IDE structural changes was achieved by analysis of the cavity and its expansion over time. Moreover, the quasi-harmonic analysis of the hinge connecting IDE domains and the angles formed over the simulations gave more insights into IDE shifts. Overall, our results could guide toward the use of different approaches to study IDE with different substrates and inhibitors, while taking into account the conformational states resolved in our study.
Collapse
Affiliation(s)
- Mariem Ghoula
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, INSERM, Université de Paris, F-75013 Paris, France;
| | - Nathalie Janel
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, Université de Paris, F-75013 Paris, France;
| | - Anne-Claude Camproux
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, INSERM, Université de Paris, F-75013 Paris, France;
- Correspondence: (A.-C.C.); (G.M.); Tel.: +33-1-57-27-83-77 (A.-C.C.); +33-1-57-27-83-85 (G.M.)
| | - Gautier Moroy
- Unité de Biologie Fonctionnelle et Adaptative, CNRS, INSERM, Université de Paris, F-75013 Paris, France;
- Correspondence: (A.-C.C.); (G.M.); Tel.: +33-1-57-27-83-77 (A.-C.C.); +33-1-57-27-83-85 (G.M.)
| |
Collapse
|
5
|
The Structures and Functions of VZV Glycoproteins. Curr Top Microbiol Immunol 2021; 438:25-58. [PMID: 34731265 DOI: 10.1007/82_2021_243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The virions of all enveloped viruses, including those of the Herpesviridae, must bind to the cell surface then undergo a process of membrane fusion between the cell plasma membrane and the virus particle envelope. As for all herpesviruses, glycoproteins in the virion envelope are the modus operandi of these events.
Collapse
|
6
|
Leissring MA. Insulin-Degrading Enzyme: Paradoxes and Possibilities. Cells 2021; 10:cells10092445. [PMID: 34572094 PMCID: PMC8472535 DOI: 10.3390/cells10092445] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/31/2022] Open
Abstract
More than seven decades have passed since the discovery of a proteolytic activity within crude tissue extracts that would become known as insulin-degrading enzyme (IDE). Certainly much has been learned about this atypical zinc-metallopeptidase; at the same time, however, many quite fundamental gaps in our understanding remain. Herein, I outline what I consider to be among the most critical unresolved questions within the field, many presenting as intriguing paradoxes. For instance, where does IDE, a predominantly cytosolic protein with no signal peptide or clearly identified secretion mechanism, interact with insulin and other extracellular substrates? Where precisely is IDE localized within the cell, and what are its functional roles in these compartments? How does IDE, a bowl-shaped protein that completely encapsulates its substrates, manage to avoid getting “clogged” and thus rendered inactive virtually immediately? Although these paradoxes are by definition unresolved, I offer herein my personal insights and informed speculations based on two decades working on the biology and pharmacology of IDE and suggest specific experimental strategies for addressing these conundrums. I also offer what I believe to be especially fruitful avenues for investigation made possible by the development of new technologies and IDE-specific reagents. It is my hope that these thoughts will contribute to continued progress elucidating the physiology and pathophysiology of this important peptidase.
Collapse
Affiliation(s)
- Malcolm A Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine (UCI MIND), Irvine, CA 92697, USA
| |
Collapse
|
7
|
Hostachy S, Utesch T, Franke K, Dornan GL, Furkert D, Türkaydin B, Haucke V, Sun H, Fiedler D. Dissecting the activation of insulin degrading enzyme by inositol pyrophosphates and their bisphosphonate analogs. Chem Sci 2021; 12:10696-10702. [PMID: 34476054 PMCID: PMC8372538 DOI: 10.1039/d1sc02975d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/06/2021] [Indexed: 11/21/2022] Open
Abstract
Inositol poly- and pyrophosphates (InsPs and PP-InsPs) are densely phosphorylated eukaryotic messengers, which are involved in numerous cellular processes. To elucidate their signaling functions at the molecular level, non-hydrolyzable bisphosphonate analogs of inositol pyrophosphates, PCP-InsPs, have been instrumental. Here, an efficient synthetic strategy to obtain these analogs in unprecedented quantities is described - relying on the use of combined phosphate ester-phosphoramidite reagents. The PCP-analogs, alongside their natural counterparts, were applied to investigate their regulatory effect on insulin-degrading enzyme (IDE), using a range of biochemical, biophysical and computational methods. A unique interplay between IDE, its substrates and the PP-InsPs was uncovered, in which the PP-InsPs differentially modulated the activity of the enzyme towards short peptide substrates. Aided by molecular docking and molecular dynamics simulations, a flexible binding mode for the InsPs/PP-InsPs was identified at the anion binding site of IDE. Targeting IDE for therapeutic purposes should thus take regulation by endogenous PP-InsP metabolites into account.
Collapse
Affiliation(s)
- Sarah Hostachy
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Tillmann Utesch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Katy Franke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Gillian Leigh Dornan
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - David Furkert
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
- Institut für Chemie, Humboldt-Universität zu Berlin Brook-Taylor-Straße 2 12489 Berlin Germany
| | - Berke Türkaydin
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Han Sun
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
| | - Dorothea Fiedler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) Robert-Rössle Str. 10 13125 Berlin Germany
- Institut für Chemie, Humboldt-Universität zu Berlin Brook-Taylor-Straße 2 12489 Berlin Germany
| |
Collapse
|
8
|
Nash Y, Ganoth A, Borenstein-Auerbach N, Levy-Barazany H, Goldsmith G, Kopelevich A, Pozyuchenko K, Sakhneny L, Lazdon E, Blanga-Kanfi S, Alhadeff R, Benromano T, Landsman L, Tsfadia Y, Frenkel D. From virus to diabetes therapy: Characterization of a specific insulin-degrading enzyme inhibitor for diabetes treatment. FASEB J 2021; 35:e21374. [PMID: 33835493 DOI: 10.1096/fj.201901945r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
Inhibition of insulin-degrading enzyme (IDE) is a possible target for treating diabetes. However, it has not yet evolved into a medical intervention, mainly because most developed inhibitors target the zinc in IDE's catalytic site, potentially causing toxicity to other essential metalloproteases. Since IDE is a cellular receptor for the varicella-zoster virus (VZV), we constructed a VZV-based inhibitor. We computationally characterized its interaction site with IDE showing that the peptide specifically binds inside IDE's central cavity, however, not in close proximity to the zinc ion. We confirmed the peptide's effective inhibition on IDE activity in vitro and showed its efficacy in ameliorating insulin-related defects in types 1 and 2 diabetes mouse models. In addition, we suggest that inhibition of IDE may ameliorate the pro-inflammatory profile of CD4+ T-cells toward insulin. Together, we propose a potential role of a designed VZV-derived peptide to serve as a selectively-targeted and as an efficient diabetes therapy.
Collapse
Affiliation(s)
- Yuval Nash
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Assaf Ganoth
- The Interdisciplinary Center (IDC), Herzliya, Israel.,Department of Physical Therapy, School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nofit Borenstein-Auerbach
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Hilit Levy-Barazany
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Guy Goldsmith
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adi Kopelevich
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Katia Pozyuchenko
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Lina Sakhneny
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ekaterina Lazdon
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shani Blanga-Kanfi
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Raphael Alhadeff
- Department of Chemistry, University of Southern California, Los Angeles, CA, USA
| | - Tali Benromano
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Development Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yossi Tsfadia
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
7,8-Dihydroxyflavone improves cognitive functions in ICV-STZ rat model of sporadic Alzheimer's disease by reversing oxidative stress, mitochondrial dysfunction, and insulin resistance. Psychopharmacology (Berl) 2021; 238:1991-2009. [PMID: 33774703 DOI: 10.1007/s00213-021-05826-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
RATIONALE Intracerebroventricular (ICV) streptozotocin (STZ) mimics sporadic Alzheimer's disease (SAD) characterized by tau pathology and neurodegeneration arising from oxidative stress, mitochondrial dysfunction, and insulin resistance. 7,8-Dihydroxyflavone (7,8-DHF) is a flavonoid having antioxidant property interlinked with mitochondrial functioning and insulin actions. OBJECTIVES To evaluate the neuroprotective and cognitive enhancement properties of 7,8-DHF in an ICV-STZ rat model of SAD. METHODS ICV-STZ (3 mg/kg) was injected into male Wistar rats. Cognitive functions were evaluated by Morris water maze (MWM) and novel object recognition (NOR). 7,8-DHF (5 mg/kg, 10 mg/kg, and 20 mg/kg) and rivastigmine (2 mg/kg) were given orally for 21 days. Reduced glutathione (GSH), catalase, superoxide dismutase (SOD), glutathione peroxidase (GPX), lipid peroxidation (LPO), protein carbonylation (PCO), and nitrite assays were performed. Mitochondrial enzyme complex I, II, III, and IV, and acetylcholinesterase (AchE) activities were determined. ELISA for the insulin-degrading enzyme (IDE) and p-tau was done. Histopathology was investigated by hematoxylin and eosin staining. RESULTS 7,8-DHF treatment attenuated ICV-STZ-induced cognitive deficit in MWM and NOR. Moreover, in the cortex and hippocampus regions of the brain, GSH, catalase, SOD, GPX, LPO, PCO, and nitrite levels were reversed. Mitochondrial enzyme complex I, II, III, and IV, and acetylcholinesterase (AchE) activities were also normalized. IDE and p-tau protein were found to be significantly altered. 7,8-DHF provided protection from neuronal cell death examined in histopathology. CONCLUSIONS Conclusively, 7,8-DHF was found to be neuroprotective in the ICV-STZ rat model by ameliorating oxidative stress, mitochondrial dysfunction, and insulin resistance, thereby improving cognitive functions evident with the behavioral results.
Collapse
|
10
|
Modulation of Insulin Sensitivity by Insulin-Degrading Enzyme. Biomedicines 2021; 9:biomedicines9010086. [PMID: 33477364 PMCID: PMC7830943 DOI: 10.3390/biomedicines9010086] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Insulin-degrading enzyme (IDE) is a highly conserved and ubiquitously expressed metalloprotease that degrades insulin and several other intermediate-size peptides. For many decades, IDE had been assumed to be involved primarily in hepatic insulin clearance, a key process that regulates availability of circulating insulin levels for peripheral tissues. Emerging evidence, however, suggests that IDE has several other important physiological functions relevant to glucose and insulin homeostasis, including the regulation of insulin secretion from pancreatic β-cells. Investigation of mice with tissue-specific genetic deletion of Ide in the liver and pancreatic β-cells (L-IDE-KO and B-IDE-KO mice, respectively) has revealed additional roles for IDE in the regulation of hepatic insulin action and sensitivity. In this review, we discuss current knowledge about IDE’s function as a regulator of insulin secretion and hepatic insulin sensitivity, both evaluating the classical view of IDE as an insulin protease and also exploring evidence for several non-proteolytic functions. Insulin proteostasis and insulin sensitivity have both been highlighted as targets controlling blood sugar levels in type 2 diabetes, so a clearer understanding the physiological functions of IDE in pancreas and liver could led to the development of novel therapeutics for the treatment of this disease.
Collapse
|
11
|
Xie Z, Wu H, Zhao J. Multifunctional roles of zinc in Alzheimer’s disease. Neurotoxicology 2020; 80:112-123. [DOI: 10.1016/j.neuro.2020.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
|
12
|
Bellia F, Lanza V, Ahmed IMM, Garcia-Vinuales S, Veiss E, Arizzi M, Calcagno D, Milardi D, Grasso G. Site directed mutagenesis of insulin-degrading enzyme allows singling out the molecular basis of peptidase versus E1-like activity: the role of metal ions. Metallomics 2020; 11:278-281. [PMID: 30627720 DOI: 10.1039/c8mt00288f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Four specifically designed IDE mutants have been used to unveil the molecular basis of peptidase versus E1-like activity of the enzyme. We have found that physiological concentrations of copper(ii) ions inhibit the proteolytic activity of the enzyme towards small and large substrates but have no effect on the E1-like activity of the enzyme.
Collapse
Affiliation(s)
- Francesco Bellia
- Institute of Biostructures and Bioimaging, National Research Council, Catania, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hu Q, Jayasinghe‐Arachchige VM, Sharma G, Serafim LF, Paul TJ, Prabhakar R. Mechanisms of peptide and phosphoester hydrolysis catalyzed by two promiscuous metalloenzymes (insulin degrading enzyme and glycerophosphodiesterase) and their synthetic analogues. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1466] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Qiaoyu Hu
- Department of Chemistry, University of Miami Coral Gables Florida
| | | | - Gaurav Sharma
- Department of Chemistry, University of Miami Coral Gables Florida
| | | | - Thomas J. Paul
- Department of Chemistry, University of Miami Coral Gables Florida
| | - Rajeev Prabhakar
- Department of Chemistry, University of Miami Coral Gables Florida
| |
Collapse
|
14
|
Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1949-1967. [DOI: 10.1016/j.bbadis.2018.11.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 02/06/2023]
|
15
|
Substrate-selective inhibitors that reprogram the activity of insulin-degrading enzyme. Nat Chem Biol 2019; 15:565-574. [PMID: 31086331 PMCID: PMC6551522 DOI: 10.1038/s41589-019-0271-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 03/14/2019] [Indexed: 11/08/2022]
Abstract
Enzymes that act on multiple substrates are common in biology but pose unique challenges as therapeutic targets. The metalloprotease insulin-degrading enzyme (IDE) modulates blood glucose levels by cleaving insulin, a hormone that promotes glucose clearance. However, IDE also degrades glucagon, a hormone that elevates glucose levels and opposes the effect of insulin. IDE inhibitors to treat diabetes therefore should prevent IDE-mediated insulin degradation, but not glucagon degradation, in contrast with traditional modes of enzyme inhibition. Using a high-throughput screen for non-active-site ligands, we discovered potent and highly specific small-molecule inhibitors that alter IDE’s substrate selectivity. X-ray co-crystal structures, including an IDE-ligand-glucagon ternary complex, revealed substrate-dependent interactions that enable these inhibitors to potently block insulin binding while allowing glucagon cleavage, even at saturating inhibitor concentrations. These findings suggest a path for developing IDE-targeting therapeutics, and offer a blueprint for modulating other enzymes in a substrate-selective manner to unlock their therapeutic potential.
Collapse
|
16
|
Kurauti MA, Ferreira SM, Soares GM, Vettorazzi JF, Carneiro EM, Boschero AC, Costa‐Júnior JM. Hyperinsulinemia is associated with increasing insulin secretion but not with decreasing insulin clearance in an age‐related metabolic dysfunction mice model. J Cell Physiol 2018; 234:9802-9809. [DOI: 10.1002/jcp.27667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Mirian A. Kurauti
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - Sandra M. Ferreira
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - Gabriela M. Soares
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - Jean F. Vettorazzi
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - Everardo M. Carneiro
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - Antonio C. Boschero
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| | - José M. Costa‐Júnior
- Obesity and Comorbidities Research Center Institute of Biology, University of Campinas (UNICAMP) Campinas Sao Paulo Brazil
| |
Collapse
|
17
|
Hayrabedyan S, Todorova K, Spinelli M, Barnea ER, Mueller M. The core sequence of PIF competes for insulin/amyloid β in insulin degrading enzyme: potential treatment for Alzheimer's disease. Oncotarget 2018; 9:33884-33895. [PMID: 30338033 PMCID: PMC6188057 DOI: 10.18632/oncotarget.26057] [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] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/16/2018] [Indexed: 11/25/2022] Open
Abstract
The central pathological feature of Alzheimer's disease (AD) is the sequential proteolytic processing of amyloid precursor protein (APP) to amyloid-β peptides (Aβ) agglomeration. The clearance of Aβ may be induced by the large zinc-binding protease insulin degrading enzyme (IDE). IDE is the common link between AD and Type II diabetes as insulin is an IDE target as well. Not surprisingly, the search for safe and effective drugs modulating IDE is ongoing. A new pregnancy derived peptide, PreImplantation Factor (PIF), inhibits neuro-inflammation and crosses the blood-brain-barrier. Importantly, we report that the (R3I4K5P6) core sequence of the PIF peptide modulates IDE function and results in decreased Aβ agglomeration in neuronal cells. Using bioinformatics we show that PIF binds to the IDE complex and sterically competes for the same place as insulin or Aβ. The predicted RIKP sequence and especially the specific I4 and P6 amino acids are essential for hydrophobic interactions with the IDE complex. In terms of potential AD treatment, PIF was successfully tested in neurodegenerative animal models of perinatal brain injury and experimental autoimmune encephalitis. Importantly, sPIF received a FDA Fast Track Approval and orphan drug designation for first-in-human trial in autoimmunity.
Collapse
Affiliation(s)
- Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Laboratory of Reproductive OMICs Technologies, Sofia, Bulgaria
| | - Krassimira Todorova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Laboratory of Reproductive OMICs Technologies, Sofia, Bulgaria
| | - Marialuigia Spinelli
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Eytan R. Barnea
- Society for The Investigation of Early Pregnancy (SIEP), New York, NY, USA
- BioIncept, New York, NY, USA
| | - Martin Mueller
- Department of Obstetrics and Gynecology, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Paediatrics, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
18
|
Lanza V, Bellia F, Rizzarelli E. An inorganic overview of natural Aβ fragments: Copper(II) and zinc(II)-mediated pathways. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
19
|
Stefanidis L, Fusco ND, Cooper SE, Smith-Carpenter JE, Alper BJ. Molecular Determinants of Substrate Specificity in Human Insulin-Degrading Enzyme. Biochemistry 2018; 57:4903-4914. [PMID: 30004674 DOI: 10.1021/acs.biochem.8b00474] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Insulin-degrading enzyme (IDE) is a 110 kDa chambered zinc metalloendopeptidase that degrades insulin, amyloid β, and other intermediate-sized aggregation prone peptides that adopt β-structures. Structural studies of IDE in complex with multiple physiological substrates have suggested a role for hydrophobic and aromatic residues of the IDE active site in substrate binding and catalysis. Here, we examine functional requirements for conserved hydrophobic and aromatic IDE active site residues that are positioned within 4.5 Å of IDE-bound insulin B chain and amyloid β peptides in the reported crystal structures for the respective enzyme-substrate complexes. Charge, size, hydrophobicity, aromaticity, and other functional group requirements for substrate binding IDE active site residues were examined through mutational analysis of the recombinant human enzyme and enzyme kinetic studies conducted using native and fluorogenic derivatives of human insulin and amyloid β peptides. A functional requirement for IDE active site residues F115, A140, F141, Y150, W199, F202, F820, and Y831 was established, and specific contributions of residue charge, size, and hydrophobicity to substrate binding, specificity, and proteolysis were demonstrated. IDE mutant alleles that exhibited enhanced or diminished proteolytic activity toward insulin or amyloid β peptides and derivative substrates were identified.
Collapse
Affiliation(s)
- Lazaros Stefanidis
- Department of Chemistry , Sacred Heart University , Fairfield , Connecticut 06825 , United States
| | - Nicholas D Fusco
- Department of Chemistry , Sacred Heart University , Fairfield , Connecticut 06825 , United States
| | - Samantha E Cooper
- Department of Chemistry and Biochemistry , Fairfield University , Fairfield , Connecticut 06824 , United States
| | - Jillian E Smith-Carpenter
- Department of Chemistry and Biochemistry , Fairfield University , Fairfield , Connecticut 06824 , United States
| | - Benjamin J Alper
- Department of Chemistry , Sacred Heart University , Fairfield , Connecticut 06825 , United States
| |
Collapse
|
20
|
Zhang Z, Liang WG, Bailey LJ, Tan YZ, Wei H, Wang A, Farcasanu M, Woods VA, McCord LA, Lee D, Shang W, Deprez-Poulain R, Deprez B, Liu DR, Koide A, Koide S, Kossiakoff AA, Li S, Carragher B, Potter CS, Tang WJ. Ensemble cryoEM elucidates the mechanism of insulin capture and degradation by human insulin degrading enzyme. eLife 2018; 7:33572. [PMID: 29596046 PMCID: PMC5910022 DOI: 10.7554/elife.33572] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/28/2018] [Indexed: 11/29/2022] Open
Abstract
Insulin degrading enzyme (IDE) plays key roles in degrading peptides vital in type two diabetes, Alzheimer's, inflammation, and other human diseases. However, the process through which IDE recognizes peptides that tend to form amyloid fibrils remained unsolved. We used cryoEM to understand both the apo- and insulin-bound dimeric IDE states, revealing that IDE displays a large opening between the homologous ~55 kDa N- and C-terminal halves to allow selective substrate capture based on size and charge complementarity. We also used cryoEM, X-ray crystallography, SAXS, and HDX-MS to elucidate the molecular basis of how amyloidogenic peptides stabilize the disordered IDE catalytic cleft, thereby inducing selective degradation by substrate-assisted catalysis. Furthermore, our insulin-bound IDE structures explain how IDE processively degrades insulin by stochastically cutting either chain without breaking disulfide bonds. Together, our studies provide a mechanism for how IDE selectively degrades amyloidogenic peptides and offers structural insights for developing IDE-based therapies.
Collapse
Affiliation(s)
- Zhening Zhang
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, United States
| | - Wenguang G Liang
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, United States
| | - Lucas J Bailey
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, United States
| | - Yong Zi Tan
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Hui Wei
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, United States
| | - Andrew Wang
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, United States
| | - Mara Farcasanu
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, United States
| | - Virgil A Woods
- Department of Medicine, University of California, San Diego, La Jolla, United States
| | - Lauren A McCord
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, United States
| | - David Lee
- Department of Medicine, University of California, San Diego, La Jolla, United States
| | - Weifeng Shang
- BioCAT, Argonne National Laboratory, Illinois, United States
| | | | - Benoit Deprez
- Univ. Lille, INSERM, Institut Pasteur de Lille, Lille, France
| | - David R Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, United States
| | - Akiko Koide
- Perlmutter Cancer Center, New York University School of Medicine, New York, United States.,New York University Langone Medical Center, New York University School of Medicine, New York, United States.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Shohei Koide
- Perlmutter Cancer Center, New York University School of Medicine, New York, United States.,New York University Langone Medical Center, New York University School of Medicine, New York, United States.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, United States
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, United States
| | - Sheng Li
- Department of Medicine, University of California, San Diego, La Jolla, United States
| | - Bridget Carragher
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Clinton S Potter
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States
| | - Wei-Jen Tang
- Ben-May Institute for Cancer Research, The University of Chicago, Chicago, United States
| |
Collapse
|
21
|
Naletova I, Nicoletti VG, Milardi D, Pietropaolo A, Grasso G. Copper, differently from zinc, affects the conformation, oligomerization state and activity of bradykinin. Metallomics 2017; 8:750-61. [PMID: 27328010 DOI: 10.1039/c6mt00067c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sole role of bradykinin (BK) as an inflammatory mediator is controversial, as recent data also support an anti-inflammatory role for BK in Alzheimer's disease (AD). The involvement of two different receptors (B1R and B2R) could be a key to understand this issue. However, although copper and zinc dyshomeostasis has been demonstrated to be largely involved in the development of AD, a detailed study of the interaction of BK with these two metal ions has never been addressed. In this work, we have applied mass spectrometry, circular dichroism as well as computational methods in order to assess if copper and zinc have the ability to modulate the conformation and oligomerization of BK. In addition, we have correlated the chemical data with the effect of metals on the activity of BK analyzed in cell cultures by biochemical procedures. The biochemical analyses on monocyte/macrophage cell culture (THP-1 Cell Line human) in line with the effect of metals on the conformation of BK showed that the presence of copper can affect the signaling cascade mediated by the BK receptors. The results obtained show a further role of metal ions, particularly copper, in the development and outcome of neuroinflammatory diseases. The possible implications in AD are discussed.
Collapse
Affiliation(s)
- Irina Naletova
- Dipartimento di Scienze Biomediche e Biotecnologiche "BIOMETEC", Università degli Studi di Catania, Via S. Sofia 64, 95125 Catania, Italy. and Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (C.I.R.C.M.S.B.), Piazza Umberto I, 1-70121 Bari, Italy
| | - Vincenzo G Nicoletti
- Dipartimento di Scienze Biomediche e Biotecnologiche "BIOMETEC", Università degli Studi di Catania, Via S. Sofia 64, 95125 Catania, Italy. and Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (C.I.R.C.M.S.B.), Piazza Umberto I, 1-70121 Bari, Italy
| | - Danilo Milardi
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Campus Universitario, Viale Europa, 88100 Catanzaro, Italy
| | - Giuseppe Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy.
| |
Collapse
|
22
|
Kurochkin IV, Guarnera E, Berezovsky IN. Insulin-Degrading Enzyme in the Fight against Alzheimer's Disease. Trends Pharmacol Sci 2017; 39:49-58. [PMID: 29132916 DOI: 10.1016/j.tips.2017.10.008] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/18/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
Abstract
After decades of research and clinical trials there is still no cure for Alzheimer's disease (AD). While impaired clearance of amyloid beta (Aβ) peptides is considered as one of the major causes of AD, it was recently complemented by a potential role of other toxic amyloidogenic species. Insulin-degrading enzyme (IDE) is the proteolytic culprit of various β-forming peptides, both extracellular and intracellular. On the basis of demonstrated allosteric activation of IDE against Aβ, it is possible to propose a new strategy for the targeted IDE-based cleansing of different toxic aggregation-prone peptides. Consequently, specific allosteric activation of IDE coupled with state-of-the-art compound delivery and CRISP-Cas9 technique of transgene insertion can be instrumental in the fight against AD and related neurodegenerative maladies.
Collapse
Affiliation(s)
- Igor V Kurochkin
- Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-ku, Kobe 651-2271, Japan
| | - Enrico Guarnera
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 07-01, Matrix, Singapore 138671
| | - Igor N Berezovsky
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, 07-01, Matrix, Singapore 138671; Department of Biological Sciences (DBS), National University of Singapore (NUS), 8 Medical Drive, Singapore 117579.
| |
Collapse
|
23
|
Hagar JA, Edin ML, Lih FB, Thurlow LR, Koller BH, Cairns BA, Zeldin DC, Miao EA. Lipopolysaccharide Potentiates Insulin-Driven Hypoglycemic Shock. THE JOURNAL OF IMMUNOLOGY 2017; 199:3634-3643. [PMID: 29038248 DOI: 10.4049/jimmunol.1700820] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/10/2017] [Indexed: 01/04/2023]
Abstract
Critically ill patients typically present with hyperglycemia. Treatment with conventional insulin therapy (targeting 144-180 mg/dl) improves patient survival; however, intensive insulin therapy (IIT) targeting normal blood glucose levels (81-108 mg/dl) increases the incidence of moderate and severe hypoglycemia, and increases mortality. Septic patients are especially prone to IIT-induced hypoglycemia, but the mechanism remains unknown. Here, we show that codelivery of insulin with otherwise sublethal doses of LPS induced hypoglycemic shock in mice within 1-2 h. LPS impaired clearance of insulin, which amplified insulin receptor signaling. These effects were mediated by caspase-11, TLR4, and complement, each of which trigger eicosanoid production that potentiates insulin signaling. Finally, in an animal model of sepsis, we observed that Salmonella typhimurium-infected mice exhibited simultaneous impaired insulin clearance coexisting with insulin resistance. Our results raise the possibility that septic patients have impaired insulin clearance, which could increase their susceptibility to hypoglycemia during IIT, contraindicating its use.
Collapse
Affiliation(s)
- Jon A Hagar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Matthew L Edin
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Fred B Lih
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Lance R Thurlow
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219
| | - Beverly H Koller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Bruce A Cairns
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and.,North Carolina Jaycee Burn Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute for Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Edward A Miao
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; .,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599.,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
24
|
Tundo GR, Sbardella D, Ciaccio C, Grasso G, Gioia M, Coletta A, Polticelli F, Di Pierro D, Milardi D, Van Endert P, Marini S, Coletta M. Multiple functions of insulin-degrading enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol 2017. [PMID: 28635330 DOI: 10.1080/10409238.2017.1337707] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Insulin-degrading enzyme (IDE) is a ubiquitous zinc peptidase of the inverzincin family, which has been initially discovered as the enzyme responsible for insulin catabolism; therefore, its involvement in the onset of diabetes has been largely investigated. However, further studies on IDE unraveled its ability to degrade several other polypeptides, such as β-amyloid, amylin, and glucagon, envisaging the possible implication of IDE dys-regulation in the "aggregopathies" and, in particular, in neurodegenerative diseases. Over the last decade, a novel scenario on IDE biology has emerged, pointing out a multi-functional role of this enzyme in several basic cellular processes. In particular, latest advances indicate that IDE behaves as a heat shock protein and modulates the ubiquitin-proteasome system, suggesting a major implication in proteins turnover and cell homeostasis. In addition, recent observations have highlighted that the regulation of glucose metabolism by IDE is not merely based on its largely proposed role in the degradation of insulin in vivo. There is increasing evidence that improper IDE function, regulation, or trafficking might contribute to the etiology of metabolic diseases. In addition, the enzymatic activity of IDE is affected by metals levels, thus suggesting a role also in the metal homeostasis (metallostasis), which is thought to be tightly linked to the malfunction of the "quality control" machinery of the cell. Focusing on the physiological role of IDE, we will address a comprehensive vision of the very complex scenario in which IDE takes part, outlining its crucial role in interconnecting several relevant cellular processes.
Collapse
Affiliation(s)
- Grazia R Tundo
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Diego Sbardella
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Chiara Ciaccio
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Giuseppe Grasso
- d Department of Chemistry , University of Catania , Catania , Italy.,e CNR IBB , Catania , Italy
| | - Magda Gioia
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Andrea Coletta
- f Department of Chemistry , University of Aarhus , Aarhus , Denmark
| | | | - Donato Di Pierro
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | | | - Peter Van Endert
- h Université Paris Descartes, INSERM, U1151, CNRS , Paris , France
| | - Stefano Marini
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Massimo Coletta
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| |
Collapse
|
25
|
Kurochkin IV, Guarnera E, Wong JH, Eisenhaber F, Berezovsky IN. Toward Allosterically Increased Catalytic Activity of Insulin-Degrading Enzyme against Amyloid Peptides. Biochemistry 2016; 56:228-239. [PMID: 27982586 DOI: 10.1021/acs.biochem.6b00783] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The physiological role of insulin-degrading enzyme (IDE) in the intracytosolic clearance of amyloid β (Aβ) and other amyloid-like peptides supports a hypothesis that human IDE hyperactivation could be therapeutically beneficial for the treatment of late-onset Alzheimer's disease (AD). The major challenge standing in the way of this goal is increasing the specific catalytic activity of IDE against the Aβ substrate. There were previous indications that the allosteric mode of IDE activity regulation could potentially provide a highly specific path toward degradation of amyloid-like peptides, while not dramatically affecting activity against other substrates. Recently developed theoretical concepts are used here to explore potential allosteric modulation of the IDE activity as a result of single-residue mutations. Five candidates are selected for experimental follow-up and allosteric free energy calculations: Ser137Ala, Lys396Ala, Asp426Ala, Phe807Ala, and Lys898Ala. Our experiments show that three mutations (Ser137Ala, Phe807Ala, and Lys898Ala) decrease the Km of the Aβ substrate. Mutation Lys898Ala results in increased catalytic activity of IDE; on the other hand, Lys364Ala does not change the activity and Asp426Ala diminishes it. Quantifying effects of mutations in terms of allosteric free energy, we show that favorable mutations lead to stabilization of the catalytic sites and other function-relevant distal sites as well as increased dynamics of the IDE-N and IDE-C halves that allow efficient substrate entrance and cleavage. A possibility for intramolecular upregulation of IDE activity against amyloid peptides via allosteric mutations calls for further investigations in this direction. Ultimately, we are hopeful it will lead to the development of IDE-based drugs for the treatment of the late-onset form of AD characterized by an overall impairment of Aβ clearance.
Collapse
Affiliation(s)
- Igor V Kurochkin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR) , 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Enrico Guarnera
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR) , 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Jin H Wong
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR) , 30 Biopolis Street, #07-01, Matrix, Singapore 138671
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR) , 30 Biopolis Street, #07-01, Matrix, Singapore 138671.,Department of Biological Sciences (DBS), National University of Singapore (NUS) , 8 Medical Drive, Singapore 117579.,School of Computer Engineering (SCE), Nanyang Technological University (NTU) , 50 Nanyang Drive, Singapore 637553
| | - Igor N Berezovsky
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR) , 30 Biopolis Street, #07-01, Matrix, Singapore 138671.,Department of Biological Sciences (DBS), National University of Singapore (NUS) , 8 Medical Drive, Singapore 117579
| |
Collapse
|
26
|
Neuroprotective effects of Caralluma tuberculata on ameliorating cognitive impairment in a d-galactose-induced mouse model. Biomed Pharmacother 2016; 84:387-394. [DOI: 10.1016/j.biopha.2016.09.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 12/21/2022] Open
|
27
|
Tundo GR, Di Muzio E, Ciaccio C, Sbardella D, Di Pierro D, Polticelli F, Coletta M, Marini S. Multiple allosteric sites are involved in the modulation of insulin-degrading-enzyme activity by somatostatin. FEBS J 2016; 283:3755-3770. [PMID: 27579517 DOI: 10.1111/febs.13841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/18/2016] [Accepted: 08/30/2016] [Indexed: 11/30/2022]
Abstract
Somatostatin is a cyclic peptide, released in the gastrointestinal system and the central nervous system, where it is involved in the regulation of cognitive and sensory functions, motor activity and sleep. It is a substrate of insulin-degrading enzyme (IDE), as well as a modulator of its activity and expression. In the present study, we have investigated the modulatory role of somatostatin on IDE activity at 37 °C and pH 7.3 for various substrates [i.e. insulin, β-amyloid (Aβ)1-40 and bradykinin], aiming to quantitatively characterize the correlation between the specific features of the substrates and the regulatory mechanism. Functional data indicate that somatostatin, in addition to the catalytic site of IDE (being a substrate), is also able to bind to two additional exosites, which play different roles according to the size of the substrate and its binding mode to the IDE catalytic cleft. In particular, one exosite, which displays high affinity for somatostatin, regulates only the interaction of IDE with larger substrates (such as insulin and Aβ1-40 ) in a differing fashion according to their various modes of binding to the enzyme. A second exosite, which is involved in the regulation of enzymatic processing by IDE of all substrates investigated (including a 10-25 amino acid long amyloid-like peptide, bradykinin and somatostatin itself, which had been studied previously), probably acts through the alteration of an 'open-closed' equilibrium.
Collapse
Affiliation(s)
- Grazia R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy.,Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, Bari, Italy
| | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy.,Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, Bari, Italy
| | - Diego Sbardella
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy.,Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, Bari, Italy
| | - Donato Di Pierro
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy
| | - Fabio Polticelli
- Department of Sciences, University of Roma Tre, Italy.,National Institute of Nuclear Physics, Roma Tre Section, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy.,Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, Bari, Italy
| | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Italy. .,Interuniversity Consortium for the Research on Chemistry of Metals in Biological Systems, Bari, Italy.
| |
Collapse
|
28
|
Hubin E, Cioffi F, Rozenski J, van Nuland NAJ, Broersen K. Characterization of insulin-degrading enzyme-mediated cleavage of Aβ in distinct aggregation states. Biochim Biophys Acta Gen Subj 2016; 1860:1281-90. [PMID: 26968463 DOI: 10.1016/j.bbagen.2016.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 02/19/2016] [Accepted: 03/07/2016] [Indexed: 12/15/2022]
Abstract
To enhance our understanding of the potential therapeutic utility of insulin-degrading enzyme (IDE) in Alzheimer's disease (AD), we studied in vitro IDE-mediated degradation of different amyloid-beta (Aβ) peptide aggregation states. Our findings show that IDE activity is driven by the dynamic equilibrium between Aβ monomers and higher ordered aggregates. We identify Met(35)-Val(36) as a novel IDE cleavage site in the Aβ sequence and show that Aβ fragments resulting from IDE cleavage form non-toxic amorphous aggregates. These findings need to be taken into account in therapeutic strategies designed to increase Aβ clearance in AD patients by modulating IDE activity.
Collapse
Affiliation(s)
- Ellen Hubin
- Nanobiophysics Group, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, Universiteit Twente, Enschede, The Netherlands; Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium
| | - Federica Cioffi
- Nanobiophysics Group, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, Universiteit Twente, Enschede, The Netherlands
| | - Jef Rozenski
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, 3000 Leuven, Belgium
| | - Nico A J van Nuland
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; Structural Biology Research Center, VIB, Pleinlaan 2, 1050 Brussels, Belgium.
| | - Kerensa Broersen
- Nanobiophysics Group, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, Universiteit Twente, Enschede, The Netherlands.
| |
Collapse
|
29
|
Tang WJ. Targeting Insulin-Degrading Enzyme to Treat Type 2 Diabetes Mellitus. Trends Endocrinol Metab 2016; 27:24-34. [PMID: 26651592 PMCID: PMC4698235 DOI: 10.1016/j.tem.2015.11.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 10/22/2022]
Abstract
Insulin-degrading enzyme (IDE) selectively degrades peptides, such as insulin, amylin, and amyloid β (Aβ) that form toxic aggregates, to maintain proteostasis. IDE defects are linked to the development of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD). Structural and biochemical analyses revealed the molecular basis for IDE-mediated destruction of amyloidogenic peptides and this information has been exploited to develop promising inhibitors of IDE to improve glucose homeostasis. However, the inhibition of IDE can also lead to glucose intolerance. In this review, I focus on recent advances regarding our understanding of the structure and function of IDE and the discovery of IDE inhibitors, as well as challenges in developing IDE-based therapy for human diseases, particularly T2DM.
Collapse
Affiliation(s)
- Wei-Jen Tang
- Ben-May Department for Cancer Research, the University of Chicago, Chicago, IL, USA.
| |
Collapse
|
30
|
Paul TJ, Barman A, Ozbil M, Bora RP, Zhang T, Sharma G, Hoffmann Z, Prabhakar R. Mechanisms of peptide hydrolysis by aspartyl and metalloproteases. Phys Chem Chem Phys 2016; 18:24790-24801. [DOI: 10.1039/c6cp02097f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Peptide hydrolysis has been involved in a wide range of biological, biotechnological, and industrial applications.
Collapse
Affiliation(s)
- Thomas J. Paul
- Department of Chemistry
- University of Miami
- Coral Gables
- USA
| | - Arghya Barman
- Department of Chemistry
- University of Miami
- Coral Gables
- USA
| | - Mehmet Ozbil
- Department of Chemistry
- University of Miami
- Coral Gables
- USA
| | | | - Tingting Zhang
- Department of Chemistry
- University of Miami
- Coral Gables
- USA
| | - Gaurav Sharma
- Department of Chemistry
- University of Miami
- Coral Gables
- USA
| | | | | |
Collapse
|
31
|
Abdul-Hay SO, Bannister TD, Wang H, Cameron MD, Caulfield TR, Masson A, Bertrand J, Howard EA, McGuire MP, Crisafulli U, Rosenberry TR, Topper CL, Thompson CR, Schürer SC, Madoux F, Hodder P, Leissring MA. Selective Targeting of Extracellular Insulin-Degrading Enzyme by Quasi-Irreversible Thiol-Modifying Inhibitors. ACS Chem Biol 2015; 10:2716-24. [PMID: 26398879 PMCID: PMC10127574 DOI: 10.1021/acschembio.5b00334] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Many therapeutically important enzymes are present in multiple cellular compartments, where they can carry out markedly different functions; thus, there is a need for pharmacological strategies to selectively manipulate distinct pools of target enzymes. Insulin-degrading enzyme (IDE) is a thiol-sensitive zinc-metallopeptidase that hydrolyzes diverse peptide substrates in both the cytosol and the extracellular space, but current genetic and pharmacological approaches are incapable of selectively inhibiting the protease in specific subcellular compartments. Here, we describe the discovery, characterization, and kinetics-based optimization of potent benzoisothiazolone-based inhibitors that, by virtue of a unique quasi-irreversible mode of inhibition, exclusively inhibit extracellular IDE. The mechanism of inhibition involves nucleophilic attack by a specific active-site thiol of the enzyme on the inhibitors, which bear an isothiazolone ring that undergoes irreversible ring opening with the formation of a disulfide bond. Notably, binding of the inhibitors is reversible under reducing conditions, thus restricting inhibition to IDE present in the extracellular space. The identified inhibitors are highly potent (IC50(app) = 63 nM), nontoxic at concentrations up to 100 μM, and appear to preferentially target a specific cysteine residue within IDE. These novel inhibitors represent powerful new tools for clarifying the physiological and pathophysiological roles of this poorly understood protease, and their unusual mechanism of action should be applicable to other therapeutic targets.
Collapse
Affiliation(s)
- Samer O. Abdul-Hay
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | | | | | | | - Thomas R. Caulfield
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Amandine Masson
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Juliette Bertrand
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Erin A. Howard
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Michael P. McGuire
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Umberto Crisafulli
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Terrone R. Rosenberry
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Caitlyn L. Topper
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| | - Caroline R. Thompson
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| | - Stephan C. Schürer
- Department
of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida 33136, United States
| | | | | | - Malcolm A. Leissring
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| |
Collapse
|
32
|
Jha NK, Jha SK, Kumar D, Kejriwal N, Sharma R, Ambasta RK, Kumar P. Impact of Insulin Degrading Enzyme and Neprilysin in Alzheimer’s Disease Biology: Characterization of Putative Cognates for Therapeutic Applications. J Alzheimers Dis 2015; 48:891-917. [DOI: 10.3233/jad-150379] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Niraj Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Saurabh Kumar Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Dhiraj Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Noopur Kejriwal
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Renu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Rashmi K. Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Shahbad Daulatpur, Delhi, India
- Department of Neurology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
33
|
Insulin-degrading enzyme is activated by the C-terminus of α-synuclein. Biochem Biophys Res Commun 2015; 466:192-5. [PMID: 26343304 DOI: 10.1016/j.bbrc.2015.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 09/01/2015] [Indexed: 11/22/2022]
Abstract
The insulin-degrading enzyme (IDE) plays a key role in type-2 diabetes and typically degrades small peptides such as insulin, amyloid β and islet amyloid polypeptide. We recently reported a novel non-proteolytical interaction in vitro between IDE and the Parkinson's disease 140-residue protein α-synuclein that resulted in dual effects: arrested α-synuclein oligomers and, simultaneously, increased IDE proteolysis activity. Here we demonstrate that these outcomes arise due to IDE interactions with the C-terminus of α-synuclein. Whereas a peptide containing the first 97 residues of α-synuclein did not improve IDE activity and its aggregation was not blocked by IDE, a peptide with the C-terminal 44 residues of α-synuclein increased IDE proteolysis to the same degree as full-length α-synuclein. Because the α-synuclein C-terminus is acidic, the interaction appears to involve electrostatic attraction with IDE's basic exosite, known to be involved in activation.
Collapse
|
34
|
Sharma SK, Chorell E, Steneberg P, Vernersson-Lindahl E, Edlund H, Wittung-Stafshede P. Insulin-degrading enzyme prevents α-synuclein fibril formation in a nonproteolytical manner. Sci Rep 2015; 5:12531. [PMID: 26228656 PMCID: PMC4521159 DOI: 10.1038/srep12531] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/29/2015] [Indexed: 11/25/2022] Open
Abstract
The insulin-degrading enzyme (IDE) degrades amyloidogenic proteins such as Amyloid β (Αβ) and Islet Amyloid Polypeptide (IAPP), i.e. peptides associated with Alzheimer’s disease and type 2 diabetes, respectively. In addition to the protease activity normally associated with IDE function an additional activity involving the formation of stable, irreversible complexes with both Αβ and α-synuclein, an amyloidogenic protein involved in Parkinson’s disease, was recently proposed. Here, we have investigated the functional consequences of IDE-α-synuclein interactions in vitro. We demonstrate that IDE in a nonproteolytic manner and at sub-stoichiometric ratios efficiently inhibits α-synuclein fibril formation by binding to α-synuclein oligomers making them inert to amyloid formation. Moreover, we show that, within a defined range of α-synuclein concentrations, interaction with α-synuclein oligomers increases IDE’s proteolytic activity on a fluorogenic substrate. We propose that the outcomes of IDE-α-synuclein interactions, i.e. protection against α-synuclein amyloid formation and stimulated IDE protease activity, may be protective in vivo.
Collapse
Affiliation(s)
| | - Erik Chorell
- Department of Chemistry, Umeå University, 90187 Umeå, Sweden
| | - Pär Steneberg
- Umeå Center for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | | | - Helena Edlund
- Umeå Center for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | | |
Collapse
|
35
|
Durham TB, Toth JL, Klimkowski VJ, Cao JXC, Siesky AM, Alexander-Chacko J, Wu GY, Dixon JT, McGee JE, Wang Y, Guo SY, Cavitt RN, Schindler J, Thibodeaux SJ, Calvert NA, Coghlan MJ, Sindelar DK, Christe M, Kiselyov VV, Michael MD, Sloop KW. Dual Exosite-binding Inhibitors of Insulin-degrading Enzyme Challenge Its Role as the Primary Mediator of Insulin Clearance in Vivo. J Biol Chem 2015; 290:20044-59. [PMID: 26085101 DOI: 10.1074/jbc.m115.638205] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Indexed: 01/07/2023] Open
Abstract
Insulin-degrading enzyme (IDE, insulysin) is the best characterized catabolic enzyme implicated in proteolysis of insulin. Recently, a peptide inhibitor of IDE has been shown to affect levels of insulin, amylin, and glucagon in vivo. However, IDE(-/-) mice display variable phenotypes relating to fasting plasma insulin levels, glucose tolerance, and insulin sensitivity depending on the cohort and age of animals. Here, we interrogated the importance of IDE-mediated catabolism on insulin clearance in vivo. Using a structure-based design, we linked two newly identified ligands binding at unique IDE exosites together to construct a potent series of novel inhibitors. These compounds do not interact with the catalytic zinc of the protease. Because one of these inhibitors (NTE-1) was determined to have pharmacokinetic properties sufficient to sustain plasma levels >50 times its IDE IC50 value, studies in rodents were conducted. In oral glucose tolerance tests with diet-induced obese mice, NTE-1 treatment improved the glucose excursion. Yet in insulin tolerance tests and euglycemic clamp experiments, NTE-1 did not enhance insulin action or increase plasma insulin levels. Importantly, IDE inhibition with NTE-1 did result in elevated plasma amylin levels, suggesting the in vivo role of IDE action on amylin may be more significant than an effect on insulin. Furthermore, using the inhibitors described in this report, we demonstrate that in HEK cells IDE has little impact on insulin clearance. In total, evidence from our studies supports a minimal role for IDE in insulin metabolism in vivo and suggests IDE may be more important in helping regulate amylin clearance.
Collapse
Affiliation(s)
| | - James L Toth
- From the Discovery Chemistry Research and Technologies
| | | | | | | | | | | | | | - James E McGee
- From the Discovery Chemistry Research and Technologies
| | - Yong Wang
- From the Discovery Chemistry Research and Technologies
| | - Sherry Y Guo
- From the Discovery Chemistry Research and Technologies
| | | | | | | | - Nathan A Calvert
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285
| | | | | | | | | | | | | |
Collapse
|
36
|
Liang WG, Ren M, Zhao F, Tang WJ. Structures of human CCL18, CCL3, and CCL4 reveal molecular determinants for quaternary structures and sensitivity to insulin-degrading enzyme. J Mol Biol 2015; 427:1345-1358. [PMID: 25636406 PMCID: PMC4355285 DOI: 10.1016/j.jmb.2015.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/07/2015] [Accepted: 01/21/2015] [Indexed: 11/23/2022]
Abstract
CC chemokine ligands (CCL) are 8-14 kDa signaling proteins involved in diverse immune functions. While CCLs share similar tertiary structures, oligomerization produces highly diverse quaternary structures that protect chemokines from proteolytic degradation and modulate their functions. CCL18 is closely related to CCL3 and CCL4 with respect to both protein sequence and genomic location, yet CCL18 has distinct biochemical and biophysical properties. Here, we report a crystal structure of human CCL18 and its oligomerization states in solution based on crystallographic and small angle X-ray scattering (SAXS) analyses. Our data shows that CCL18 adopts an α-helical conformation at its N-terminus that weakens its dimerization, explaining CCL18’s preference for the monomeric state. Multiple contacts between monomers allow CCL18 to reversibly form a unique open-ended oligomer different from those of CCL3, CCL4, and CCL5. Furthermore, these differences hinge on proline 8, which is conserved in CCL3 and CCL4, but is replaced by lysine in human CCL18. Our structural analyses suggest that a proline 8 to alanine mutation stabilizes a type I β-turn at the N-terminus of CCL4 to prevent dimerization but prevents dimers from making key contacts with each other in CCL3. Thus, the P8A mutation induces depolymerization of CCL3 and CCL4 by distinct mechanisms. Finally, we used structural, biochemical, and functional analyses to unravel why insulin-degrading enzyme (IDE) degrades CCL3 and CCL4 but not CCL18. Our results elucidate the molecular basis for the oligomerization of three closely related CC chemokines and suggest how oligomerization shapes CCL chemokine function.
Collapse
Affiliation(s)
- Wenguang G Liang
- Ben May Department for Cancer Research, The University of Chicago, IL 60637, USA
| | - Min Ren
- Ben May Department for Cancer Research, The University of Chicago, IL 60637, USA
| | - Fan Zhao
- Ben May Department for Cancer Research, The University of Chicago, IL 60637, USA
| | - Wei-Jen Tang
- Ben May Department for Cancer Research, The University of Chicago, IL 60637, USA.
| |
Collapse
|
37
|
da Cruz CHB, Seabra GM. QM/MM simulations of amyloid-β 42 degradation by IDE in the presence and absence of ATP. J Chem Inf Model 2015; 55:72-83. [PMID: 25539133 DOI: 10.1021/ci500544c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ability of the insulin-degrading enzyme (IDE) to degrade amyloid-β 42 (Aβ42), a process regulated by ATP, has been studied as an alternative path in the development of drugs against Alzheimer's disease. In this study, we calculated the potential of mean force for the degradation of Aβ42 by IDE in the presence and absence of ATP by umbrella sampling with hybrid quantum mechanics and molecular mechanics (QM/MM) calculations, using the SCC-DFTB QM Hamiltonian and Amber ff99SB force field. Results indicate that the reaction occurs in two steps: The first step is characterized by the formation of the intermediate. The second step is characterized by breaking the peptide bond of the substrate, the latter being the rate-determining step. In our simulations, the activation energy barrier in the absence of ATP is 15 ± 2 kcal mol(-1), which is 7 kcal mol(-1) lower than in the presence of ATP, indicating that the presence of the nucleotide decreases the reaction rate by about 10(5) times.
Collapse
Affiliation(s)
- Carlos H B da Cruz
- Departamento de Química Fundamental, Universidade Federal de Pernambuco , Av. Jornalista Aníbal Fernandes, s/n, Cidade Universitária, Recife-PE, Brazil , 50.740-560
| | | |
Collapse
|
38
|
Wilson BA, Cruz-Diaz N, Marshall AC, Pirro NT, Su Y, Gwathmey TM, Rose JC, Chappell MC. An angiotensin-(1-7) peptidase in the kidney cortex, proximal tubules, and human HK-2 epithelial cells that is distinct from insulin-degrading enzyme. Am J Physiol Renal Physiol 2015; 308:F594-601. [PMID: 25568136 DOI: 10.1152/ajprenal.00609.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Angiotensin 1-7 [ANG-(1-7)] is expressed within the kidney and exhibits renoprotective actions that antagonize the inflammatory, fibrotic, and pro-oxidant effects of ANG II. We previously identified an peptidase that preferentially metabolized ANG-(1-7) to ANG-(1-4) in the brain medulla and cerebrospinal fluid (CSF) of sheep (Marshall AC, Pirro NT, Rose JC, Diz DI, Chappell MC. J Neurochem 130: 313-323, 2014); thus the present study established the expression of the peptidase in the kidney. Utilizing a sensitive HPLC-based approach, we demonstrate a peptidase activity that hydrolyzed ANG-(1-7) to ANG-(1-4) in the sheep cortex, isolated tubules, and human HK-2 renal epithelial cells. The peptidase was markedly sensitive to the metallopeptidase inhibitor JMV-390; human HK-2 cells expressed subnanomolar sensitivity (IC50 = 0.5 nM) and the highest specific activity (123 ± 5 fmol·min(-1)·mg(-1)) compared with the tubules (96 ± 12 fmol·min(-1)·mg(-1)) and cortex (107 ± 9 fmol·min(-1)·mg(-1)). The peptidase was purified 41-fold from HK-2 cells; the activity was sensitive to JMV-390, the chelator o-phenanthroline, and the mercury-containing compound p-chloromercuribenzoic acid (PCMB), but not to selective inhibitors against neprilysin, neurolysin and thimet oligopeptidase. Both ANG-(1-7) and its endogenous analog [Ala(1)]-ANG-(1-7) (alamandine) were preferentially hydrolyzed by the peptidase compared with ANG II, [Asp(1)]-ANG II, ANG I, and ANG-(1-12). Although the ANG-(1-7) peptidase and insulin-degrading enzyme (IDE) share similar inhibitor characteristics of a metallothiolendopeptidase, we demonstrate marked differences in substrate specificity, which suggest these peptidases are distinct. We conclude that an ANG-(1-7) peptidase is expressed within the renal proximal tubule and may play a potential role in the renal renin-angiotensin system to regulate ANG-(1-7) tone.
Collapse
Affiliation(s)
- Bryan A Wilson
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| | - Nildris Cruz-Diaz
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| | - Allyson C Marshall
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| | - Nancy T Pirro
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| | - Yixin Su
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, North Cartolina
| | - TanYa M Gwathmey
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| | - James C Rose
- Department of Obstetrics and Gynecology, Wake Forest University School of Medicine, Winston-Salem, North Cartolina
| | - Mark C Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, North Cartolina; and
| |
Collapse
|
39
|
Anti-diabetic activity of insulin-degrading enzyme inhibitors mediated by multiple hormones. Nature 2014; 511:94-8. [PMID: 24847884 DOI: 10.1038/nature13297] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 03/27/2014] [Indexed: 02/07/2023]
Abstract
Despite decades of speculation that inhibiting endogenous insulin degradation might treat type-2 diabetes, and the identification of IDE (insulin-degrading enzyme) as a diabetes susceptibility gene, the relationship between the activity of the zinc metalloprotein IDE and glucose homeostasis remains unclear. Although Ide(-/-) mice have elevated insulin levels, they exhibit impaired, rather than improved, glucose tolerance that may arise from compensatory insulin signalling dysfunction. IDE inhibitors that are active in vivo are therefore needed to elucidate IDE's physiological roles and to determine its potential to serve as a target for the treatment of diabetes. Here we report the discovery of a physiologically active IDE inhibitor identified from a DNA-templated macrocycle library. An X-ray structure of the macrocycle bound to IDE reveals that it engages a binding pocket away from the catalytic site, which explains its remarkable selectivity. Treatment of lean and obese mice with this inhibitor shows that IDE regulates the abundance and signalling of glucagon and amylin, in addition to that of insulin. Under physiological conditions that augment insulin and amylin levels, such as oral glucose administration, acute IDE inhibition leads to substantially improved glucose tolerance and slower gastric emptying. These findings demonstrate the feasibility of modulating IDE activity as a new therapeutic strategy to treat type-2 diabetes and expand our understanding of the roles of IDE in glucose and hormone regulation.
Collapse
|
40
|
Wang C, He L, Yan M, Zheng GY, Liu XY. Effects of polyprenols from pine needles of Pinus massoniana on ameliorating cognitive impairment in a D-galactose-induced mouse model. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9676. [PMID: 24981114 PMCID: PMC4150899 DOI: 10.1007/s11357-014-9676-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 06/18/2014] [Indexed: 05/14/2023]
Abstract
Cognitive deficiency and oxidative stress have been well documented in aging and in neurodegenerative disorders such as Alzheimer's disease. In this study, we assessed the therapeutic effect of polyprenols on D-galactose-induced cognitive impairment in mice by testing on of behavioral and cognitive performance. In order to explore the possible role of polyprenols against D-galactose-induced oxidative damages, we assessed various biochemical indicators. Chronic administration of D-galactose (150 mg/kg·d, s.c.) for 7 weeks significantly impaired cognitive performance (both in step-through passive and active avoidance tests) and locomotor activity (in open-field test) and the ability of spatial learning and memory (in Morris water maze test) compared with the control group. The results revealed that polyprenols treatment for 2 weeks significantly ameliorated model mice's cognitive performance and oxidative defense. All groups of polyprenols enhanced the learning and memory ability in step-through passive and active avoidance tests, locomotor activity in open-field test, and the ability of spatial learning and memory in Morris water maze test. Furthermore, high and middle level of polyprenols significantly increased total antioxidative capacity (T-AOC), glutathione peroxidase (GSH-Px), super oxide dismutase (SOD) activity, neprilysin (NEP), and β-site AβPP cleaving enzyme 1 (BACE1) expression, while nitric oxide (NO), nitric oxide synthase (NOS) activity, malondialdehyde (MDA) concentration, and the level of Aβ1-42 and presenilin 1 (PS1) were decreased. Polyprenols have a significant relieving effect on learning, memory, and spontaneous activities in a D-galactose-induced mouse model and ameliorates cognitive impairment and biochemical dysfunction in mice. In summary, we have demonstrated that polyprenols may ameliorate memory and cognitive impairment via enhancing oxidative defense and affecting generation and dissimilation of Aβ-related enzymes, suggesting that polyprenols represent a novel drug for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Cong Wang
- />Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, JiangSu Province China
| | - Ling He
- />Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, JiangSu Province China
| | - Ming Yan
- />National Drug Screening Laboratory, China Pharmaceutical University, Nanjing, 210009 China
| | - Guang-yao Zheng
- />Institute of Chemical Industry of Forest Products, CAF, National Engineering Lab for Biomass Chemical Utilization, Key and Open Lab of Forest Chemical Engineering, SFA, Key Lab of Biomass Energy and Material, Jiangsu Province, Nanjing, 210042 China
| | - Xiao-yang Liu
- />Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, 210009, JiangSu Province China
| |
Collapse
|
41
|
Conformational states and recognition of amyloidogenic peptides of human insulin-degrading enzyme. Proc Natl Acad Sci U S A 2013; 110:13827-32. [PMID: 23922390 DOI: 10.1073/pnas.1304575110] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Insulin-degrading enzyme (IDE) selectively degrades the monomer of amyloidogenic peptides and contributes to clearance of amyloid β (Aβ). Thus, IDE retards the progression of Alzheimer's disease. IDE possesses an enclosed catalytic chamber that engulfs and degrades its peptide substrates; however, the molecular mechanism of IDE function, including substrate access to the chamber and recognition, remains elusive. Here, we captured a unique IDE conformation by using a synthetic antibody fragment as a crystallization chaperone. An unexpected displacement of a door subdomain creates an ~18-Å opening to the chamber. This swinging-door mechanism permits the entry of short peptides into the catalytic chamber and disrupts the catalytic site within IDE door subdomain. Given the propensity of amyloidogenic peptides to convert into β-strands for their polymerization into amyloid fibrils, they also use such β-strands to stabilize the disrupted catalytic site resided at IDE door subdomain for their degradation by IDE. Thus, action of the swinging door allows IDE to recognize amyloidogenicity by substrate-induced stabilization of the IDE catalytic cleft. Small angle X-ray scattering (SAXS) analysis revealed that IDE exists as a mixture of closed and open states. These open states, which are distinct from the swinging door state, permit entry of larger substrates (e.g., Aβ, insulin) to the chamber and are preferred in solution. Mutational studies confirmed the critical roles of the door subdomain and hinge loop joining the N- and C-terminal halves of IDE for catalysis. Together, our data provide insights into the conformational changes of IDE that govern the selective destruction of amyloidogenic peptides.
Collapse
|
42
|
Nakamura T, Tu S, Akhtar MW, Sunico CR, Okamoto SI, Lipton SA. Aberrant protein s-nitrosylation in neurodegenerative diseases. Neuron 2013; 78:596-614. [PMID: 23719160 DOI: 10.1016/j.neuron.2013.05.005] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2013] [Indexed: 12/14/2022]
Abstract
S-Nitrosylation is a redox-mediated posttranslational modification that regulates protein function via covalent reaction of nitric oxide (NO)-related species with a cysteine thiol group on the target protein. Under physiological conditions, S-nitrosylation can be an important modulator of signal transduction pathways, akin to phosphorylation. However, with aging or environmental toxins that generate excessive NO, aberrant S-nitrosylation reactions can occur and affect protein misfolding, mitochondrial fragmentation, synaptic function, apoptosis or autophagy. Here, we discuss how aberrantly S-nitrosylated proteins (SNO-proteins) play a crucial role in the pathogenesis of neurodegenerative diseases, including Alzheimer's and Parkinson's diseases. Insight into the pathophysiological role of aberrant S-nitrosylation pathways will enhance our understanding of molecular mechanisms leading to neurodegenerative diseases and point to potential therapeutic interventions.
Collapse
Affiliation(s)
- Tomohiro Nakamura
- Del E. Web Center for Neuroscience, Aging, and Stem Cell Research, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Song ES, Melikishvili M, Fried MG, Juliano MA, Juliano L, Rodgers DW, Hersh LB. Cysteine 904 is required for maximal insulin degrading enzyme activity and polyanion activation. PLoS One 2012; 7:e46790. [PMID: 23077523 PMCID: PMC3471918 DOI: 10.1371/journal.pone.0046790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 09/06/2012] [Indexed: 11/19/2022] Open
Abstract
Cysteine residues in insulin degrading enzyme have been reported as non-critical for its activity. We found that converting the twelve cysteine residues in rat insulin degrading enzyme (IDE) to serines resulted in a cysteine-free form of the enzyme with reduced activity and decreased activation by polyanions. Mutation of each cysteine residue individually revealed cysteine 904 as the key residue required for maximal activity and polyanion activation, although other cysteines affect polyanion binding to a lesser extent. Based on the structure of IDE, Asn 575 was identified as a potential hydrogen bond partner for Cys904 and mutation of this residue also reduced activity and decreased polyanion activation. The oligomerization state of IDE did not correlate with its activity, with the dimer being the predominant form in all the samples examined. These data suggest that there are several conformational states of the dimer that affect activity and polyanion activation.
Collapse
Affiliation(s)
- Eun Suk Song
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Manana Melikishvili
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Michael G. Fried
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Maria A. Juliano
- Department of Biophysics, Universidade Federal de Sao Paulo, Escola Paulista de Medicina, Rua Tres de Maio, Sao Paulo, Brazil
| | - Luiz Juliano
- Department of Biophysics, Universidade Federal de Sao Paulo, Escola Paulista de Medicina, Rua Tres de Maio, Sao Paulo, Brazil
| | - David W. Rodgers
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Louis B. Hersh
- Department of Molecular and Cellular Biochemistry and the Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
44
|
Grasso G, Salomone F, Tundo GR, Pappalardo G, Ciaccio C, Spoto G, Pietropaolo A, Coletta M, Rizzarelli E. Metal ions affect insulin-degrading enzyme activity. J Inorg Biochem 2012; 117:351-8. [PMID: 22819648 DOI: 10.1016/j.jinorgbio.2012.06.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/07/2012] [Accepted: 06/10/2012] [Indexed: 01/24/2023]
Abstract
Insulin degradation is a finely tuned process that plays a major role in controlling insulin action and most evidence supports IDE (insulin-degrading enzyme) as the primary degradative agent. However, the biomolecular mechanisms involved in the interaction between IDE and its substrates are often obscure, rendering the specific enzyme activity quite difficult to target. On the other hand, biometals, such as copper, aluminum and zinc, have an important role in pathological conditions such as Alzheimer's disease or diabetes mellitus. The metabolic disorders connected with the latter lead to some metallostasis alterations in the human body and many studies point at a high level of interdependence between diabetes and several cations. We have previously reported (Grasso et al., Chem. Eur. J. 17 (2011) 2752-2762) that IDE activity toward Aβ peptides can be modulated by metal ions. Here, we have investigated the effects of different metal ions on the IDE proteolytic activity toward insulin as well as a designed peptide comprising a portion of the insulin B chain (B20-30), which has a very low affinity for metal ions. The results obtained by different experimental techniques clearly show that IDE is irreversibly inhibited by copper(I) but is still able to process its substrates when it is bound to copper(II).
Collapse
Affiliation(s)
- Giuseppe Grasso
- Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Structure based discovery of small molecules to regulate the activity of human insulin degrading enzyme. PLoS One 2012; 7:e31787. [PMID: 22355395 PMCID: PMC3280214 DOI: 10.1371/journal.pone.0031787] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 01/19/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Insulin-degrading enzyme (IDE) is an allosteric Zn(+2) metalloprotease involved in the degradation of many peptides including amyloid-β, and insulin that play key roles in Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM), respectively. Therefore, the use of therapeutic agents that regulate the activity of IDE would be a viable approach towards generating pharmaceutical treatments for these diseases. Crystal structure of IDE revealed that N-terminal has an exosite which is ∼30 Å away from the catalytic region and serves as a regulation site by orientation of the substrates of IDE to the catalytic site. It is possible to find small molecules that bind to the exosite of IDE and enhance its proteolytic activity towards different substrates. METHODOLOGY/PRINCIPAL FINDINGS In this study, we applied structure based drug design method combined with experimental methods to discover four novel molecules that enhance the activity of human IDE. The novel compounds, designated as D3, D4, D6, and D10 enhanced IDE mediated proteolysis of substrate V, insulin and amyloid-β, while enhanced degradation profiles were obtained towards substrate V and insulin in the presence of D10 only. CONCLUSION/SIGNIFICANCE This paper describes the first examples of a computer-aided discovery of IDE regulators, showing that in vitro and in vivo activation of this important enzyme with small molecules is possible.
Collapse
|
46
|
Guttmann RP, Ghoshal S. Thiol-protease oxidation in age-related neuropathology. Free Radic Biol Med 2011; 51:282-8. [PMID: 21565267 DOI: 10.1016/j.freeradbiomed.2011.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 04/07/2011] [Accepted: 04/07/2011] [Indexed: 11/27/2022]
Abstract
Increased oxidative stress is a hallmark of every major neurodegenerative disease that has been studied. Numerous biomarkers of oxidative stress have been found, indicating that waves of oxidation had, at one time or another, overwhelmed antioxidant defenses, leaving behind a host of oxidized DNA, lipids, and proteins in their path. Although some level of oxidation may be beneficial, perhaps mediated by a hormetic response, the extent and types of oxidation detected in neuropathological states would suggest that oxidative stress contributes to a loss of homeostasis and cellular dysfunction. Although there are many targets of oxidants, this review emphasizes protein oxidation with a focus on an important group of redox-sensitive enzymes, the thiol-proteases. Both the direct and the indirect effects of oxidation and their potential importance in neurodegeneration are considered.
Collapse
Affiliation(s)
- Rodney P Guttmann
- Department of Gerontology, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| | | |
Collapse
|
47
|
Noinaj N, Bhasin SK, Song ES, Scoggin KE, Juliano MA, Juliano L, Hersh LB, Rodgers DW. Identification of the allosteric regulatory site of insulysin. PLoS One 2011; 6:e20864. [PMID: 21731629 PMCID: PMC3123307 DOI: 10.1371/journal.pone.0020864] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 05/14/2011] [Indexed: 11/29/2022] Open
Abstract
Background Insulin degrading enzyme (IDE) is responsible for the metabolism of insulin and plays a role in clearance of the Aβ peptide associated with Alzheimer's disease. Unlike most proteolytic enzymes, IDE, which consists of four structurally related domains and exists primarily as a dimer, exhibits allosteric kinetics, being activated by both small substrate peptides and polyphosphates such as ATP. Principal Findings The crystal structure of a catalytically compromised mutant of IDE has electron density for peptide ligands bound at the active site in domain 1 and a distal site in domain 2. Mutating residues in the distal site eliminates allosteric kinetics and activation by a small peptide, as well as greatly reducing activation by ATP, demonstrating that this site plays a key role in allostery. Comparison of the peptide bound IDE structure (using a low activity E111F IDE mutant) with unliganded wild type IDE shows a change in the interface between two halves of the clamshell-like molecule, which may enhance enzyme activity by altering the equilibrium between closed and open conformations. In addition, changes in the dimer interface suggest a basis for communication between subunits. Conclusions/Significance Our findings indicate that a region remote from the active site mediates allosteric activation of insulysin by peptides. Activation may involve a small conformational change that weakens the interface between two halves of the enzyme.
Collapse
Affiliation(s)
- Nicholas Noinaj
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sonia K. Bhasin
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Eun Suk Song
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kirsten E. Scoggin
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Maria A. Juliano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Luiz Juliano
- Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Louis B. Hersh
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
| | - David W. Rodgers
- Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
48
|
Clayton D, Hanchapola I, Hausler N, Unabia S, Lew RA, Widdop RE, Smith AI, Perlmutter P, Aguilar MI. β-Amino acid substitution to investigate the recognition of angiotensin II (AngII) by angiotensin converting enzyme 2 (ACE2). J Mol Recognit 2011; 24:235-44. [PMID: 20549604 DOI: 10.1002/jmr.1041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In spite of the important role of angiotensin converting enzyme 2 (ACE2) in the cardiovascular system, little is known about the substrate structural requirements of the AngII-ACE2 interaction. Here we investigate how changes in angiotensin II (AngII) structure affect binding and cleavage by ACE2. A series of C3 β-amino acid AngII analogs were generated and their secondary structure, ACE2 inhibition, and proteolytic stability assessed by circular dichroism (CD), quenched fluorescence substrate (QFS) assay, and LC-MS analysis, respectively. The β-amino acid-substituted AngII analogs showed differences in secondary structure, ACE2 binding and proteolytic stability. In particular, three different subsets of structure-activity profiles were observed corresponding to substitutions in the N-terminus, the central region and the C-terminal region of AngII. The results show that β-substitution can dramatically alter the structure of AngII and changes in structure correlated with ACE2 inhibition and/or substrate cleavage. β-amino acid substitution in the N-terminal region of AngII caused little change in structure or substrate cleavage, while substitution in the central region of AngII lead to increased β-turn structure and enhanced substrate cleavage. β-amino acid substitution in the C-terminal region significantly diminished both secondary structure and proteolytic processing by ACE2. The β-AngII analogs with enhanced or decreased proteolytic stability have potential application for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Daniel Clayton
- Biochemistry & Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Glebov K, Schütze S, Walter J. Functional relevance of a novel SlyX motif in non-conventional secretion of insulin-degrading enzyme. J Biol Chem 2011; 286:22711-5. [PMID: 21576244 DOI: 10.1074/jbc.c110.217893] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Insulin-degrading enzyme (IDE) is a Zn(2+) metalloprotease with a characteristic inverted catalytic motif. IDE is ubiquitously expressed and degrades peptide substrates including insulin, endorphin, and the amyloid-β peptide. Although IDE is mainly expressed in the cytosol, it can also be found on the cell surface and in secreted form in extracellular fluids. As IDE lacks a characteristic signal sequence that targets the protein to the classical secretory pathway, release of the enzyme involves non-conventional mechanisms. However, functional domains of IDE involved in its secretion remain elusive. By bioinformatical, biochemical, and cell biological methods, we identified a novel amino acid motif ((853)EKPPHY(858)) close to the C terminus of IDE and characterized its function in the non-conventional secretion of the protein. Because of its close homology to an amino acid sequence found in bacterial proteins belonging to the SlyX family, we propose to call it the SlyX motif. Mutagenesis revealed that deletion of this motif strongly decreased the release of IDE, whereas deletion of a potential microbody-targeting signal at the extreme C terminus had little effect on secretion. The combined data indicate that the non-conventional secretion of IDE is regulated by the newly identified SlyX motif.
Collapse
|
50
|
Structural basis for antigenic peptide precursor processing by the endoplasmic reticulum aminopeptidase ERAP1. Nat Struct Mol Biol 2011; 18:604-13. [PMID: 21478864 PMCID: PMC3087843 DOI: 10.1038/nsmb.2021] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 01/26/2011] [Indexed: 02/07/2023]
Abstract
ERAP1 trims antigen precursors to fit into MHC class I proteins. To perform this function, ERAP1 has unique substrate preferences, trimming long peptides while sparing shorter ones. To identify the structural basis for ERAP1's unusual properties, we determined the X-ray crystal structure of human ERAP1 bound to bestatin. The structure reveals an open conformation with a large interior compartment. An extended groove originating from the enzyme's catalytic center can accommodate long peptides and has features that explain ERAP1's broad specificity for antigenic peptide precursors. Structural and biochemical analysis suggest a mechanism for ERAP1's length-dependent trimming activity, whereby binding of long but not short substrates induces a conformational change with reorientation of a key catalytic residue towards the active site. ERAP1's unique structural elements suggest how a generic aminopeptidase structure has been adapted for the specialized function of trimming antigenic precursors.
Collapse
|