1
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
2
|
Pinheiro FI, Araújo-Filho I, do Rego ACM, de Azevedo EP, Cobucci RN, Guzen FP. Hepatopancreatic metabolic disorders and their implications in the development of Alzheimer's disease and vascular dementia. Ageing Res Rev 2024; 96:102250. [PMID: 38417711 DOI: 10.1016/j.arr.2024.102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Dementia has been faced with significant public health challenges and economic burdens that urges the need to develop safe and effective interventions. In recent years, an increasing number of studies have focused on the relationship between dementia and liver and pancreatic metabolic disorders that result in diseases such as diabetes, obesity, hypertension and dyslipidemia. Previous reports have shown that there is a plausible correlation between pathologies caused by hepatopancreatic dysfunctions and dementia. Glucose, insulin and IGF-1 metabolized in the liver and pancreas probably have an important influence on the pathophysiology of the most common dementias: Alzheimer's and vascular dementia. This current review highlights recent studies aimed at identifying convergent mechanisms, such as insulin resistance and other diseases, linked to altered hepatic and pancreatic metabolism, which are capable of causing brain changes that ultimately lead to dementia.
Collapse
Affiliation(s)
- Francisco I Pinheiro
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Irami Araújo-Filho
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Department of Surgical, Federal University of Rio Grande do Norte, Natal 59010-180, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Amália C M do Rego
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Institute of Education, Research and Innovation of the Liga Norte Rio-Grandense Against Cancer
| | - Eduardo P de Azevedo
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil
| | - Ricardo N Cobucci
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil; Postgraduate Program in Science Applied to Women`s Health, Medical School, Federal University of Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | - Fausto P Guzen
- Postgraduate Program in Biotechnology, Health School, Potiguar University (UnP), Natal, RN, Brazil; Postgraduate Program in Health and Society, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil; Postgraduate Program in Physiological Sciences, Department of Biomedical Sciences, Faculty of Health Sciences, State University of Rio Grande do Norte (UERN), Mossoró, Brazil.
| |
Collapse
|
3
|
Majou D, Dermenghem AL. Effects of DHA (omega-3 fatty acid) and estradiol on amyloid β-peptide regulation in the brain. Brain Res 2024; 1823:148681. [PMID: 37992797 DOI: 10.1016/j.brainres.2023.148681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
In the early stages of sporadic Alzheimer's disease (SAD), there is a strong correlation between memory impairment and cortical levels of soluble amyloid-β peptide oligomers (Aβ). It has become clear that Aβ disrupt glutamatergic synaptic function, which can in turn lead to the characteristic cognitive deficits of SAD, but the actual pathways are still not well understood. This opinion article describes the pathogenic mechanisms underlying cerebral amyloidosis. These mechanisms are dependent on the amyloid precursor protein and concern the synthesis of Aβ peptides with competition between the non-amyloidogenic pathway and the amyloidogenic pathway (i.e. a competition between the ADAM10 and BACE1 enzymes), on the one hand, and the various processes of Aβ residue clearance, on the other hand. This clearance mobilizes both endopeptidases (NEP, and IDE) and removal transporters across the blood-brain barrier (LRP1, ABCB1, and RAGE). Lipidated ApoE also plays a major role in all processes. The disturbance of these pathways induces an accumulation of Aβ. The description of the mechanisms reveals two key molecules in particular: (i) free estradiol, which has genomic and non-genomic action, and (ii) free DHA as a preferential ligand of PPARα-RXRα and PPARɣ-RXRα heterodimers. DHA and free estradiol are also self-regulating, and act in synergy. When a certain level of chronic DHA and free estradiol deficiency is reached, a permanent imbalance is established in the central nervous system. The consequences of these deficits are revealed in particular by the presence of Aβ peptide deposits, as well as other markers of the etiology of SAD.
Collapse
Affiliation(s)
- Didier Majou
- ACTIA, 149, rue de Bercy, 75595 Paris Cedex 12, France.
| | | |
Collapse
|
4
|
Tundo GR, Grasso G, Persico M, Tkachuk O, Bellia F, Bocedi A, Marini S, Parravano M, Graziani G, Fattorusso C, Sbardella D. The Insulin-Degrading Enzyme from Structure to Allosteric Modulation: New Perspectives for Drug Design. Biomolecules 2023; 13:1492. [PMID: 37892174 PMCID: PMC10604886 DOI: 10.3390/biom13101492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/11/2023] [Accepted: 09/17/2023] [Indexed: 10/29/2023] Open
Abstract
The insulin-degrading enzyme (IDE) is a Zn2+ peptidase originally discovered as the main enzyme involved in the degradation of insulin and other amyloidogenic peptides, such as the β-amyloid (Aβ) peptide. Therefore, a role for the IDE in the cure of diabetes and Alzheimer's disease (AD) has been long envisaged. Anyway, its role in degrading amyloidogenic proteins remains not clearly defined and, more recently, novel non-proteolytic functions of the IDE have been proposed. From a structural point of view, the IDE presents an atypical clamshell structure, underscoring unique enigmatic enzymological properties. A better understanding of the structure-function relationship may contribute to solving some existing paradoxes of IDE biology and, in light of its multifunctional activity, might lead to novel therapeutic approaches.
Collapse
Affiliation(s)
- Grazia Raffaella Tundo
- Department of Clinical Science and Traslational Medicine, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (G.R.T.)
| | - Giuseppe Grasso
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy;
| | - Marco Persico
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.P.); (O.T.)
| | - Oleh Tkachuk
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.P.); (O.T.)
| | - Francesco Bellia
- Institute of Crystallography, CNR, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Alessio Bocedi
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Stefano Marini
- Department of Clinical Science and Traslational Medicine, University of Rome Tor Vergata, Via Della Ricerca Scientifica 1, 00133 Rome, Italy; (G.R.T.)
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Caterina Fattorusso
- Department of Pharmacy, University of Naples “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy; (M.P.); (O.T.)
| | | |
Collapse
|
5
|
Dutta BJ, Singh S, Seksaria S, Das Gupta G, Singh A. Inside the diabetic brain: Insulin resistance and molecular mechanism associated with cognitive impairment and its possible therapeutic strategies. Pharmacol Res 2022; 182:106358. [PMID: 35863719 DOI: 10.1016/j.phrs.2022.106358] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/02/2022] [Accepted: 07/15/2022] [Indexed: 01/21/2023]
Abstract
Type 2 diabetes mellitus (T2DM) the most prevalent metabolic disease that has evolved into a major public health issue. Concerning about its secondary complications, a growing body of evidence links T2DM to cognitive impairment and neurodegenerative disorders. The underlying pathology behind this secondary complication disease is yet to be fully known. Nonetheless, they are likely to be associated with poor insulin signaling as a result of insulin resistance. We have combed through a rising body of literature on insulin signaling in the normal and diabetic brains along with various factors like insulin resistance, hyperglycemia, obesity, oxidative stress, neuroinflammation and Aβ plaques which can act independently or synergistically to link T2DM with cognitive impairments. Finally, we explored several pharmacological and non-pharmacological methods in the hopes of accelerating the rational development of medications for cognitive impairment in T2DM by better understanding these shared pathways.
Collapse
Affiliation(s)
- Bhaskar Jyoti Dutta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Sanket Seksaria
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India
| | - Amrita Singh
- Department of Pharmacology, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, Punjab, India.
| |
Collapse
|
6
|
Sousa L, Guarda M, Meneses MJ, Macedo MP, Vicente Miranda H. Insulin-degrading enzyme: an ally against metabolic and neurodegenerative diseases. J Pathol 2021; 255:346-361. [PMID: 34396529 DOI: 10.1002/path.5777] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/01/2021] [Accepted: 08/09/2021] [Indexed: 11/11/2022]
Abstract
Insulin-degrading enzyme (IDE) function goes far beyond its known proteolytic role as a regulator of insulin levels. IDE has a wide substrate promiscuity, degrading several proteins such as amyloid-β peptide, glucagon, islet amyloid polypeptide (IAPP) and insulin-like growth factors, that have diverse physiological and pathophysiological functions. Importantly, IDE plays other non-proteolytical functions such as a chaperone/dead-end chaperone, an E1-ubiquitin activating enzyme, and a proteasome modulator. It also responds as a heat shock protein, regulating cellular proteostasis. Notably, amyloidogenic proteins such as IAPP, amyloid-β and α-synuclein have been reported as substrates for IDE chaperone activity. This is of utmost importance as failure of IDE may result in increased protein aggregation, a key hallmark in the pathogenesis of beta cells in type 2 diabetes mellitus and of neurons in neurodegenerative diseases such as Alzheimer's and Parkinson's disease. In this review, we focus on the biochemical and biophysical properties of IDE and the regulation of its physiological functions. We further raise the hypothesis that IDE plays a central role in the pathological context of dysmetabolic and neurodegenerative diseases and discuss its potential as a therapeutic target. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Luís Sousa
- CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Mariana Guarda
- CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| | - Maria João Meneses
- CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal.,APDP-Diabetes Portugal Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - M Paula Macedo
- CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal.,APDP-Diabetes Portugal Education and Research Center (APDP-ERC), Lisbon, Portugal.,Departamento de Ciências Médicas, Instituto de Biomedicina - iBiMED, Universidade de Aveiro, Aveiro, Portugal
| | - Hugo Vicente Miranda
- CEDOC, NOVA Medical School, NMS, Universidade Nova de Lisboa, 1169-056, Lisbon, Portugal
| |
Collapse
|
7
|
Ding D, Wang X, Li Q, Li L, Wu J. Research on the Glial-Lymphatic System and Its Relationship With Alzheimer's Disease. Front Neurosci 2021; 15:605586. [PMID: 34220413 PMCID: PMC8242204 DOI: 10.3389/fnins.2021.605586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
Metabolic waste clearance is essential to maintain body homeostasis, in which the lymphatic system plays a vital role. Conversely, in recent years, studies have identified the glial-lymphatic system in the brain, which primarily comprises the inflow of fluid along the para-arterial space. Aquaporin-4 mediates the convection of interstitial fluid in the brain and outflow along the paravenous space. β-Amyloid deposition is a characteristic pathological change in Alzheimer's disease, and some studies have found that the glial-lymphatic system plays an important role in its clearance. Thus, the glial-lymphatic system may influence Alzheimer's disease severity and outcome; therefore, this review summarizes the current and available research on the glial-lymphatic system and Alzheimer's disease.
Collapse
Affiliation(s)
- Danhua Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyu Wang
- Department of Rheumatology, Peking University Third Hospital, Beijing, China
| | - Qianqian Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lanjun Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Sofer Y, Nash Y, Osher E, Fursht O, Goldsmith G, Nahary L, Shaklai S, Tordjman KM, Serebro M, Touati EB, Yacobi Bach M, Marcus Y, Tal B, Sack J, Shefer G, Margaliot M, Landis N, Goldiner I, Abu Ahmad W, Stern N, Benhar I, Frenkel D. Insulin-degrading enzyme higher in subjects with metabolic syndrome. Endocrine 2021; 71:357-364. [PMID: 33398768 DOI: 10.1007/s12020-020-02548-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022]
Abstract
Metabolic syndrome (MS) is comprised of a cluster of abnormalities in glucose, lipid, and vascular homeostasis, which is most commonly linked to abdominal obesity. MS heralds increased risk for development of diabetes and is linked to impairment in insulin signaling. Insulin-degrading enzyme (IDE) is one of the mechanisms through which insulin blood levels are maintained. It has been previously suggested that controlling IDE levels could provide yet another potential therapeutic approach in diabetes. Here we aim to investigate whether changes in serum IDE levels correlate with the severity of MS. Using a highly sensitive ELISA assay of active IDE in human serum, we found a strong correlation between circulating IDE levels and circulating levels of triglycerides, insulin, and c-peptide and an inverse correlation with HDL cholesterol (HDLc). Serum IDE levels were higher in MS subjects than in control subjects. Hence, circulating IDE may serve as a tool to identify subjects with abnormal insulin metabolism, possibly those with MS that are at risk to develop diabetes.
Collapse
Affiliation(s)
- Y Sofer
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel.
| | - Y Nash
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - E Osher
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - O Fursht
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - G Goldsmith
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - L Nahary
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - S Shaklai
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - K M Tordjman
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - M Serebro
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - E B Touati
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - M Yacobi Bach
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Y Marcus
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - B Tal
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - J Sack
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - G Shefer
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - M Margaliot
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - N Landis
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - I Goldiner
- Laboratory Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - W Abu Ahmad
- Hebrew University-Hadassah Braun School of Public Health and Community Medicine, Jerusalem, Israel
| | - N Stern
- Institute of Endocrinology, Metabolism and Hypertension and The Sagol Center for Epigenetics of Metabolism and Aging, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - I Benhar
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - D Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, 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
|
Abstract
Aging is characterized by a progressive loss of physiological function leading to increase in the vulnerability to death. This deterioration process occurs in all living organisms and is the primary risk factor for pathological conditions including obesity, type 2 diabetes mellitus, Alzheimer's disease and cardiovascular diseases. Most of the age-related diseases have been associated with impairment of action of an important hormone, namely insulin. It is well-known that this hormone is a critical mediator of metabolism, growth, proliferation and differentiation. Insulin action depends on two processes that determine its circulating levels, insulin secretion and clearance, and insulin sensitivity in its target tissues. Aging has deleterious effects on these three mechanisms, impairing insulin action, thereby increasing the risk for diseases and death. Thus, improving insulin action may be an important strategy to have a healthier and longer life.
Collapse
|
10
|
Yoo SK, Kang JY, Lee U, Park SK, Kim JM, Han HJ, Kim DO, Heo HJ. Improving effect of Actinidia arguta leaf on hyperglycemia-induced cognitive dysfunction. J Funct Foods 2021. [DOI: 10.1016/j.jff.2020.104315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
11
|
Marmentini C, Soares GM, Bronczek GA, Piovan S, Mareze-Costa CE, Carneiro EM, Boschero AC, Kurauti MA. Aging Reduces Insulin Clearance in Mice. Front Endocrinol (Lausanne) 2021; 12:679492. [PMID: 34054736 PMCID: PMC8150109 DOI: 10.3389/fendo.2021.679492] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Hyperinsulinemia is frequently associated with aging and may cause insulin resistance in elderly. Since insulin secretion and clearance decline with age, hyperinsulinemia seems to be maintained, primarily, due to a decrease in the insulin clearance. To investigate these aging effects, 3- and 18-month-old male C57BL/6 mice were subjected to intraperitoneal glucose and insulin tolerance tests (ipGTT and ipITT) and, during the ipGTT, plasma c-peptide and insulin were measure to evaluate in vivo insulin clearance. Glucose-stimulated insulin secretion in isolated pancreatic islets was also assessed, and liver samples were collected for molecular analyses (western blot). Although insulin sensitivity was not altered in the old mice, glucose tolerance, paradoxically, seems to be increased, accompanied by higher plasma insulin, during ipGTT. While insulin secretion did not increase, insulin clearance was reduced in the old mice, as suggested by the lower c-peptide:insulin ratio, observed during ipGTT. Carcinoembryonic antigen-related cell adhesion molecule-1 (CEACAM1) and insulin-degrading enzyme (IDE), as well as the activity of this enzyme, were reduced in the liver of old mice, justifying the decreased insulin clearance observed in these mice. Therefore, loss of hepatic CEACAM1 and IDE function may be directly related to the decline in insulin clearance during aging.
Collapse
Affiliation(s)
- Carine Marmentini
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela M. Soares
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Gabriela A. Bronczek
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Silvano Piovan
- Department of Physiological Sciences, Biological Sciences Center, State University of Maringa (UEM), Maringa, Brazil
| | - Cecília E. Mareze-Costa
- Department of Physiological Sciences, Biological Sciences Center, State University of Maringa (UEM), Maringa, Brazil
| | - Everardo M. Carneiro
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Antonio C. Boschero
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
| | - Mirian A. Kurauti
- Laboratory of Endocrine Pancreas and Metabolism, Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas, Brazil
- Department of Physiological Sciences, Biological Sciences Center, State University of Maringa (UEM), Maringa, Brazil
- *Correspondence: Mirian A. Kurauti, ;
| |
Collapse
|
12
|
Microglia Do Not Take Up Soluble Amyloid-beta Peptides, But Partially Degrade Them by Secreting Insulin-degrading Enzyme. Neuroscience 2020; 443:30-43. [PMID: 32697980 DOI: 10.1016/j.neuroscience.2020.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
Abstract
Microglia play important roles in the pathogenesis of Alzheimer's disease (AD), in part, by affecting the clearance of amyloid-β (Aβ) peptides. Most studies, however, used synthetic soluble Aβ (sAβ) at higher concentrations. The exact mechanisms underlying microglia-mediated clearance of physiological sAβ at very low concentrations remain unclear. Here we reported that there were much more Iba-1- and CD68-positive microglia and significantly less sAβ left in the brain of adult mice 5 days after the surgery of sAβ microinjection compared to 2 h after the surgery (p < 0.05). However, very few Iba-1- and CD68-positive microglia co-localized with microinjected fluorescently labeled sAβ (FLsAβ42) 5 days after the surgery. Also, there was no co-localization of FLsAβ42 with a lysosomal marker (LAMP-1) 5 days after the surgery. There was no significant difference in the percentage of Aβ+/PE-CD11b+/APC-CD45low microglia between the control group and the group microinjected with TBS-soluble Aβ extracted from the brains of AD patients (p > 0.05). The degradation of physiological sAβ was prevented by a highly selective insulin-degrading enzyme inhibitor (Ii1) but not by a phagocytosis inhibitor (polyinosinic acid) or pinocytosis inhibitor (cytochalasin B) in vitro. Furthermore, the reduction of synthetic and physiological sAβ in the brain was partially prevented by the co-injection of Ii1 in vivo (p < 0.05). Our results demonstrate that microglia do not take up synthetic or physiological sAβ, but partially degrade it via the secretion of insulin-degrading enzyme, which will be beneficial for understanding how sAβ is removed from the brain by microglia.
Collapse
|
13
|
Jash K, Gondaliya P, Kirave P, Kulkarni B, Sunkaria A, Kalia K. Cognitive dysfunction: A growing link between diabetes and Alzheimer's disease. Drug Dev Res 2020; 81:144-164. [DOI: 10.1002/ddr.21579] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/12/2019] [Accepted: 06/30/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Kavya Jash
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| | - Piyush Gondaliya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| | - Prathibha Kirave
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| | - Bhagyashri Kulkarni
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| | - Aditya Sunkaria
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Ahmedabad Gandhinagar Gujarat India
| |
Collapse
|
14
|
Nalivaeva NN, Turner AJ. Targeting amyloid clearance in Alzheimer's disease as a therapeutic strategy. Br J Pharmacol 2019; 176:3447-3463. [PMID: 30710367 PMCID: PMC6715594 DOI: 10.1111/bph.14593] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/08/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022] Open
Abstract
Targeting the amyloid-β (Aβ) peptide cascade has been at the heart of therapeutic developments in Alzheimer's disease (AD) research for more than 25 years, yet no successful drugs have reached the marketplace based on this hypothesis. Nevertheless, the genetic and other evidence remains strong, if not overwhelming, that Aβ is central to the disease process. Most attention has focused on the biosynthesis of Aβ from its precursor protein through the successive actions of the β- and γ-secretases leading to the development of inhibitors of these membrane proteases. However, the levels of Aβ are maintained through a balance of its biosynthesis and clearance, which occurs both through further proteolysis by a family of amyloid-degrading enzymes (ADEs) and by a variety of transport processes. The development of late-onset AD appears to arise from a failure of these clearance mechanisms rather than by overproduction of the peptide. This review focuses on the nature of these clearance mechanisms, particularly the various proteases known to be involved, and their regulation and potential as therapeutic targets in AD drug development. The majority of the ADEs are zinc metalloproteases [e.g., the neprilysin (NEP) family, insulin-degrading enzyme, and angiotensin converting enzymes (ACE)]. Strategies for up-regulating the expression and activity of these enzymes, such as genetic, epigenetic, stem cell technology, and other pharmacological approaches, will be highlighted. Modifiable physiological mechanisms affecting the efficiency of Aβ clearance, including brain perfusion, obesity, diabetes, and sleep, will also be outlined. These new insights provide optimism for future therapeutic developments in AD research. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Natalia N. Nalivaeva
- School of Biomedical SciencesUniversity of LeedsLeedsUK
- Laboratory of Physiology and Pathology of CNSI.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of RASSt. PetersburgRussia
| | | |
Collapse
|
15
|
Delikkaya B, Moriel N, Tong M, Gallucci G, de la Monte SM. Altered expression of insulin-degrading enzyme and regulator of calcineurin in the rat intracerebral streptozotocin model and human apolipoprotein E-ε4-associated Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:392-404. [PMID: 31193223 PMCID: PMC6522644 DOI: 10.1016/j.dadm.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION This study assesses insulin-degrading enzyme (IDE) and regulator of calcineurin 1 (RCAN1) as potential mediators of brain insulin deficiency and neurodegeneration in experimental and human Alzheimer's disease (AD). METHODS Temporal lobes from Long Evans rats treated with intracerebral streptozotocin or vehicle and postmortem frontal lobes from humans with normal aging AD (Braak 0-2), moderate (Braak 3-4) AD, or advanced (Braak 5-6) AD were used to measure IDE and RCAN mRNA and protein. RESULTS Intracerebral streptozotocin significantly increased IDE and RCAN mRNA and protein. In humans with apolipoprotein E (ApoE) ε3/ε4 or ε4/ε4 and AD, IDE was elevated at Braak 3-4, but at Braak 5-6, IDE expression was significantly reduced. RCAN1 mRNA was similarly reduced in ApoE ε4+ patients with moderate or severe AD, whereas RCAN1 protein declined with the severity of AD and ApoE ε4 dose. DISCUSSION The findings suggest that IDE and RCAN1 differentially modulate brain insulin signaling in relation to AD severity and ApoE genotype.
Collapse
Affiliation(s)
- Büşra Delikkaya
- Istanbul University-Cerrahpasa Cerrahpasa Medical Faculty, Istanbul, Turkey
| | - Natalia Moriel
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Alpert Medical School of Brown University, Providence, RI, USA
| | - Gina Gallucci
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA
| | - Suzanne M. de la Monte
- Department of Medicine, Rhode Island Hospital, Providence, RI, USA,Alpert Medical School of Brown University, Providence, RI, USA,Departments of Neurology and Neurosurgery, Rhode Island Hospital, Providence, RI, USA,Department of Pathology and Laboratory Medicine, Providence VA Medical Center, Providence, RI, USA,Corresponding author. Tel.: +401-444-7364; Fax: +401-444-2939.
| |
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
|
Sbardella D, Tundo GR, Coletta A, Marcoux J, Koufogeorgou EI, Ciaccio C, Santoro AM, Milardi D, Grasso G, Cozza P, Bousquet-Dubouch MP, Marini S, Coletta M. The insulin-degrading enzyme is an allosteric modulator of the 20S proteasome and a potential competitor of the 19S. Cell Mol Life Sci 2018; 75:3441-3456. [PMID: 29594388 PMCID: PMC11105570 DOI: 10.1007/s00018-018-2807-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/12/2018] [Accepted: 03/22/2018] [Indexed: 01/09/2023]
Abstract
The interaction of insulin-degrading enzyme (IDE) with the main intracellular proteasome assemblies (i.e, 30S, 26S and 20S) was analyzed by enzymatic activity, mass spectrometry and native gel electrophoresis. IDE was mainly detected in association with assemblies with at least one free 20S end and biochemical investigations suggest that IDE competes with the 19S in vitro. IDE directly binds the 20S and affects its proteolytic activities in a bimodal fashion, very similar in human and yeast 20S, inhibiting at (IDE) ≤ 30 nM and activating at (IDE) ≥ 30 nM. Only an activating effect is observed in a yeast mutant locked in the "open" conformation (i.e., the α-3ΔN 20S), envisaging a possible role of IDE as modulator of the 20S "open"-"closed" allosteric equilibrium. Protein-protein docking in silico proposes that the interaction between IDE and the 20S could involve the C-term helix of the 20S α-3 subunit which regulates the gate opening of the 20S.
Collapse
Affiliation(s)
- Diego Sbardella
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Interuniversitary Center for the Research on the Chemistry of Metals in Biological Systems, Bari, Italy
- Interdepartmental Center for TeleInfrastructures, University of Roma Tor Vergata, Rome, Italy
| | - Grazia R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Interuniversitary Center for the Research on the Chemistry of Metals in Biological Systems, Bari, Italy
| | - Andrea Coletta
- Department of Chemistry, Aarhus University, Aarhus, Denmark
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Chiara Ciaccio
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Interuniversitary Center for the Research on the Chemistry of Metals in Biological Systems, Bari, Italy
| | - Anna M Santoro
- Institute of Biostructures and Bioimaging, National Research Council, Catania, Italy
| | - Danilo Milardi
- Institute of Biostructures and Bioimaging, National Research Council, Catania, Italy
| | - Giuseppe Grasso
- Department of Chemistry, University of Catania, Catania, Italy
| | - Paola Cozza
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Interdepartmental Center for TeleInfrastructures, University of Roma Tor Vergata, Rome, Italy
| | | | - Stefano Marini
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
- Interuniversitary Center for the Research on the Chemistry of Metals in Biological Systems, Bari, Italy
- Interdepartmental Center for TeleInfrastructures, University of Roma Tor Vergata, Rome, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Via Montpellier 1, 00133, Rome, Italy.
- Interuniversitary Center for the Research on the Chemistry of Metals in Biological Systems, Bari, Italy.
- Interdepartmental Center for TeleInfrastructures, University of Roma Tor Vergata, Rome, Italy.
| |
Collapse
|
18
|
Li H, Cao L, Ren Y, Jiang Y, Xie W, Li D. GLP-1 receptor regulates cell growth through regulating IDE expression level in Aβ1-42-treated PC12 cells. Biosci Rep 2018; 38:BSR20171284. [PMID: 29263141 PMCID: PMC6043719 DOI: 10.1042/bsr20171284] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 12/25/2022] Open
Abstract
This study aimed to validate whether glucagon-like peptide-1 receptor (GLP-1R) / cyclic adenosine monophosphate (cAMP) / protein kinase (PKA) / insulin-degrading enzyme (IDE) signaling pathway was associated with neuronal apoptosis. We developed an animal model presenting both Alzheimer's disease (AD) and type 2 diabetes (T2D), by crossing APP/PS1 mice (AD model) with streptozotocin (STZ)-treated mice (a T2D model). Neuronal apoptosis was detected by TUNEL staining and the expression levels of apoptosis-related proteins were examined by Western blotting. The viability of PC12 cells was analyzed by MTT assay and apoptosis of PC12 cells was detected by flow cytometry. The mRNA expression level was detected by qRT-PCR. T2D contributes to AD progress by prompting neuronal apoptosis and increasing expression of pro-apoptotic protein. β-Amyloid peptide1-42 (Aβ1-42) was shown to exert effects on inhibiting cell viability and prompting cell apoptosis of PC12 cells. However, GLP-1R agonist geniposide (Gen) significantly reversed them, exerting a protective role on PC12 cells. And IDE antagonist bacitracin (Bac) markedly reversed the protective effects of Gen on Aβ1-42-treated PC12 cells. Besides, Gen significantly reversed the effects of Aβ1-42 treatment on IDE expression, and the inhibitor of cAMP/PKA signaling pathway markedly reversed the effects of Gen on IDE expression level in Aβ1-42-treated PC12 cells. In conclusion, GLP-1R regulates cell growth, at least partially, through regulating cAMP/PKA/IDE signaling pathway in Aβ1-42-treated PC12 cells.
Collapse
Affiliation(s)
- Huajie Li
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| | - Liping Cao
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| | - Yi Ren
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| | - Ying Jiang
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| | - Wei Xie
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| | - Dawen Li
- Department of Neurology, the First People's Hospital of Chang Zhou, Jiang Su 213003, China
| |
Collapse
|
19
|
Yang D, Qin W, Shi X, Zhu B, Xie M, Zhao H, Teng B, Wu Y, Zhao R, Yin F, Ren P, Liu L, Li Z. Stabilized β-Hairpin Peptide Inhibits Insulin Degrading Enzyme. J Med Chem 2018; 61:8174-8185. [DOI: 10.1021/acs.jmedchem.8b00418] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Dan Yang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
- Department of Science & Technology of Shandong Province, Jinan 250101, Shandong, China
| | - Weirong Qin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Xiaodong Shi
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Bili Zhu
- School of Medicine, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Mingsheng Xie
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Hui Zhao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Bin Teng
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Yujie Wu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Rongtong Zhao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| | - Peigen Ren
- Center for Translational Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Lizhong Liu
- School of Medicine, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, Guangdong, China
| |
Collapse
|
20
|
Novosadová Z, Polidarová L, Sládek M, Sumová A. Alteration in glucose homeostasis and persistence of the pancreatic clock in aged mPer2 Luc mice. Sci Rep 2018; 8:11668. [PMID: 30076390 PMCID: PMC6076295 DOI: 10.1038/s41598-018-30225-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/23/2018] [Indexed: 12/16/2022] Open
Abstract
The physiological function of the pancreas is controlled by the circadian clock. The aim of this study was to determine whether aging-induced changes in glucose homeostasis affect properties of the circadian clock in the pancreas and/or its sensitivity to disturbances in environmental lighting conditions. mPer2Luc mice aged 24-26 months developed hyperinsulinemic hypoglycaemia, which was likely due to the Pclo-mediated insulin hyper-secretion and Slc2a2-mediated glucose transport impairment in the pancreas, and due to the alterations in Pp1r3c-related glycogen storage and Sgk1-related glucose transport in the liver. In the pancreatic tissue, aging affected clock gene expression only marginally, it upregulated Bmal1 and downregulated Clock expression. Whereas aging significantly impaired the circadian clock in lung explants, which were used as a control tissue, the properties of the pancreatic clock in vitro were not affected. The data suggest a non-circadian role of Bmal1 in changes of pancreatic function that occur during aging. Additionally, the pancreatic clock was more sensitive to exposure of animals to constant light conditions. These findings provide an explanation for the previously demonstrated relationship between disturbances in the circadian system and disordered glucose homeostasis, including diabetes mellitus type 2, in subjects exposed to long-term shift work.
Collapse
Affiliation(s)
- Zuzana Novosadová
- Department of Neurohumoral Regulations, Institute of Physiology, the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Sciences, Charles University, Prague, Czech Republic
| | - Lenka Polidarová
- Department of Neurohumoral Regulations, Institute of Physiology, the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Sládek
- Department of Neurohumoral Regulations, Institute of Physiology, the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Sumová
- Department of Neurohumoral Regulations, Institute of Physiology, the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
21
|
Rotermund C, Machetanz G, Fitzgerald JC. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front Endocrinol (Lausanne) 2018; 9:400. [PMID: 30072954 PMCID: PMC6060268 DOI: 10.3389/fendo.2018.00400] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/27/2018] [Indexed: 12/12/2022] Open
Abstract
The search for treatments for neurodegenerative diseases is a major concern in light of today's aging population and an increasing burden on individuals, families, and society. Although great advances have been made in the last decades to understand the underlying genetic and biological cause of these diseases, only some symptomatic treatments are available. Metformin has long since been used to treat Type 2 Diabetes and has been shown to be beneficial in several other conditions. Metformin is well-tested in vitro and in vivo and an approved compound that targets diverse pathways including mitochondrial energy production and insulin signaling. There is growing evidence for the benefits of metformin to counteract age-related diseases such as cancer, cardiovascular disease, and neurodegenerative diseases. We will discuss evidence showing that certain neurodegenerative diseases and diabetes are explicitly linked and that metformin along with other diabetes drugs can reduce neurological symptoms in some patients and reduce disease phenotypes in animal and cell models. An interesting therapeutic factor might be how metformin is able to balance survival and death signaling in cells through pathways that are commonly associated with neurodegenerative diseases. In healthy neurons, these overarching signals keep energy metabolism, oxidative stress, and proteostasis in check, avoiding the dysfunction and neuronal death that defines neurodegenerative disease. We will discuss the biological mechanisms involved and the relevance of neuronal vulnerability and potential difficulties for future trials and development of therapies.
Collapse
Affiliation(s)
| | - Gerrit Machetanz
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Julia C. Fitzgerald
- German Centre for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
Rotermund C, Machetanz G, Fitzgerald JC. The Therapeutic Potential of Metformin in Neurodegenerative Diseases. Front Endocrinol (Lausanne) 2018; 9:400. [PMID: 30072954 DOI: 10.3389/fendo.2018.00400/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/27/2018] [Indexed: 05/25/2023] Open
Abstract
The search for treatments for neurodegenerative diseases is a major concern in light of today's aging population and an increasing burden on individuals, families, and society. Although great advances have been made in the last decades to understand the underlying genetic and biological cause of these diseases, only some symptomatic treatments are available. Metformin has long since been used to treat Type 2 Diabetes and has been shown to be beneficial in several other conditions. Metformin is well-tested in vitro and in vivo and an approved compound that targets diverse pathways including mitochondrial energy production and insulin signaling. There is growing evidence for the benefits of metformin to counteract age-related diseases such as cancer, cardiovascular disease, and neurodegenerative diseases. We will discuss evidence showing that certain neurodegenerative diseases and diabetes are explicitly linked and that metformin along with other diabetes drugs can reduce neurological symptoms in some patients and reduce disease phenotypes in animal and cell models. An interesting therapeutic factor might be how metformin is able to balance survival and death signaling in cells through pathways that are commonly associated with neurodegenerative diseases. In healthy neurons, these overarching signals keep energy metabolism, oxidative stress, and proteostasis in check, avoiding the dysfunction and neuronal death that defines neurodegenerative disease. We will discuss the biological mechanisms involved and the relevance of neuronal vulnerability and potential difficulties for future trials and development of therapies.
Collapse
Affiliation(s)
| | - Gerrit Machetanz
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Julia C Fitzgerald
- German Centre for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neurodegenerative Diseases, Centre of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Xin SH, Tan L, Cao X, Yu JT, Tan L. Clearance of Amyloid Beta and Tau in Alzheimer's Disease: from Mechanisms to Therapy. Neurotox Res 2018; 34:733-748. [PMID: 29626319 DOI: 10.1007/s12640-018-9895-1] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/02/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Pathological proteins of AD mainly contain amyloid-beta (Aβ) and tau. Their deposition will lead to neuron damage by a series of pathways, and then induce memory and cognitive impairment. Thus, it is pivotal to understand the clearance pathways of Aβ and tau in order to delay or even halt AD. Aβ clearance mechanisms include ubiquitin-proteasome system, autophagy-lysosome, proteases, microglial phagocytosis, and transport from the brain to the blood via the blood-brain barrier (BBB), arachnoid villi and blood-CSF barrier, which can be named blood circulatory clearance. Recently, lymphatic clearance has been demonstrated to play a key role in transport of Aβ into cervical lymph nodes. The discovery of meningeal lymphatic vessels is another direct evidence for lymphatic clearance in the brain. Furthermore, periphery clearance also contributes to Aβ clearance. Tau clearance is almost the same as Aβ clearance. In this review, we will mainly introduce the clearance mechanisms of Aβ and tau proteins, and summarize corresponding targeted drug therapies for AD.
Collapse
Affiliation(s)
- Shu-Hui Xin
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, Shandong, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, Shandong, China
| | - Xipeng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, Shandong, China. .,Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, No.5 Donghai Middle Road, Qingdao, 266071, Shandong, China.
| |
Collapse
|
24
|
Insulin degrading enzyme contributes to the pathology in a mixed model of Type 2 diabetes and Alzheimer's disease: possible mechanisms of IDE in T2D and AD. Biosci Rep 2018; 38:BSR20170862. [PMID: 29222348 PMCID: PMC6435468 DOI: 10.1042/bsr20170862] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 02/03/2023] Open
Abstract
Insulin degrading enzyme (IDE) is believed to act as a junction point of Type 2 diabetes (T2D) and Alzheimer's disease (AD); however, the underlying mechanism was not completely clear yet. Transgenic APPSwe/PS1 mice were used as the AD model and were treated with streptozocin/streptozotocin (STZ) to develop a mixed mice model presenting both AD and T2D. Morris Water Maze (MWM) and recognition task were performed to trace the cognitive function. The detection of fasting plasma glucose (FPG) and plasma insulin concentration, and oral glucose tolerance test (OGTT) were used to trace the metabolism evolution. Aβ40 and Aβ42 were quantified by colorimetric ELISA kits. The mRNA or protein expression levels were determined by quantitative real-time RT-PCR and Western blotting analysis respectively. T2D contributes to the AD progress by accelerating and worsening spatial learning and recognition impairments. Metabolic parameters and glucose tolerance were significantly changed in the presence of the AD and T2D. The expression levels of IDE, PPARγ, and AMPK were down-regulated in mice with AD and T2D. PPARγ activator rosiglitazone (RSZ) or AMPK activator AICAR increased the expression level of IDE and decreased Aβ levels in mice with AD and T2D. RSZ or AICAR treatment also alleviated the spatial learning and recognition impairments in AD and T2D mice. Our results found that, in the mice with T2D and AD, the activators of PPARγ/AMPK signaling pathway significantly increased the expression level of IDE, and decreased the accumulation of Aβ40 and Aβ42, as well as alleviated the spatial learning and recognition impairments.
Collapse
|
25
|
Li H, Yang S, Wu J, Ji L, Zhu L, Cao L, Huang J, Jiang Q, Wei J, Liu M, Mao K, Wei N, Xie W, Yang Z. cAMP/PKA signaling pathway contributes to neuronal apoptosis via regulating IDE expression in a mixed model of type 2 diabetes and Alzheimer's disease. J Cell Biochem 2017; 119:1616-1626. [PMID: 28771808 DOI: 10.1002/jcb.26321] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/02/2017] [Indexed: 12/30/2022]
Abstract
Type 2 diabetes (T2D) may play a relevant role in the development of Alzheimer's disease (AD), however, the underlying mechanism was not clear yet. We developed an animal model presenting both AD and T2D, morris water maze (MWM) test and recognition task were performed to trace the cognitive function. Fasting plasma glucose (FPG) and oral glucose tolerance test (OGTT) were determined to trace the metabolism evolution. TUNEL assay and apoptosis-related protein levels were analyzed for the detection of neuronal apoptosis. Cyclic adenosine monophosphate (cAMP) agonist bucladesine or protein kinase (PKA) inhibitor H-89 were used to determine the effects of cAMP/PKA signaling pathway on IDE expression and neuronal apoptosis. The results showed that T2D contributes to the AD progress by accelerating and worsening spatial memory and recognition dysfunctions. Metabolic parameters and glucose tolerance were significantly changed in the presence of the AD and T2D. The significantly induced neuronal apoptosis and increased pro-apoptotic proteins in mice with AD and T2D were also observed. We showed the decreased expression level of IDE and the activating of cAMP/PKA signaling pathway in AD and T2D mice. Further studies indicated that cAMP agonist decreased the expression level of IDE and induced the neuronal apoptosis in mice with AD and T2D; whereas PKA inhibitor H-89 treatment showed the completely opposite results. Our study indicated that, in the T2D and AD mice, cAMP/PKA signaling pathway and IDE may participate in the contribute role of T2D in accelerating the pathological process of AD via causing the accumulation of Aβ and neuronal apoptosis.
Collapse
Affiliation(s)
- Huajie Li
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Song Yang
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Jian Wu
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Lei Ji
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Linfeng Zhu
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Liping Cao
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Jinzhong Huang
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Qingqing Jiang
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Jiang Wei
- Department of General Laboratory, The First People's Hospital of Changzhou, Jiangsu, China
| | - Meng Liu
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Keshi Mao
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Ning Wei
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Wei Xie
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| | - Zhilong Yang
- Department of Neurology, The First People's Hospital of Changzhou, Jiangsu, China
| |
Collapse
|
26
|
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
|
27
|
Song MK, Bischoff DS, Song AM, Uyemura K, Yamaguchi DT. Metabolic relationship between diabetes and Alzheimer's Disease affected by Cyclo(His-Pro) plus zinc treatment. BBA CLINICAL 2017; 7:41-54. [PMID: 28070499 PMCID: PMC5219633 DOI: 10.1016/j.bbacli.2016.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 09/28/2016] [Accepted: 09/29/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Association of Alzheimer's Disease (AD) with Type 2 Diabetes (T2D) has been well established. Cyclo(His-Pro) plus zinc (Cyclo-Z) treatment ameliorated diabetes in rats and similar improvements have been seen in human patients. Treatment of amyloid precursor protein (APP) transgenic mice with Cyclo-Z exhibited memory improvements and significantly reduced Aβ-40 and Aβ-42 protein levels in the brain tissues of the mice. SCOPE OF REVIEW Metabolic relationship between AD and T2D will be described with particular attention to insulin sensitivity and Aβ degradation in brain and plasma tissues. Mechanistic effect of insulin degrading enzyme (IDE) in decreasing blood glucose and brain Aβ levels will be elucidated. Cyclo-Z effects on these biochemical parameters will be discussed. MAJOR CONCLUSION Stimulation of IDE synthesis is effective for the clinical treatment of metabolic diseases including AD and T2D. GENERAL SIGNIFICANCE Cyclo-Z might be the effective treatment of AD and T2D by stimulating IDE synthesis.
Collapse
Affiliation(s)
- Moon K. Song
- VA Greater Los Angeles Healthcare System, 16111, Plummer Street, North Hills, CA 91343
- UCLA School of Medicine, 1O833 Le Conte Avenue, Los Angeles, CA 90095
| | - David S. Bischoff
- VA Greater Los Angeles Healthcare System, 16111, Plummer Street, North Hills, CA 91343
- UCLA School of Medicine, 1O833 Le Conte Avenue, Los Angeles, CA 90095
| | - Albert M. Song
- Kaiser Permanente Medical Center, 13651 Willard Street, Panorama City, CA 91402
| | - Koichi Uyemura
- VA Greater Los Angeles Healthcare System, 16111, Plummer Street, North Hills, CA 91343
| | - Dean T. Yamaguchi
- VA Greater Los Angeles Healthcare System, 16111, Plummer Street, North Hills, CA 91343
- UCLA School of Medicine, 1O833 Le Conte Avenue, Los Angeles, CA 90095
| |
Collapse
|
28
|
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
|
29
|
Schilling MA. Unraveling Alzheimer's: Making Sense of the Relationship between Diabetes and Alzheimer's Disease1. J Alzheimers Dis 2016; 51:961-77. [PMID: 26967215 PMCID: PMC4927856 DOI: 10.3233/jad-150980] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2016] [Indexed: 12/11/2022]
Abstract
Numerous studies have documented a strong association between diabetes and Alzheimer's disease (AD). The nature of the relationship, however, has remained a puzzle, in part because of seemingly incongruent findings. For example, some studies have concluded that insulin deficiency is primarily at fault, suggesting that intranasal insulin or inhibiting the insulin-degrading enzyme (IDE) could be beneficial. Other research has concluded that hyperinsulinemia is to blame, which implies that intranasal insulin or the inhibition of IDE would exacerbate the disease. Such antithetical conclusions pose a serious obstacle to making progress on treatments. However, careful integration of multiple strands of research, with attention to the methods used in different studies, makes it possible to disentangle the research on AD. This integration suggests that there is an important relationship between insulin, IDE, and AD that yields multiple pathways to AD depending on the where deficiency or excess in the cycle occurs. I review evidence for each of these pathways here. The results suggest that avoiding excess insulin, and supporting robust IDE levels, could be important ways of preventing and lessening the impact of AD. I also describe what further tests need to be conducted to verify the arguments made in the paper, and their implications for treating AD.
Collapse
|
30
|
Kerridge C, Kozlova DI, Nalivaeva NN, Turner AJ. Hypoxia Affects Neprilysin Expression Through Caspase Activation and an APP Intracellular Domain-dependent Mechanism. Front Neurosci 2015; 9:426. [PMID: 26617481 PMCID: PMC4643132 DOI: 10.3389/fnins.2015.00426] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
While gene mutations in the amyloid precursor protein (APP) and the presenilins lead to an accumulation of the amyloid β-peptide (Aβ) in the brain causing neurodegeneration and familial Alzheimer's disease (AD), over 95% of all AD cases are sporadic. Despite the pathologies being indistinguishable, relatively little is known about the mechanisms affecting generation of Aβ in the sporadic cases. Vascular disorders such as ischaemia and stroke are well established risk factors for the development of neurodegenerative diseases and systemic hypoxic episodes have been shown to increase Aβ production and accumulation. We have previously shown that hypoxia causes a significant decrease in the expression of the major Aβ-degrading enzyme neprilysin (NEP) which might deregulate Aβ clearance. Aβ itself is derived from the transmembrane APP along with several other biologically active metabolites including the C-terminal fragment (CTF) termed the APP intracellular domain (AICD), which regulates the expression of NEP and some other genes in neuronal cells. Here we show that in hypoxia there is a significantly increased expression of caspase-3, 8, and 9 in human neuroblastoma NB7 cells, which can degrade AICD. Using chromatin immunoprecipitation we have revealed that there was also a reduction of AICD bound to the NEP promoter region which underlies the decreased expression and activity of the enzyme under hypoxic conditions. Incubation of the cells with a caspase-3 inhibitor Z-DEVD-FMK could rescue the effect of hypoxia on NEP activity protecting the levels of AICD capable of binding the NEP promoter. These data suggest that activation of caspases might play an important role in regulation of NEP levels in the brain under pathological conditions such as hypoxia and ischaemia leading to a deficit of Aβ clearance and increasing the risk of development of AD.
Collapse
Affiliation(s)
- Caroline Kerridge
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds Leeds, UK ; Neuroscience, Eli Lilly and Company Limited, Lilly Research Centre Surrey, UK
| | - Daria I Kozlova
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences St. Petersburg, Russia
| | - Natalia N Nalivaeva
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds Leeds, UK ; I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences St. Petersburg, Russia
| | - Anthony J Turner
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds Leeds, UK
| |
Collapse
|
31
|
New Insights into Epigenetic and Pharmacological Regulation of Amyloid-Degrading Enzymes. Neurochem Res 2015; 41:620-30. [PMID: 26376806 DOI: 10.1007/s11064-015-1703-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/12/2015] [Accepted: 08/18/2015] [Indexed: 10/23/2022]
Abstract
Currently, deficit of amyloid β-peptide (Aβ) clearance from the brain is considered as one of the possible causes of amyloid accumulation and neuronal death in the sporadic form of Alzheimer's disease (AD). Aβ clearance can involve either specific proteases present in the brain or Aβ-binding/transport proteins. Among amyloid-degrading enzymes the most intensively studied are neprilysin (NEP) and insulin-degrading enzyme (IDE). Since ageing and development of brain pathologies is often accompanied by a deficit in the levels of expression and activity of these enzymes in the brain, there is an urgent need to understand the mechanisms involved in their regulation. We have recently reported that NEP and also an Aβ-transport protein, transthyretin are epigenetically co-regulated by the APP intracellular domain (AICD) and this regulation depends on the cell type and APP695 isoform expression in a process that can be regulated by the tyrosine kinase inhibitor, Gleevec. We have now extended our work and shown that, unlike NEP, another amyloid-degrading enzyme, IDE, is not related to over-expression of APP695 in neuroblastoma SH-SY5Y cells but is up-regulated by APP751 and APP770 isoforms independently of AICD but correlating with reduced HDAC1 binding to its promoter. Studying the effect of the nuclear retinoid X receptor agonist, bexarotene, on NEP and IDE expression, we have found that both enzymes can be up-regulated by this compound but this mechanism is not APP-isoform specific and does not involve AICD but, on the contrary, affects HDAC1 occupancy on the NEP gene promoter. These new insights into the mechanisms of NEP and IDE regulation suggest possible pharmacological targets in developing AD therapies.
Collapse
|