1
|
Rhea EM, Leclerc M, Yassine HN, Capuano AW, Tong H, Petyuk VA, Macauley SL, Fioramonti X, Carmichael O, Calon F, Arvanitakis Z. State of the Science on Brain Insulin Resistance and Cognitive Decline Due to Alzheimer's Disease. Aging Dis 2024; 15:1688-1725. [PMID: 37611907 PMCID: PMC11272209 DOI: 10.14336/ad.2023.0814] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 08/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is common and increasing in prevalence worldwide, with devastating public health consequences. While peripheral insulin resistance is a key feature of most forms of T2DM and has been investigated for over a century, research on brain insulin resistance (BIR) has more recently been developed, including in the context of T2DM and non-diabetes states. Recent data support the presence of BIR in the aging brain, even in non-diabetes states, and found that BIR may be a feature in Alzheimer's disease (AD) and contributes to cognitive impairment. Further, therapies used to treat T2DM are now being investigated in the context of AD treatment and prevention, including insulin. In this review, we offer a definition of BIR, and present evidence for BIR in AD; we discuss the expression, function, and activation of the insulin receptor (INSR) in the brain; how BIR could develop; tools to study BIR; how BIR correlates with current AD hallmarks; and regional/cellular involvement of BIR. We close with a discussion on resilience to both BIR and AD, how current tools can be improved to better understand BIR, and future avenues for research. Overall, this review and position paper highlights BIR as a plausible therapeutic target for the prevention of cognitive decline and dementia due to AD.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA.
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
| | - Hussein N Yassine
- Departments of Neurology and Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Ana W Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Han Tong
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99352, USA.
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508, USA.
| | - Xavier Fioramonti
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France.
| | - Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Frederic Calon
- Faculty of Pharmacy, Laval University, Quebec, Quebec, Canada.
- Neuroscience Axis, CHU de Québec Research Center - Laval University, Quebec, Quebec, Canada.
- International Associated Laboratory OptiNutriBrain, Bordeaux, France and Quebec, Canada.
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
2
|
Abdulhameed N, Babin A, Hansen K, Weaver R, Banks WA, Talbot K, Rhea EM. Comparing regional brain uptake of incretin receptor agonists after intranasal delivery in CD-1 mice and the APP/PS1 mouse model of Alzheimer's disease. Alzheimers Res Ther 2024; 16:173. [PMID: 39085976 PMCID: PMC11293113 DOI: 10.1186/s13195-024-01537-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
Targeting brain insulin resistance (BIR) has become an attractive alternative to traditional therapeutic treatments for Alzheimer's disease (AD). Incretin receptor agonists (IRAs), targeting either or both of the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors, have proven to reverse BIR and improve cognition in mouse models of AD. We previously showed that many, but not all, IRAs can cross the blood-brain barrier (BBB) after intravenous (IV) delivery. Here we determined if widespread brain uptake of IRAs could be achieved by circumventing the BBB using intranasal (IN) delivery, which has the added advantage of minimizing adverse gastrointestinal effects of systemically delivered IRAs. Of the 5 radiolabeled IRAs tested (exenatide, dulaglutide, semaglutide, DA4-JC, and DA5-CH) in CD-1 mice, exenatide, dulaglutide, and DA4-JC were successfully distributed throughout the brain following IN delivery. We observed significant sex differences in uptake for DA4-JC. Dulaglutide and DA4-JC exhibited high uptake by the hippocampus and multiple neocortical areas. We further tested and found the presence of AD-associated Aβ pathology minimally affected uptake of dulaglutide and DA4-JC. Of the 5 tested IRAs, dulaglutide and DA4-JC are best capable of accessing brain regions most vulnerable in AD (neocortex and hippocampus) after IN administration. Future studies will need to be performed to determine if IN IRA delivery can reduce BIR in AD or animal models of that disorder.
Collapse
Affiliation(s)
- Noor Abdulhameed
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Alice Babin
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Kim Hansen
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - Riley Weaver
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA
| | - Konrad Talbot
- Departments of Neurosurgery, Pathology and Human Anatomy, and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98498, USA.
| |
Collapse
|
3
|
Zhou AL, Swaminathan SK, Salian VS, Wang L, Curran GL, Min HK, Lowe VJ, Kandimalla KK. Insulin Signaling Differentially Regulates the Trafficking of Insulin and Amyloid Beta Peptides at the Blood-Brain Barrier. Mol Pharm 2024; 21:2176-2186. [PMID: 38625027 PMCID: PMC11929044 DOI: 10.1021/acs.molpharmaceut.3c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
The blood-brain barrier (BBB) is instrumental in clearing toxic metabolites from the brain, such as amyloid-β (Aβ) peptides, and in delivering essential nutrients to the brain, like insulin. In Alzheimer's disease (AD) brain, increased Aβ levels are paralleled by decreased insulin levels, which are accompanied by insulin signaling deficits at the BBB. Thus, we investigated the impact of insulin-like growth factor and insulin receptor (IGF1R and IR) signaling on Aβ and insulin trafficking at the BBB. Following intravenous infusion of an IGF1R/IR kinase inhibitor (AG1024) in wild-type mice, the BBB trafficking of 125I radiolabeled Aβ peptides and insulin was assessed by dynamic SPECT/CT imaging. The brain efflux of [125I]iodo-Aβ42 decreased upon AG1024 treatment. Additionally, the brain influx of [125I]iodoinsulin, [125I]iodo-Aβ42, [125I]iodo-Aβ40, and [125I]iodo-BSA (BBB integrity marker) was decreased, increased, unchanged, and unchanged, respectively, upon AG1024 treatment. Subsequent mechanistic studies were performed using an in vitro BBB cell model. The cell uptake of [125I]iodoinsulin, [125I]iodo-Aβ42, and [125I]iodo-Aβ40 was decreased, increased, and unchanged, respectively, upon AG1024 treatment. Further, AG1024 reduced the phosphorylation of insulin signaling kinases (Akt and Erk) and the membrane expression of Aβ and insulin trafficking receptors (LRP-1 and IR-β). These findings reveal that insulin signaling differentially regulates the BBB trafficking of Aβ peptides and insulin. Moreover, deficits in IGF1R and IR signaling, as observed in the brains of type II diabetes and AD patients, are expected to increase Aβ accumulation while decreasing insulin delivery to the brain, which has been linked to the progression of cognitive decline in AD.
Collapse
Affiliation(s)
- Andrew L Zhou
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Vrishali S Salian
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Lushan Wang
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Geoffry L Curran
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, United States
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, United States
| | - Hoon-Ki Min
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, United States
| | - Val J Lowe
- Department of Radiology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, United States
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota College of Pharmacy, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [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] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
5
|
Wang J, Fu J, Zhao Y, Liu Q, Yan X, Su J. Iron and Targeted Iron Therapy in Alzheimer's Disease. Int J Mol Sci 2023; 24:16353. [PMID: 38003544 PMCID: PMC10671546 DOI: 10.3390/ijms242216353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. β-amyloid plaque (Aβ) deposition and hyperphosphorylated tau, as well as dysregulated energy metabolism in the brain, are key factors in the progression of AD. Many studies have observed abnormal iron accumulation in different regions of the AD brain, which is closely correlated with the clinical symptoms of AD; therefore, understanding the role of brain iron accumulation in the major pathological aspects of AD is critical for its treatment. This review discusses the main mechanisms and recent advances in the involvement of iron in the above pathological processes, including in iron-induced oxidative stress-dependent and non-dependent directions, summarizes the hypothesis that the iron-induced dysregulation of energy metabolism may be an initiating factor for AD, based on the available evidence, and further discusses the therapeutic perspectives of targeting iron.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (J.W.); (J.F.); (Y.Z.); (Q.L.); (X.Y.)
| |
Collapse
|
6
|
Hnilicova P, Kantorova E, Sutovsky S, Grofik M, Zelenak K, Kurca E, Zilka N, Parvanovova P, Kolisek M. Imaging Methods Applicable in the Diagnostics of Alzheimer's Disease, Considering the Involvement of Insulin Resistance. Int J Mol Sci 2023; 24:3325. [PMID: 36834741 PMCID: PMC9958721 DOI: 10.3390/ijms24043325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease and the most frequently diagnosed type of dementia, characterized by (1) perturbed cerebral perfusion, vasculature, and cortical metabolism; (2) induced proinflammatory processes; and (3) the aggregation of amyloid beta and hyperphosphorylated Tau proteins. Subclinical AD changes are commonly detectable by using radiological and nuclear neuroimaging methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and single-photon emission computed tomography (SPECT). Furthermore, other valuable modalities exist (in particular, structural volumetric, diffusion, perfusion, functional, and metabolic magnetic resonance methods) that can advance the diagnostic algorithm of AD and our understanding of its pathogenesis. Recently, new insights into AD pathoetiology revealed that deranged insulin homeostasis in the brain may play a role in the onset and progression of the disease. AD-related brain insulin resistance is closely linked to systemic insulin homeostasis disorders caused by pancreas and/or liver dysfunction. Indeed, in recent studies, linkages between the development and onset of AD and the liver and/or pancreas have been established. Aside from standard radiological and nuclear neuroimaging methods and clinically fewer common methods of magnetic resonance, this article also discusses the use of new suggestive non-neuronal imaging modalities to assess AD-associated structural changes in the liver and pancreas. Studying these changes might be of great clinical importance because of their possible involvement in AD pathogenesis during the prodromal phase of the disease.
Collapse
Affiliation(s)
- Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ema Kantorova
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 813 67 Bratislava, Slovakia
| | - Milan Grofik
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Kamil Zelenak
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Egon Kurca
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
7
|
Kara B, Gordon MN, Gifani M, Dorrance AM, Counts SE. Vascular and Nonvascular Mechanisms of Cognitive Impairment and Dementia. Clin Geriatr Med 2023; 39:109-122. [PMID: 36404024 PMCID: PMC10062062 DOI: 10.1016/j.cger.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aging, familial gene mutations, and genetic, environmental, and modifiable lifestyle risk factors predispose individuals to cognitive impairment or dementia by influencing the efficacy of multiple, often interdependent cellular and molecular homeostatic pathways mediating neuronal, glial, and vascular integrity and, ultimately, cognitive status. This review summarizes data from foundational and recent breakthrough studies to highlight common and differential vascular and nonvascular pathogenic mechanisms underlying the progression of Alzheimer disease, vascular dementia, frontotemporal dementia, and dementia with Lewy bodies.
Collapse
Affiliation(s)
- Betul Kara
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Avenue Northwest, Grand Rapids, MI 49503, USA
| | - Marcia N Gordon
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Avenue Northwest, Grand Rapids, MI 49503, USA
| | - Mahsa Gifani
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Avenue Northwest, Grand Rapids, MI 49503, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, USA
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, 400 Monroe Avenue Northwest, Grand Rapids, MI 49503, USA; Department of Family Medicine, Michigan State University, 15 Michigan Street Northeast, Grand Rapids, MI 49503, USA; Hauenstein Neurosciences Center, Mercy Health Saint Mary's Medical Center, 20 Jefferson Avenue Southeast, Grand Rapids, MI 49503, USA.
| |
Collapse
|
8
|
Sanaie S, Nikanfar S, Kalekhane ZY, Azizi-Zeinalhajlou A, Sadigh-Eteghad S, Araj-Khodaei M, Ayati MH, Andalib S. Saffron as a promising therapy for diabetes and Alzheimer's disease: mechanistic insights. Metab Brain Dis 2023; 38:137-162. [PMID: 35986812 DOI: 10.1007/s11011-022-01059-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/29/2022] [Indexed: 02/03/2023]
Abstract
The prevalence of both Alzheimer's disease (AD) and diabetes mellitus is increasing with the societies' aging and has become an essential social concern worldwide. Accumulation of amyloid plaques and neurofibrillary tangles (NFTs) of tau proteins in the brain are hallmarks of AD. Diabetes is an underlying risk factor for AD. Insulin resistance has been proposed to be involved in amyloid-beta (Aβ) aggregation in the brain. It seems that diabetic conditions can result in AD pathology by setting off a cascade of processes, including inflammation, mitochondrial dysfunction, and ROS and advanced glycation end products (AGEs) synthesis. Due to the several side effects of chemical drugs and their high cost, using herbal medicine has recently attracted attention for the treatment of diabetes and AD. Saffron and its active ingredients have been used for its anti-inflammatory, anti-oxidant, anti-diabetic, and anti-AD properties. Therefore, in the present review paper, we take account of the clinical, in vivo and in vitro evidence regarding the anti-diabetic and anti-AD effects of saffron and discuss the preventive or postponing properties of saffron or its components on AD development via its anti-diabetic effects.
Collapse
Affiliation(s)
- Sarvin Sanaie
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Nikanfar
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Yousefi Kalekhane
- Research Center of Psychiatry and Behavioral Sciences, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Psychology, Faculty of Educational Sciences and Psychology, University of Tabriz, Tabriz, Iran
| | - Akbar Azizi-Zeinalhajlou
- Student Research Committee, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Araj-Khodaei
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Persian Medicine, School of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Hossein Ayati
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sasan Andalib
- Research Unit of Clinical Physiology and Nuclear Medicine, Department of Clinical Research, Odense University Hospital, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Rhea EM, Banks WA, Raber J. Insulin Resistance in Peripheral Tissues and the Brain: A Tale of Two Sites. Biomedicines 2022; 10:1582. [PMID: 35884888 PMCID: PMC9312939 DOI: 10.3390/biomedicines10071582] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/12/2022] Open
Abstract
The concept of insulin resistance has been around since a few decades after the discovery of insulin itself. To allude to the classic Charles Dicken's novel published 62 years before the discovery of insulin, in some ways, this is the best of times, as the concept of insulin resistance has expanded to include the brain, with the realization that insulin has a life beyond the regulation of glucose. In other ways, it is the worst of times as insulin resistance is implicated in devastating diseases, including diabetes mellitus, obesity, and Alzheimer's disease (AD) that affect the brain. Peripheral insulin resistance affects nearly a quarter of the United States population in adults over age 20. More recently, it has been implicated in AD, with the degree of brain insulin resistance correlating with cognitive decline. This has led to the investigation of brain or central nervous system (CNS) insulin resistance and the question of the relation between CNS and peripheral insulin resistance. While both may involve dysregulated insulin signaling, the two conditions are not identical and not always interlinked. In this review, we compare and contrast the similarities and differences between peripheral and CNS insulin resistance. We also discuss how an apolipoprotein involved in insulin signaling and related to AD, apolipoprotein E (apoE), has distinct pools in the periphery and CNS and can indirectly affect each system. As these systems are both separated but also linked via the blood-brain barrier (BBB), we discuss the role of the BBB in mediating some of the connections between insulin resistance in the brain and in the peripheral tissues.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - William A. Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA 98195, USA; (E.M.R.); (W.A.B.)
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
10
|
Zhou AL, Sharda N, Sarma VV, Ahlschwede KM, Curran GL, Tang X, Poduslo JF, Kalari KR, Lowe VJ, Kandimalla KK. Age-Dependent Changes in the Plasma and Brain Pharmacokinetics of Amyloid-β Peptides and Insulin. J Alzheimers Dis 2022; 85:1031-1044. [PMID: 34924382 PMCID: PMC10846947 DOI: 10.3233/jad-215128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Age is the most common risk factor for Alzheimer's disease (AD), a neurodegenerative disorder characterized by the hallmarks of toxic amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. Moreover, sub-physiological brain insulin levels have emerged as a pathological manifestation of AD. OBJECTIVE Identify age-related changes in the plasma disposition and blood-brain barrier (BBB) trafficking of Aβ peptides and insulin in mice. METHODS Upon systemic injection of 125I-Aβ40, 125I-Aβ42, or 125I-insulin, the plasma pharmacokinetics and brain influx were assessed in wild-type (WT) or AD transgenic (APP/PS1) mice at various ages. Additionally, publicly available single-cell RNA-Seq data [GSE129788] was employed to investigate pathways regulating BBB transport in WT mice at different ages. RESULTS The brain influx of 125I-Aβ40, estimated as the permeability-surface area product, decreased with age, accompanied by an increase in plasma AUC. In contrast, the brain influx of 125I-Aβ42 increased with age, accompanied by a decrease in plasma AUC. The age-dependent changes observed in WT mice were accelerated in APP/PS1 mice. As seen with 125I-Aβ40, the brain influx of 125I-insulin decreased with age in WT mice, accompanied by an increase in plasma AUC. This finding was further supported by dynamic single-photon emission computed tomography (SPECT/CT) imaging studies. RAGE and PI3K/AKT signaling pathways at the BBB, which are implicated in Aβ and insulin transcytosis, respectively, were upregulated with age in WT mice, indicating BBB insulin resistance. CONCLUSION Aging differentially affects the plasma pharmacokinetics and brain influx of Aβ isoforms and insulin in a manner that could potentially augment AD risk.
Collapse
Affiliation(s)
- Andrew L. Zhou
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Nidhi Sharda
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Vidur V. Sarma
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Kristen M. Ahlschwede
- Department of Pharmaceutical Sciences, Rosalind Franklin University of Medicine and Science, College of Pharmacy, North Chicago, IL, USA
| | - Geoffry L. Curran
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Xiaojia Tang
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Joseph F. Poduslo
- Department of Neurology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Krishna R. Kalari
- Department of Health Sciences, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, College of Medicine, Rochester, MN, USA
| | - Karunya K. Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| |
Collapse
|
11
|
García-Aviles JE, Méndez-Hernández R, Guzmán-Ruiz MA, Cruz M, Guerrero-Vargas NN, Velázquez-Moctezuma J, Hurtado-Alvarado G. Metabolic Disturbances Induced by Sleep Restriction as Potential Triggers for Alzheimer's Disease. Front Integr Neurosci 2021; 15:722523. [PMID: 34539357 PMCID: PMC8447653 DOI: 10.3389/fnint.2021.722523] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sleep has a major role in learning, memory consolidation, and metabolic function. Although it is known that sleep restriction increases the accumulation of amyloid β peptide (Aβ) and the risk to develop Alzheimer's disease (AD), the mechanism behind these effects remains unknown. In this review, we discuss how chronic sleep restriction induces metabolic and cognitive impairments that could result in the development of AD in late life. Here, we integrate evidence regarding mechanisms whereby metabolic signaling becomes disturbed after short or chronic sleep restriction in the context of cognitive impairment, particularly in the accumulation of Aβ in the brain. We also discuss the role of the blood-brain barrier in sleep restriction with an emphasis on the transport of metabolic signals into the brain and Aβ clearance. This review presents the unexplored possibility that the alteration of peripheral metabolic signals induced by sleep restriction, especially insulin resistance, is responsible for cognitive deficit and, subsequently, implicated in AD development.
Collapse
Affiliation(s)
- Jesús Enrique García-Aviles
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Rebeca Méndez-Hernández
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Mara A Guzmán-Ruiz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel Cruz
- Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Hospital de Especialidades, Unidad de Investigación Médica en Bioquímica, Mexico City, Mexico
| | - Natalí N Guerrero-Vargas
- Departamento de Anatomía, Facultad de Medicina, Universidad Nacional Autónoma de México, México City, Mexico
| | - Javier Velázquez-Moctezuma
- Area of Neurosciences, Biology of Reproduction Department, Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico City, Mexico
| | - Gabriela Hurtado-Alvarado
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
12
|
Leclerc M, Dudonné S, Calon F. Can Natural Products Exert Neuroprotection without Crossing the Blood-Brain Barrier? Int J Mol Sci 2021; 22:ijms22073356. [PMID: 33805947 PMCID: PMC8037419 DOI: 10.3390/ijms22073356] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/17/2022] Open
Abstract
The scope of evidence on the neuroprotective impact of natural products has been greatly extended in recent years. However, a key question that remains to be answered is whether natural products act directly on targets located in the central nervous system (CNS), or whether they act indirectly through other mechanisms in the periphery. While molecules utilized for brain diseases are typically bestowed with a capacity to cross the blood–brain barrier, it has been recently uncovered that peripheral metabolism impacts brain functions, including cognition. The gut–microbiota–brain axis is receiving increasing attention as another indirect pathway for orally administered compounds to act on the CNS. In this review, we will briefly explore these possibilities focusing on two classes of natural products: omega-3 polyunsaturated fatty acids (n-3 PUFAs) from marine sources and polyphenols from plants. The former will be used as an example of a natural product with relatively high brain bioavailability but with tightly regulated transport and metabolism, and the latter as an example of natural compounds with low brain bioavailability, yet with a growing amount of preclinical and clinical evidence of efficacy. In conclusion, it is proposed that bioavailability data should be sought early in the development of natural products to help identifying relevant mechanisms and potential impact on prevalent CNS disorders, such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Axe Neurosciences, Centre de Recherche du CHU de Québec–Université Laval, Québec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
| | - Stéphanie Dudonné
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Axe Neurosciences, Centre de Recherche du CHU de Québec–Université Laval, Québec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-(418)-525-4444 (ext. 48697); Fax: +1-(418)-654-2761
| |
Collapse
|
13
|
Nash Y, Frenkel D. Inflammation and insulin resistance in Alzheimer’s disease. GENETICS, NEUROLOGY, BEHAVIOR, AND DIET IN DEMENTIA 2020:389-405. [DOI: 10.1016/b978-0-12-815868-5.00025-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Rhea EM, Banks WA. Role of the Blood-Brain Barrier in Central Nervous System Insulin Resistance. Front Neurosci 2019; 13:521. [PMID: 31213970 PMCID: PMC6558081 DOI: 10.3389/fnins.2019.00521] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/06/2019] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) mediates the communication between the periphery and the central nervous system (CNS). Recently, CNS insulin resistance has been elucidated to play a role in neurodegenerative disease. This has stimulated a wealth of information on the molecular impact of insulin in the brain, particularly in the improvement of cognition. Since the BBB regulates the transport of insulin into the brain and thus, helps to regulate CNS levels, alterations in the BBB response to insulin could impact CNS insulin resistance. In this review, we summarize the effect of insulin on some of the cell types that make up the BBB, including endothelial cells, neurons, astrocytes, and pericytes. We broadly discuss how these changes in specific cell types could ultimately impact the BBB. We also summarize how insulin can regulate levels of the pathological hallmarks of Alzheimer's disease, including amyloid beta (Aβ) and tau within each cell type. Finally, we suggest interventional approaches to overcome detrimental effects on the BBB in regards to changes in insulin transport.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
Wei W, Chen M, Li G, Sang N. Atmospheric PM 2.5 aspiration causes tauopathy by disturbing the insulin signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:301-305. [PMID: 30458396 DOI: 10.1016/j.ecoenv.2018.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/29/2018] [Accepted: 11/01/2018] [Indexed: 06/09/2023]
Abstract
Epidemiological and toxicological studies have shown that ambient fine particulate matter (PM2.5) is a healthy risk factor for neurodegenerative diseases. Hyperphosphorylated tau is the common feature of numerous neurodegenerative diseases known as tauopathy, which could be inhibited by insulin stimulation. However, the effects of PM2.5 on tau protein injury by disturbing the insulin signaling pathway still need to be illuminated. In present study, male C57BL/6 J mice were administered with PM2.5 to determine whether PM2.5 inhalation can induce tauopathy via the insulin resistance (IR) related pathway (IRS-1/AKT/GSK-3β signaling pathway). The results showed that PM2.5 treatment induced the generation of phosphorylated tau (P-tau) and contributed to the development of tauopathy because of the insulin signaling disorders in insulin targeting organs. As expected, the occurrence of central and peripheral IR and accompanying hyperinsulinemia aggravated the disturbance of the IRS-1/AKT/GSK-3β signaling pathway. These observations indicated that PM2.5 exposure led to neurodegenerative tau lesion, and insulin signaling pathway might be a potential therapeutic target for tauopathy.
Collapse
Affiliation(s)
- Wei Wei
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Minjun Chen
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
16
|
Bourassa P, Alata W, Tremblay C, Paris-Robidas S, Calon F. Transferrin Receptor-Mediated Uptake at the Blood-Brain Barrier Is Not Impaired by Alzheimer's Disease Neuropathology. Mol Pharm 2019; 16:583-594. [PMID: 30609376 DOI: 10.1021/acs.molpharmaceut.8b00870] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transferrin receptor (TfR) is highly expressed by brain capillary endothelial cells (BCECs) forming the blood-brain barrier (BBB) and is therefore considered as a potential target for brain drug delivery. Monoclonal antibodies binding to the TfR, such as clone Ri7, have been shown to internalize into BCECs in vivo. However, since Alzheimer's disease (AD) is accompanied by a BBB dysfunction, it raises concerns about whether TfR-mediated transport becomes inefficient during the progression of the disease. Measurements of TfR levels using Western blot analysis in whole homogenates from human post-mortem parietal cortex and hippocampus did not reveal any significant difference between individuals with or without a neuropathological diagnosis of AD (respectively, n = 19 and 22 for the parietal cortex and n = 12 and 14 for hippocampus). Similarly, TfR concentrations in isolated human brain microvessels from parietal cortex were similar between controls and AD cases. TfR levels in isolated murine brain microvessels were not significantly different between groups of 12- and 18-month-old NonTg and 3xTg-AD mice, the latter modeling Aβ and τ neuropathologies. In situ brain perfusion assays were then conducted to measure the brain uptake and internalization of fluorolabeled Ri7 in BCECs upon binding. Consistently, TfR-mediated uptake in BCECs was similar between 3xTg-AD mice and nontransgenic controls (∼0.3 μL·g-1·s-1) at 12, 18, and 22 months of age. Fluorescence microscopy analysis following intravenous administration of fluorolabeled Ri7 highlighted that the signal from the antibody was widely distributed throughout the cerebral vasculature but not in neurons or astrocytes. Overall, our data suggest that both TfR protein levels and TfR-dependent internalization mechanisms are preserved in the presence of Aβ and τ neuropathologies, supporting the potential of TfR as a vector target for drug delivery into BCECs in AD.
Collapse
Affiliation(s)
- Philippe Bourassa
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Wael Alata
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Cyntia Tremblay
- Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Sarah Paris-Robidas
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Frédéric Calon
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| |
Collapse
|
17
|
Duarte A, Santos M, Oliveira C, Moreira P. Brain insulin signalling, glucose metabolism and females' reproductive aging: A dangerous triad in Alzheimer's disease. Neuropharmacology 2018; 136:223-242. [PMID: 29471055 DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
18
|
Kuai Z, Xu Y, Zhao Q, Liu J, Guan S, Qiao Y, Gong X, Nie J, Li P, Liu D, Xing Y, Li H, Sun Z, Wang W, Ning C, Shi Y, Kong W, Shan Y. Effects of insulin on transcriptional response and permeability in an in vitro model of human blood‐brain barrier. J Cell Biochem 2018; 119:5657-5664. [DOI: 10.1002/jcb.26744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ziyu Kuai
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Yan Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Qi Zhao
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Jie Liu
- Faculty of Health SciencesUniversity of MacauMacauChina
| | - Shanshan Guan
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Xin Gong
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Pengju Li
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Dongni Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Yifan Xing
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Huiwen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Zixiao Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Wenqi Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Chunan Ning
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
- Key Laboratory for Molecular Enzymology and EngineeringThe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunJilinChina
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life SciencesJilin UniversityChangchunJilinChina
- Key Laboratory for Molecular Enzymology and EngineeringThe Ministry of EducationSchool of Life SciencesJilin UniversityChangchunJilinChina
| |
Collapse
|
19
|
Ribarič S. Peptides as Potential Therapeutics for Alzheimer's Disease. Molecules 2018; 23:E283. [PMID: 29385735 PMCID: PMC6017258 DOI: 10.3390/molecules23020283] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/22/2022] Open
Abstract
Intracellular synthesis, folding, trafficking and degradation of proteins are controlled and integrated by proteostasis. The frequency of protein misfolding disorders in the human population, e.g., in Alzheimer's disease (AD), is increasing due to the aging population. AD treatment options are limited to symptomatic interventions that at best slow-down disease progression. The key biochemical change in AD is the excessive accumulation of per-se non-toxic and soluble amyloid peptides (Aβ(1-37/44), in the intracellular and extracellular space, that alters proteostasis and triggers Aβ modification (e.g., by reactive oxygen species (ROS)) into toxic intermediate, misfolded soluble Aβ peptides, Aβ dimers and Aβ oligomers. The toxic intermediate Aβ products aggregate into progressively less toxic and less soluble protofibrils, fibrils and senile plaques. This review focuses on peptides that inhibit toxic Aβ oligomerization, Aβ aggregation into fibrils, or stabilize Aβ peptides in non-toxic oligomers, and discusses their potential for AD treatment.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
20
|
Byun MS, Kim HJ, Yi D, Choi HJ, Baek H, Lee JH, Choe YM, Sohn BK, Lee JY, Lee Y, Ko H, Kim YK, Lee YS, Sohn CH, Woo JI, Lee DY. Differential effects of blood insulin and HbA1c on cerebral amyloid burden and neurodegeneration in nondiabetic cognitively normal older adults. Neurobiol Aging 2017; 59:15-21. [DOI: 10.1016/j.neurobiolaging.2017.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023]
|
21
|
Smith DK, He M, Zhang CL, Zheng JC. The therapeutic potential of cell identity reprogramming for the treatment of aging-related neurodegenerative disorders. Prog Neurobiol 2017; 157:212-229. [PMID: 26844759 PMCID: PMC5848468 DOI: 10.1016/j.pneurobio.2016.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/25/2015] [Accepted: 01/04/2016] [Indexed: 12/12/2022]
Abstract
Neural cell identity reprogramming strategies aim to treat age-related neurodegenerative disorders with newly induced neurons that regenerate neural architecture and functional circuits in vivo. The isolation and neural differentiation of pluripotent embryonic stem cells provided the first in vitro models of human neurodegenerative disease. Investigation into the molecular mechanisms underlying stem cell pluripotency revealed that somatic cells could be reprogrammed to induced pluripotent stem cells (iPSCs) and these cells could be used to model Alzheimer disease, amyotrophic lateral sclerosis, Huntington disease, and Parkinson disease. Additional neural precursor and direct transdifferentiation strategies further enabled the induction of diverse neural linages and neuron subtypes both in vitro and in vivo. In this review, we highlight neural induction strategies that utilize stem cells, iPSCs, and lineage reprogramming to model or treat age-related neurodegenerative diseases, as well as, the clinical challenges related to neural transplantation and in vivo reprogramming strategies.
Collapse
Affiliation(s)
- Derek K Smith
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Miao He
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Physical Therapy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, The University of Texas Southwestern Medical Center, 6000 Harry Hines Boulevard, Dallas, TX 75390, USA.
| | - Jialin C Zheng
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Family Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Center for Translational Neurodegeneration and Regenerative Therapy, the Collaborative Innovation Center for Brain Science, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
22
|
Tsuneki H, Yoshida H, Endo K, Mori N, Hosoh S, Tsuda M, Wada T, Sasaoka T. Different impacts of acylated and non-acylated long-acting insulin analogs on neural functions in vitro and in vivo. Diabetes Res Clin Pract 2017; 129:62-72. [PMID: 28511140 DOI: 10.1016/j.diabres.2017.03.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/13/2017] [Accepted: 03/28/2017] [Indexed: 01/06/2023]
Abstract
AIMS Centrally administered insulin improves cognitive functions in patients with Alzheimer's disease; however, it remains unknown whether long-acting insulin analogs exert more pronounced effects than insulin. In the present study, we directly compared the effects of insulin and its analogs on neural functions in vitro and in vivo. METHODS Cultured rat cerebral cortical neurons were treated with insulin, insulin glargine U100 (Gla), insulin detemir (Det), or insulin degludec (Deg). Moreover, these drugs were intracerebroventricularly administered to mice. Their efficacies were evaluated by biochemical and behavioral analyses. RESULTS In cultured neurons, insulin, Gla, and Det increased phosphorylation of Akt and enhanced gene expression of brain-derived neurotrophic factor to a similar extent, although Deg was less effective. The effects of Det and Deg, but not insulin and Gla were suppressed by addition of albumin. When the drug was centrally administered, the increasing effects of insulin on the Akt phosphorylation were comparable to those of Gla but greater than those of Det in hippocampus and cerebral cortex of diabetic db/db and non-diabetic db/m+ mice. Moreover, insulin and Gla enhanced memory functions in Y-maze test and suppressed depression-like behavior in forced swim test in normal mice to a similar extent, and these effects were more potent than those of Det. CONCLUSIONS Insulin and Gla have greater impacts on central nervous system than insulin analogs with high albumin sensitivity, such as Det and Deg. These pharmacological profiles should be taken into account for developing an insulin-based therapy to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Hitomi Yoshida
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kosuke Endo
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Norihiko Mori
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Shuji Hosoh
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masaaki Tsuda
- Department of Biological Chemistry, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
23
|
Marie-Thérèse T, Milène V, Cyntia T, Marine T, Ariane GR, David BA, Frédéric C. Altered cerebral insulin response in transgenic mice expressing the epsilon-4 allele of the human apolipoprotein E gene. Psychoneuroendocrinology 2017; 77:203-210. [PMID: 28088659 PMCID: PMC9162491 DOI: 10.1016/j.psyneuen.2016.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/11/2016] [Accepted: 11/21/2016] [Indexed: 10/20/2022]
Abstract
Apolipoprotein E epsilon-4 (APOEε4 or APOE4), an allelic variation of the APOE gene, not only increases the risk of developing the late-onset form of Alzheimer's disease (AD), but also influences the outcome of treatment. Indeed, data from clinical studies show that the beneficial effect of insulin on cognition is blunted in APOE4 carriers. To investigate how APOE impacts insulin response, we assessed the effects of an acute insulin injection in APOE3- and APOE4-targeted replacement mice that respectively express the human APOE3 or APOE4 isoform instead of the endogenous murine ApoE protein. We evaluated cognition, insulin signaling and proteins implicated in Aβ transport and tau phosphorylation in the cortex and brain capillaries. We found that a single acute insulin injection increased Akt pSer473 in APOE4 compared to APOE3 mice (+113% versus +78.5%), indicating that APOE4 carriage potentiates activation of insulin upstream signaling pathway in the brain. Insulin also led to decreased concentrations of the receptor for advanced glycation endproducts (RAGE) in brain capillaries in both groups of mice. Moreover, higher phosphorylation of tau at Ser202, one of the key markers of AD neuropathology, was observed in insulin-injected APOE4 mice (+44%), consistent with findings in human APOE4 carriers (+400% compared to non-carriers). Therefore, our data suggest that APOE4 carriage leads to an increased insulin-induced activation of cerebral Akt pathway, associated with higher AD-like tau neuropathology. Our results provide evidence of altered insulin signaling in APOE4 carriers as well as a possible mechanism to explain the absence of cognitive benefit from insulin therapy in these individuals.
Collapse
Affiliation(s)
- Traversy Marie-Thérèse
- Faculté de pharmacie, Université Laval, Québec, Qc, Canada.,Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada
| | - Vandal Milène
- Faculté de pharmacie, Université Laval, Québec, Qc, Canada.,Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada.,Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Qc, Canada
| | - Tremblay Cyntia
- Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada
| | - Tournissac Marine
- Faculté de pharmacie, Université Laval, Québec, Qc, Canada.,Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada.,Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Qc, Canada
| | - Giguère-Rancourt Ariane
- Faculté de pharmacie, Université Laval, Québec, Qc, Canada.,Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada
| | - Bennett A. David
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Calon Frédéric
- Faculté de pharmacie, Université Laval, Québec, Qc, Canada.,Axe Neurosciences, Centre de recherche du CHU-Q (Pavillon CHUL), Québec, Qc, Canada.,Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, Qc, Canada.,Corresponding Author : Centre de recherche du CHU-Q (Pavillon CHUL), Room T-2-67, 2705 boul. Laurier, Québec (Québec) G1V 4G2, (418) 525-4444 x48697
| |
Collapse
|
24
|
Bloch K, Vanichkin A, Gil-Ad I, Vardi P, Weizman A. Insulin delivery to the brain using intracranial implantation of alginate-encapsulated pancreatic islets. J Tissue Eng Regen Med 2017; 11:3263-3272. [DOI: 10.1002/term.2235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 04/13/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler Faculty of Medicine; Tel Aviv University; Beilinson Campus Petah Tikva Israel
| | - Alexey Vanichkin
- Laboratory of Transplantation, Felsenstein Medical Research Center, Sackler Faculty of Medicine; Tel Aviv University; Beilinson Campus Petah Tikva Israel
| | - Irit Gil-Ad
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine; Tel Aviv University; Beilinson Campus Petah Tikva Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler Faculty of Medicine; Tel Aviv University; Beilinson Campus Petah Tikva Israel
| | - Abraham Weizman
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine; Tel Aviv University; Beilinson Campus Petah Tikva Israel
- Research Unit; Geha Mental Health Center; Petah Tikva Israel
| |
Collapse
|
25
|
Yan W, Ku T, Yue H, Li G, Sang N. NO 2 inhalation causes tauopathy by disturbing the insulin signaling pathway. CHEMOSPHERE 2016; 165:248-256. [PMID: 27657817 DOI: 10.1016/j.chemosphere.2016.09.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 06/06/2023]
Abstract
Air pollution has been evidenced as a risk factor for neurodegenerative tauopathies. NO2, a primary component of air pollution, is negatively linked to neurodegenerative disorders, but its independent and direct association with tau lesion remains to be elucidated. Considering the fact that the insulin signaling pathway can be targeted by air pollutants and regulate tau function, this study focused on the role of insulin signaling in this NO2-induced tauopathy. Using a dynamic inhalation treatment, we demonstrated that exposure to NO2 induced a disruption of insulin signaling in skeletal muscle, liver, and brain, with associated p38 MAPK and/or JNK activation. We also found that in parallel with these kinase signaling cascades, the compensatory hyperinsulinemia triggered by whole-body insulin resistance (IR) further attenuated the IRS-1/AKT/GSK-3β signaling pathway in the central nervous system, which consequently increased the phosphorylation of tau and reduced the expression of synaptic proteins that contributed to the development of the tau pathology. These findings provide new insight into the possible mechanisms involved in the etiopathogenesis of NO2-induced tauopathy, suggesting that the targeting of insulin signaling may be a promising therapeutic strategy to prevent this disease.
Collapse
Affiliation(s)
- Wei Yan
- College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Tingting Ku
- College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huifeng Yue
- College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guangke Li
- College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Nan Sang
- College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
26
|
Ribarič S. The Rationale for Insulin Therapy in Alzheimer's Disease. Molecules 2016; 21:molecules21060689. [PMID: 27240327 PMCID: PMC6273626 DOI: 10.3390/molecules21060689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/14/2016] [Accepted: 05/19/2016] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with a prevalence that increases with age. By 2050, the worldwide number of patients with AD is projected to reach more than 140 million. The prominent signs of AD are progressive memory loss, accompanied by a gradual decline in cognitive function and premature death. AD is the clinical manifestation of altered proteostasis. The initiating step of altered proteostasis in most AD patients is not known. The progression of AD is accelerated by several chronic disorders, among which the contribution of diabetes to AD is well understood at the cell biology level. The pathological mechanisms of AD and diabetes interact and tend to reinforce each other, thus accelerating cognitive impairment. At present, only symptomatic interventions are available for treating AD. To optimise symptomatic treatment, a personalised therapy approach has been suggested. Intranasal insulin administration seems to open the possibility for a safe, and at least in the short term, effective symptomatic intervention that delays loss of cognition in AD patients. This review summarizes the interactions of AD and diabetes from the cell biology to the patient level and the clinical results of intranasal insulin treatment of cognitive decline in AD.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
27
|
Yuan X, Mei B, Zhang L, Zhang C, Zheng M, Liang H, Wang W, Zheng J, Ding L, Zheng K. Enhanced penetration of exogenous EPCs into brains of APP/PS1 transgenic mice. Am J Transl Res 2016; 8:1460-1470. [PMID: 27186272 PMCID: PMC4859631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/19/2016] [Indexed: 06/05/2023]
Abstract
The aim of this study was to investigate the repair function of exogenous Endothelial progenitor cells (EPCs) for brain microvascular damage of the APP/PS1 transgenic mouse model of Alzheimer's disease (AD). This study used a density-gradient centrifugation method to isolate mononuclear cells (MNCs) from mouse bone marrow, which were subsequently seeded and cultured. Cells were characterized by morphology and detection of the surface markers CD34 and CD133 at different time points by immunofluorescence (IF) and flow cytometry (FCM). Then, EPCs were transfected with GFP adenoviral vectors (GFP-EPCs). Wild-type (WT) and APP/PS1 transgenic mice both received GFP-EPCs injection through the tail vein, and using a PBS buffer injection as the control. Seven days later, the animals' brain tissue was isolated. Expression of GFP was detected by quantitative polymerase chain reaction (qPCR) and western-blot (WB), while the fluorescence of GFP within the brains of mice was observed under a fluorescence microscope. Higher mRNA and protein expression of GFP, accompanied with increased green fluorescence, were detected in the brain of GFP-EPCs-injected APP/PS1 mice, as compared with GFP-EPCs-injected WT mice. The results show that the APP/PS1 transgenic mouse model of AD exhibited enhanced penetration of exogenous EPCs into brains than the WT mice.
Collapse
Affiliation(s)
- Xiaoyang Yuan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Bin Mei
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Miao Zheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Huifang Liang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Wei Wang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Jie Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Ling Ding
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| |
Collapse
|