201
|
Avgerinos KI, Kalaitzidis G, Malli A, Kalaitzoglou D, Myserlis PG, Lioutas VA. Intranasal insulin in Alzheimer's dementia or mild cognitive impairment: a systematic review. J Neurol 2018; 265:1497-1510. [PMID: 29392460 PMCID: PMC6465964 DOI: 10.1007/s00415-018-8768-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS Due to common pathophysiological findings of Alzheimer's disease (AD) with diabetes mellitus (DM), insulin has been suggested as a possible treatment of AD or mild cognitive impairment (MCI). A safe alternative of IV insulin is intranasal (IN) insulin. The aim of this systematic review is to investigate the effects of IN insulin on cognitive function of patients with either AD or MCI. METHODS A literature search of the electronic databases Medline, Scopus and CENTRAL was performed to identify RCTs investigating the effect of IN insulin administration on cognitive tasks, in patients with AD or MCI. RESULTS Seven studies (293 patients) met our inclusion criteria. Most studies showed that verbal memory and especially story recall was improved after IN insulin administration. Sometimes the effect was restricted for apoe4 (-) patients. Intranasal insulin did not affect other cognitive functions. However, there were some positive results in functional status and daily activity. Data suggested that different insulin types and doses may have different effects on different apoe4 groups. In addition, the effects of treatment on Αβ levels differed from study to study. Finally, IN insulin resulted in minor adverse effects. CONCLUSIONS Intranasal insulin improved story recall performance of apoe4 (-) patients with AD or MCI. Other cognitive functions were not affected, but there were some positive results in functional status and daily activity. Since IN insulin is a safe intervention, future studies should be conducted with larger doses and after proper selection of patients and insulin types.
Collapse
Affiliation(s)
- Konstantinos Ioannis Avgerinos
- 251 Hellenic Airforce General Hospital, Kanellopoulou 3, 11525, Athens, Greece.
- Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
- Society of Junior Doctors, Athens, Greece.
| | - Grigorios Kalaitzidis
- Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Society of Junior Doctors, Athens, Greece
| | - Antonia Malli
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Society of Junior Doctors, Athens, Greece
| | - Dimitrios Kalaitzoglou
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Society of Junior Doctors, Athens, Greece
| | - Pavlos Gr Myserlis
- 401 General Army Hospital, Athens, Greece
- Society of Junior Doctors, Athens, Greece
| | - Vasileios-Arsenios Lioutas
- Department of Neurology, Division of Cerebrovascular Diseases, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| |
Collapse
|
202
|
Cao L, Wang Z, Wan W. Suppressor of Cytokine Signaling 3: Emerging Role Linking Central Insulin Resistance and Alzheimer's Disease. Front Neurosci 2018; 12:417. [PMID: 29973864 PMCID: PMC6020761 DOI: 10.3389/fnins.2018.00417] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 06/01/2018] [Indexed: 01/09/2023] Open
Abstract
Currently, the etiology of Alzheimer’s disease (AD) is still elusive. Central insulin resistance has been determined to play an important role in the progress of AD. However, the mechanism underlying the development of disrupted insulin signaling pathways in AD is unclear. Suppressor of cytokine signaling 3 (SOCS3) is a member of the SOCS protein family that acts as a negative modulator of insulin signaling in sensitive tissues, such as hepatocytes and adipocytes. However, little is known about its role in neurological diseases. Recent evidence indicates that the level of SOCS3 is increased in the brains of individuals with AD, especially in areas with amyloid beta deposition, suggesting that SOCS3 may regulate the central insulin signaling pathways in AD. Here, we discuss the potential role of SOCS3 in AD and speculate that SOCS3 may be a promising therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Lan Cao
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zigao Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenbin Wan
- Department of Neurology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
203
|
Lee JH, Jahrling JB, Denner L, Dineley KT. Targeting Insulin for Alzheimer’s Disease: Mechanisms, Status and Potential Directions. J Alzheimers Dis 2018; 64:S427-S453. [DOI: 10.3233/jad-179923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jung Hyun Lee
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jordan B. Jahrling
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Larry Denner
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Kelly T. Dineley
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
204
|
Bayunova LV, Zorina II, Zakharova IO, Avrova NF. Insulin Increases Viability of Neurons in Rat Cerebral Cortex and Normalizes Bax/Bcl-2 Ratio under Conditions of Oxidative Stress. Bull Exp Biol Med 2018; 165:14-17. [DOI: 10.1007/s10517-018-4088-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Indexed: 10/16/2022]
|
205
|
Aderibigbe BA, Naki T. Design and Efficacy of Nanogels Formulations for Intranasal Administration. Molecules 2018; 23:E1241. [PMID: 29789506 PMCID: PMC6100477 DOI: 10.3390/molecules23061241] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Nanogels are drug delivery systems that can bypass the blood-brain barrier and deliver drugs to the desired site when administered intranasally. They have been used as a drug delivery platform for the management of brain diseases such as Alzheimer disease, migraine, schizophrenia and depression. nanogels have also been developed as vaccine carriers for the protection of bacterial infections such as influenza, meningitis, pneumonia and as veterinary vaccine carriers for the protection of animals from encephalomyelitis and mouth to foot disease. It has been developed as vaccine carriers for the prevention of lifestyle disease such as obesity. Intranasal administration of therapeutics using nanogels for the management of brain diseases revealed that the drug transportation was via the olfactory nerve pathway resulting in rapid drug delivery to the brain with excellent neuroprotective effect. The application of nanogels as vaccine carriers also induced significant responses associated with protective immunity against selected bacterial and viral infections. This review provides a detailed information on the enhanced therapeutic effects, mechanisms and biological efficacy of nanogels for intranasal administration.
Collapse
Affiliation(s)
- Blessing A Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| | - Tobeka Naki
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa.
| |
Collapse
|
206
|
Nakamura N, Ohyagi Y, Imamura T, Yanagihara YT, Iinuma KM, Soejima N, Murai H, Yamasaki R, Kira JI. Apomorphine Therapy for Neuronal Insulin Resistance in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 58:1151-1161. [PMID: 28550243 DOI: 10.3233/jad-160344] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Apomorphine (APO) promotes intraneuronal amyloid-β (Aβ) degradation and improves memory function in an Alzheimer's disease (AD) model, 3xTg-AD mice. Since insulin resistance is increased in AD neurons, we investigated the effects of APO on brain insulin resistance in 3xTg-AD mice at early and late stages. After 1-month subcutaneous injection of Apokyn® to 3xTg-AD mice at 6 or 12 months of age, memory function was significantly improved in both age groups. Protein levels of insulin-degrading enzyme (IDE), which is linked to insulin signaling and degrades Aβ, significantly increased in the 3xTg-AD mice brain compared with non-transgenic mice, and were further increased by APO. Protein levels of two types of serine-phosphorylated insulin receptor substrate-1 (IRS-1), pS616 and pS636/639, significantly decreased following APO treatment in the 13-month-old 3xTg-AD mice brain, suggesting improved brain insulin resistance. Immunostaining of the IDE, pS616 and pS636/639 IRS-1 demonstrated similar changes due to APO treatment. Thus, brain insulin resistance is considered an important therapeutic target in AD, and APO may provide improved neuronal insulin resistance.
Collapse
Affiliation(s)
- Norimichi Nakamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasumasa Ohyagi
- Department of Geriatric Medicine and Neurology, Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Tomohiro Imamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki T Yanagihara
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoko Soejima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Murai
- Department of Neurological Therapeutics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
207
|
Benedict C, Grillo CA. Insulin Resistance as a Therapeutic Target in the Treatment of Alzheimer's Disease: A State-of-the-Art Review. Front Neurosci 2018; 12:215. [PMID: 29743868 PMCID: PMC5932355 DOI: 10.3389/fnins.2018.00215] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/19/2018] [Indexed: 01/10/2023] Open
Abstract
Research in animals and humans has shown that type 2 diabetes and its prodromal state, insulin resistance, promote major pathological hallmarks of Alzheimer's disease (AD), such as the formation of amyloid plaques and neurofibrillary tangles (NFT). Worrisomely, dysregulated amyloid beta (Aβ) metabolism has also been shown to promote central nervous system insulin resistance; although the role of tau metabolism remains controversial. Collectively, as proposed in this review, these findings suggest the existence of a mechanistic interplay between AD pathogenesis and disrupted insulin signaling. They also provide strong support for the hypothesis that pharmacologically restoring brain insulin signaling could represent a promising strategy to curb the development and progression of AD. In this context, great hopes have been attached to the use of intranasal insulin. This drug delivery method increases cerebrospinal fluid concentrations of insulin in the absence of peripheral side effects, such as hypoglycemia. With this in mind, the present review will also summarize current knowledge on the efficacy of intranasal insulin to mitigate major pathological symptoms of AD, i.e., cognitive impairment and deregulation of Aβ and tau metabolism.
Collapse
Affiliation(s)
| | - Claudia A Grillo
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina-School of Medicine, Columbia, SC, United States
| |
Collapse
|
208
|
Lehrer S, Rheinstein PH, Rosenzweig KE. Association of Radon Background and Total Background Ionizing Radiation with Alzheimer's Disease Deaths in U.S. States. J Alzheimers Dis 2018; 59:737-741. [PMID: 28671130 DOI: 10.3233/jad-170308] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Exposure of the brain to ionizing radiation might promote the development of Alzheimer's disease (AD). OBJECTIVE Analysis of AD death rates versus radon background radiation and total background radiation in U.S. states. METHODS Total background, radon background, cosmic and terrestrial background radiation measurements are from Assessment of Variations in Radiation Exposure in the United States and Report No. 160 - Ionizing Radiation Exposure of the Population of the United States. 2013 AD death rates by U.S. state are from the Alzheimer's Association. RESULTS Radon background ionizing radiation was significantly correlated with AD death rate in 50 states and the District of Columbia (r = 0.467, p = 0.001). Total background ionizing radiation was also significantly correlated with AD death rate in 50 states and the District of Columbia (r = 0.452, p = 0.001). Multivariate linear regression weighted by state population demonstrated that AD death rate was significantly correlated with radon background (β= 0.169, p < 0.001), age (β= 0.231, p < 0.001), hypertension (β= 0.155, p < 0.001), and diabetes (β= 0.353, p < 0.001). CONCLUSION Our findings, like other studies, suggest that ionizing radiation is a risk factor for AD. Intranasal inhalation of radon gas could subject the rhinencephalon and hippocampus to damaging radiation that initiates AD. The damage would accumulate over time, causing age to be a powerful risk factor.
Collapse
Affiliation(s)
- Steven Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Kenneth E Rosenzweig
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
209
|
Batista AF, Forny-Germano L, Clarke JR, Lyra E Silva NM, Brito-Moreira J, Boehnke SE, Winterborn A, Coe BC, Lablans A, Vital JF, Marques SA, Martinez AM, Gralle M, Holscher C, Klein WL, Houzel JC, Ferreira ST, Munoz DP, De Felice FG. The diabetes drug liraglutide reverses cognitive impairment in mice and attenuates insulin receptor and synaptic pathology in a non-human primate model of Alzheimer's disease. J Pathol 2018; 245:85-100. [PMID: 29435980 PMCID: PMC5947670 DOI: 10.1002/path.5056] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/13/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder that still lacks an effective treatment, and this has stimulated an intense pursuit of disease-modifying therapeutics. Given the increasingly recognized link between AD and defective brain insulin signaling, we investigated the actions of liraglutide, a glucagon-like peptide-1 (GLP-1) analog marketed for treatment of type 2 diabetes, in experimental models of AD. Insulin receptor pathology is an important feature of AD brains that impairs the neuroprotective actions of central insulin signaling. Here, we show that liraglutide prevented the loss of brain insulin receptors and synapses, and reversed memory impairment induced by AD-linked amyloid-β oligomers (AβOs) in mice. Using hippocampal neuronal cultures, we determined that the mechanism of neuroprotection by liraglutide involves activation of the PKA signaling pathway. Infusion of AβOs into the lateral cerebral ventricle of non-human primates (NHPs) led to marked loss of insulin receptors and synapses in brain regions related to memory. Systemic treatment of NHPs with liraglutide provided partial protection, decreasing AD-related insulin receptor, synaptic, and tau pathology in specific brain regions. Synapse damage and elimination are amongst the earliest known pathological changes and the best correlates of memory impairment in AD. The results illuminate mechanisms of neuroprotection by liraglutide, and indicate that GLP-1 receptor activation may be harnessed to protect brain insulin receptors and synapses in AD. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Andre F Batista
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia M Lyra E Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jordano Brito-Moreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susan E Boehnke
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Brian C Coe
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ann Lablans
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Juliana F Vital
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suelen A Marques
- Departament of Neurobiology, Fluminense Federal University, Niteroi, Brazil
| | - Ana Mb Martinez
- Department of Pathology, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, UFRJ, Rio de Janeiro, Brazil
| | - Matthias Gralle
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christian Holscher
- Division of Biomed and Life Sciences, Faculty of Health and Medicine Lancaster University, Lancaster, UK
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Jean-Christophe Houzel
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
210
|
Zorina II, Bayunova LV, Zakharova IO, Avrova NF. The Dependence of the Protective Effect of Insulin on Its Concentration and Modulation of ERK1/2 Activity under the Conditions of Oxidative Stress in Cortical Neurons. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712417040110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
211
|
Mandal A, Prabhavalkar KS, Bhatt LK. Gastrointestinal hormones in regulation of memory. Peptides 2018; 102:16-25. [PMID: 29466709 DOI: 10.1016/j.peptides.2018.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/16/2022]
Abstract
The connection between the gastrointestinal hormones and the brain has been established many years ago. This relation is termed the gut-brain axis (GBA). The GBA is a bidirectional communication which not only regulates gastrointestinal homeostasis but is also linked with higher emotional and cognitive functions. Hypothalamus plays a critical role in the regulation of energy metabolism, nutrient partitioning and control of feeding behaviors. Various gut hormones are released inside the gastrointestinal tract on food intake. These hormones act peripherally and influence the different responses of the tissues to the food intake, but do also have effects on the brain. The hypothalamus, in turn, integrates visceral function with limbic system structures such as hippocampus, amygdala, and cerebral cortex. The hippocampus has been known for its involvement in the cognitive function and the modulation of synaptic plasticity. This review aims to establish the role of various gut hormones in learning and memory, through the interaction of various receptors in the hippocampus. Understanding their role in memory can also aid in finding novel therapeutic strategies for the treatment of the neurological disorders associated with memory dysfunctions.
Collapse
Affiliation(s)
- Anwesha Mandal
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| | - Lokesh K Bhatt
- Department of Pharmacology, SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| |
Collapse
|
212
|
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
|
213
|
Li T, Jiao JJ, Hölscher C, Wu MN, Zhang J, Tong JQ, Dong XF, Qu XS, Cao Y, Cai HY, Su Q, Qi JS. A novel GLP-1/GIP/Gcg triagonist reduces cognitive deficits and pathology in the 3xTg mouse model of Alzheimer's disease. Hippocampus 2018; 28:358-372. [PMID: 29473979 DOI: 10.1002/hipo.22837] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is an important risk factor for Alzheimer's disease (AD). Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) have been identified to be effective in T2DM treatment and neuroprotection. In this study, we further explored the effects of a novel unimolecular GLP-1/GIP/Gcg triagonist on the cognitive behavior and cerebral pathology in the 7-month-old triple transgenic mouse model of AD (3xTg-AD), and investigated its possible electrophysiological and molecular mechanisms. After chronic administration of the GLP-1/GIP/Gcg triagonist (10 nmol/kg bodyweight, once daily, i.p.) for 30 days, open field, Y maze and Morris water maze tests were performed, followed by in vivo electrophysiological recording, immunofluorescence and Western blotting experiments. We found that the chronic treatment with the triagonist could improve long-term spatial memory of 3xTg-AD mice in Morris water maze, as well as the working memory in Y maze task. The triagonist also alleviated the suppression of long-term potentiation (LTP) in the CA1 region of hippocampus. In addition, the triagonist significantly reduced hippocampal pathological damages, including amyloid-β (Aβ) and phosphorylated tau aggregates, and upregulated the expression levels of S133 p-CREB, T286 p-CAMKII and S9 p-GSK3β in the hippocampus of the 3xTg-AD mice. These results demonstrate for the first time that the novel GLP-1/GIP/Gcg triagonist is efficacious in ameliorating cognitive deficits and pathological damages of 3xTg-AD mice, suggesting that the triagonist might be potentially beneficial in the treatment of AD.
Collapse
Affiliation(s)
- Tian Li
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Juan-Juan Jiao
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Christian Hölscher
- Division of Biomed and Life Sciences, Faculty of Health and Medicine Lancaster University, Lancaster, LA1 4YQ, United Kingdom
| | - Mei-Na Wu
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Jun Zhang
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Jia-Qing Tong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Xue-Fan Dong
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Xue-Song Qu
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Yue Cao
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| | - Hong-Yan Cai
- Department of Immunology and Microbiology, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Qiang Su
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Jin-Shun Qi
- Department of Neurobiology and National Key Discipline of Physiology, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China
| |
Collapse
|
214
|
Tateiwa H, Kawano T, Nishigaki A, Yamanaka D, Aoyama B, Shigematsu-Locatelli M, Eguchi S, Locatelli FM, Yokoyama M. The role of hippocampal brain-derived neurotrophic factor in age-related differences in neuropathic pain behavior in rats. Life Sci 2018; 197:56-66. [DOI: 10.1016/j.lfs.2018.01.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/16/2018] [Accepted: 01/30/2018] [Indexed: 01/22/2023]
|
215
|
Fried PJ, Schilberg L, Brem AK, Saxena S, Wong B, Cypess AM, Horton ES, Pascual-Leone A. Humans with Type-2 Diabetes Show Abnormal Long-Term Potentiation-Like Cortical Plasticity Associated with Verbal Learning Deficits. J Alzheimers Dis 2018; 55:89-100. [PMID: 27636847 DOI: 10.3233/jad-160505] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Type-2 diabetes mellitus (T2DM) accelerates cognitive aging and increases risk of Alzheimer's disease. Rodent models of T2DM show altered synaptic plasticity associated with reduced learning and memory. Humans with T2DM also show cognitive deficits, including reduced learning and memory, but the relationship of these impairments to the efficacy of neuroplastic mechanisms has never been assessed. OBJECTIVE Our primary objective was to compare mechanisms of cortical plasticity in humans with and without T2DM. Our secondary objective was to relate plasticity measures to standard measures of cognition. METHODS A prospective cross-sectional cohort study was conducted on 21 adults with T2DM and 15 demographically-similar non-diabetic controls. Long-term potentiation-like plasticity was assessed in primary motor cortex by comparing the amplitude of motor evoked potentials (MEPs) from single-pulse transcranial magnetic stimulation before and after intermittent theta-burst stimulation (iTBS). Plasticity measures were compared between groups and related to neuropsychological scores. RESULTS In T2DM, iTBS-induced modulation of MEPs was significantly less than controls, even after controlling for potential confounds. Furthermore, in T2DM, modulation of MEPs 10-min post-iTBS was significantly correlated with Rey Auditory Verbal Learning Task (RAVLT) performance. CONCLUSION Humans with T2DM show abnormal cortico-motor plasticity that is correlated with reduced verbal learning. Since iTBS after-effects and the RAVLT are both NMDA receptor-dependent measures, their relationship in T2DM may reflect brain-wide alterations in the efficacy of NMDA receptors. These findings offer novel mechanistic insights into the brain consequences of T2DM and provide a reliable means to monitor brain health and evaluate the efficacy of clinical interventions.
Collapse
Affiliation(s)
- Peter J Fried
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lukas Schilberg
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Anna-Katharine Brem
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Sadhvi Saxena
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical School, Baltimore, MD, USA
| | - Bonnie Wong
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Frontotemporal Dementia Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.,Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Edward S Horton
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Interventional Cognitive Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
216
|
Kennedy G, Hardman RJ, Macpherson H, Scholey AB, Pipingas A. How Does Exercise Reduce the Rate of Age-Associated Cognitive Decline? A Review of Potential Mechanisms. J Alzheimers Dis 2018; 55:1-18. [PMID: 27636853 DOI: 10.3233/jad-160665] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The rate of age-associated cognitive decline varies considerably between individuals. It is important, both on a societal and individual level, to investigate factors that underlie these differences in order to identify those which might realistically slow cognitive decline. Physical activity is one such factor with substantial support in the literature. Regular exercise can positively influence cognitive ability, reduce the rate of cognitive aging, and even reduce the risk of Alzheimer's disease (AD) and other dementias. However, while there is substantial evidence in the extant literature for the effect of exercise on cognition, the processes that mediate this relationship are less clear. This review examines cardiovascular health, production of brain derived neurotrophic factor (BDNF), insulin sensitivity, stress, and inflammation as potential pathways, via which exercise may maintain or improve cognitive functioning, and may be particularly pertinent in the context of the aging brain. A greater understanding of these mechanisms and their potential relationships with exercise and cognition will be invaluable in providing biomarkers for investigating the efficacy of differing exercise regimes on cognitive outcomes.
Collapse
Affiliation(s)
- Greg Kennedy
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Roy J Hardman
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Helen Macpherson
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia.,Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, VIC, Australia
| | - Andrew B Scholey
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Andrew Pipingas
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
217
|
Zhao N, Liu CC, Qiao W, Bu G. Apolipoprotein E, Receptors, and Modulation of Alzheimer's Disease. Biol Psychiatry 2018; 83:347-357. [PMID: 28434655 PMCID: PMC5599322 DOI: 10.1016/j.biopsych.2017.03.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 12/31/2022]
Abstract
Apolipoprotein E (apoE) is a lipid carrier in both the peripheral and the central nervous systems. Lipid-loaded apoE lipoprotein particles bind to several cell surface receptors to support membrane homeostasis and injury repair in the brain. Considering prevalence and relative risk magnitude, the ε4 allele of the APOE gene is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). ApoE4 contributes to AD pathogenesis by modulating multiple pathways, including but not limited to the metabolism, aggregation, and toxicity of amyloid-β peptide, tauopathy, synaptic plasticity, lipid transport, glucose metabolism, mitochondrial function, vascular integrity, and neuroinflammation. Emerging knowledge on apoE-related pathways in the pathophysiology of AD presents new opportunities for AD therapy. We describe the biochemical and biological features of apoE and apoE receptors in the central nervous system. We also discuss the evidence and mechanisms addressing differential effects of apoE isoforms and the role of apoE receptors in AD pathogenesis, with a particular emphasis on the clinical and preclinical studies related to amyloid-β pathology. Finally, we summarize the current strategies of AD therapy targeting apoE, and postulate that effective strategies require an apoE isoform-specific approach.
Collapse
Affiliation(s)
- Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
218
|
Mechanism of intranasal drug delivery directly to the brain. Life Sci 2018; 195:44-52. [DOI: 10.1016/j.lfs.2017.12.025] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/08/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023]
|
219
|
Alford S, Patel D, Perakakis N, Mantzoros CS. Obesity as a risk factor for Alzheimer's disease: weighing the evidence. Obes Rev 2018; 19:269-280. [PMID: 29024348 DOI: 10.1111/obr.12629] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/03/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death in the USA today; therefore, it is imperative that public health initiatives and clinical strategies are developed to prevent and effectively treat AD. Despite the enormous impact that AD has on individuals, families, society, and the health care system, there are no biomarkers to clearly identify those at risk for AD, public health prevention strategies in place, or treatments to address the underlying pathology or stop the progression of AD. There is ample scientific as well as empirical evidence that obesity and its metabolic and vascular comorbidities are related to AD and likely in the causative pathway. Obesity prevention and treatment could prove to be an efficacious and safe approach to preventing AD, a serious and daunting epidemic disease. In this review, we present the current pathophysiological and clinical evidence linking obesity and obesity-related comorbidities (eg, insulin resistance, hyperglycaemia, and type 2 diabetes) with AD. Additionally, we discuss which population to target and when to consider treatment for AD. Finally, we summarize the current evidence regarding the efficacy of anti-obesity and anti-diabetic pharmacotherapeutic agents for the treatment of AD.
Collapse
Affiliation(s)
| | - D Patel
- MCPHS University, Boston, MA, USA.,VA Boston Healthcare System, Boston, MA, USA
| | - N Perakakis
- Mantzoros Lab, Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - C S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
220
|
Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer's and Parkinson's disease models. Neuropharmacology 2018; 136:251-259. [PMID: 29402504 DOI: 10.1016/j.neuropharm.2018.01.040] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/10/2018] [Accepted: 01/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes is a risk factor for several chronic neurodegenerative disorders such as Alzheimer's or Parkinson's disease. The link appears to be insulin de-sensitisation in the brain. Insulin is an important neuroprotective growth factor. GLP-1 and GIP are growth factors that re-sensitise insulin and GLP-1 mimetics are used in the clinic to treat diabetes. GLP-1 and GIP mimetics initially designed to treat diabetes show good protective effects in animal models of Alzheimer's and Parkinson's disease. Based on these results, several clinical trials have shown first encouraging effects in patients with Alzheimer's or Parkinson' disease. Novel dual GLP-1/GIP receptor agonists have been developed to treat diabetes, and they also show good neuroprotective effects that are superior to single GLP-1 analogues. Several newer dual analogues have been tested that have been engineered to cross the blood -brain barrier. They show clear neuroprotective effects by reducing inflammation and oxidative stress and apoptotic signalling and protecting memory formation, synaptic numbers and synaptic activity, motor activity, dopaminergic neurons, cortical activity and energy utilisation in the brain. These results demonstrate the potential of developing disease-modifying treatments for Alzheimer's and Parkinson's disease that are superior to current single GLP-1 mimetics. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
|
221
|
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
|
222
|
Nday CM, Eleftheriadou D, Jackson G. Shared pathological pathways of Alzheimer's disease with specific comorbidities: current perspectives and interventions. J Neurochem 2018; 144:360-389. [PMID: 29164610 DOI: 10.1111/jnc.14256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) belongs to one of the most multifactorial, complex and heterogeneous morbidity-leading disorders. Despite the extensive research in the field, AD pathogenesis is still at some extend obscure. Mechanisms linking AD with certain comorbidities, namely diabetes mellitus, obesity and dyslipidemia, are increasingly gaining importance, mainly because of their potential role in promoting AD development and exacerbation. Their exact cognitive impairment trajectories, however, remain to be fully elucidated. The current review aims to offer a clear and comprehensive description of the state-of-the-art approaches focused on generating in-depth knowledge regarding the overlapping pathology of AD and its concomitant ailments. Thorough understanding of associated alterations on a number of molecular, metabolic and hormonal pathways, will contribute to the further development of novel and integrated theranostics, as well as targeted interventions that may be beneficial for individuals with age-related cognitive decline.
Collapse
Affiliation(s)
- Christiane M Nday
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Despoina Eleftheriadou
- Department of Chemical Engineering, Laboratory of Inorganic Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Graham Jackson
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa
| |
Collapse
|
223
|
Takeda M. Current status and promising future of nasal drug delivery. Nihon Yakurigaku Zasshi 2018; 150:148-152. [PMID: 28890477 DOI: 10.1254/fpj.150.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
224
|
Cheng YS, Chen ZT, Liao TY, Lin C, Shen HCH, Wang YH, Chang CW, Liu RS, Chen RPY, Tu PH. An intranasally delivered peptide drug ameliorates cognitive decline in Alzheimer transgenic mice. EMBO Mol Med 2017; 9:703-715. [PMID: 28356312 PMCID: PMC5412883 DOI: 10.15252/emmm.201606666] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Imbalance between the production and clearance of amyloid β (Aβ) peptides is considered to be the primary mechanism of AD pathogenesis. This amyloid hypothesis is supported by the recent success of the human anti‐amyloid antibody aducanumab, in clearing plaque and slowing clinical impairment in prodromal or mild patients in a phase Ib trial. Here, a peptide combining polyarginines (polyR) (for charge repulsion) and a segment derived from the core region of Aβ amyloid (for sequence recognition) was designed. The efficacy of the designed peptide, R8‐Aβ(25–35), on amyloid reduction and the improvement of cognitive functions were evaluated using APP/PS1 double transgenic mice. Daily intranasal administration of PEI‐conjugated R8‐Aβ(25–35) peptide significantly reduced Aβ amyloid accumulation and ameliorated the memory deficits of the transgenic mice. Intranasal administration is a feasible route for peptide delivery. The modular design combining polyR and aggregate‐forming segments produced a desirable therapeutic effect and could be easily adopted to design therapeutic peptides for other proteinaceous aggregate‐associated diseases.
Collapse
Affiliation(s)
- Yu-Sung Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zih-Ten Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Tai-Yan Liao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chen Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Howard C-H Shen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ya-Han Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Chi-Wei Chang
- Biomedical Imaging Research Center, Department of Nuclear Medicine, National Yang-Ming University and Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ren-Shyan Liu
- Biomedical Imaging Research Center, Department of Nuclear Medicine, National Yang-Ming University and Taipei Veterans General Hospital, Taipei, Taiwan.,Molecular and Genetic Imaging Core, Taiwan Mouse Clinic, Academia Sinica, Taipei, Taiwan
| | - Rita P-Y Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan .,Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Pang-Hsien Tu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
225
|
Picone P, Sabatino MA, Ditta LA, Amato A, San Biagio PL, Mulè F, Giacomazza D, Dispenza C, Di Carlo M. Nose-to-brain delivery of insulin enhanced by a nanogel carrier. J Control Release 2017; 270:23-36. [PMID: 29196041 DOI: 10.1016/j.jconrel.2017.11.040] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/21/2017] [Accepted: 11/25/2017] [Indexed: 02/07/2023]
Abstract
Recent evidences suggest that insulin delivery to the brain can be an important pharmacological therapy for some neurodegenerative pathologies, including Alzheimer disease (AD). Due to the presence of the Blood Brain Barrier, a suitable carrier and an appropriate route of administration are required to increase the efficacy and safety of the treatment. Here, poly(N-vinyl pyrrolidone)-based nanogels (NG), synthetized by e-beam irradiation, alone and with covalently attached insulin (NG-In) were characterized for biocompatibility and brain delivery features in a mouse model. Preliminarily, the biodistribution of the "empty" nanocarrier after intraperitoneal (i.p.) injection was investigated by using a fluorescent-labeled NG. By fluorescence spectroscopy, SEM and dynamic light scattering analyses we established that urine clearance occurs in 24h. Histological liver and kidneys inspections indicated that no morphological alterations of tissues occurred and no immunological response was activated after NG injection. Furthermore, after administration of the insulin-conjugated nanogels (NG-In) through the intranasal route (i.n.) no alteration or immunogenic response of the nasal mucosa was observed, suggesting that the formulation is well tolerated in mouse. Moreover, an enhancement of NG-In delivery to the different brain areas and of its biological activity, measured as Akt activation levels, with reference to free insulin administration was demonstrated. Taken together, these results indicate that the synthesized NG-In enhances brain insulin delivery upon i.n. administration and strongly encourage its further evaluation as therapeutic agent against some neurodegenerative diseases.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Maria Antonietta Sabatino
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy
| | - Lorena Anna Ditta
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Edificio 16, 90128 Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Edificio 16, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| | - Clelia Dispenza
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy; Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| | - Marta Di Carlo
- Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| |
Collapse
|
226
|
Freitas HR, Ferreira GDC, Trevenzoli IH, Oliveira KDJ, de Melo Reis RA. Fatty Acids, Antioxidants and Physical Activity in Brain Aging. Nutrients 2017; 9:nu9111263. [PMID: 29156608 PMCID: PMC5707735 DOI: 10.3390/nu9111263] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Polyunsaturated fatty acids and antioxidants are important mediators in the central nervous system. Lipid derivatives may control the production of proinflammatory agents and regulate NF-κB activity, microglial activation, and fatty acid oxidation; on the other hand, antioxidants, such as glutathione and ascorbate, have been shown to signal through transmitter receptors and protect against acute and chronic oxidative stress, modulating the activity of different signaling pathways. Several authors have investigated the role of these nutrients in the brains of the young and the aged in degenerative diseases such as Alzheimer’s and Parkinson’s, and during brain aging due to adiposity- and physical inactivity-mediated metabolic disturbances, chronic inflammation, and oxidative stress. Through a literature review, we aimed to highlight recent data on the role of adiposity, fatty acids, antioxidants, and physical inactivity in the pathophysiology of the brain and in the molecular mechanisms of senescence. Data indicate the complexity and necessity of endogenous/dietary antioxidants for the maintenance of redox status and the control of neuroglial signaling under stress. Recent studies also indicate that omega-3 and -6 fatty acids act in a competitive manner to generate mediators for energy metabolism, influencing feeding behavior, neural plasticity, and memory during aging. Finding pharmacological or dietary resources that mitigate or prevent neurodegenerative affections continues to be a great challenge and requires additional effort from researchers, clinicians, and nutritionists in the field.
Collapse
Affiliation(s)
- Hércules Rezende Freitas
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Gustavo da Costa Ferreira
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| | - Karen de Jesus Oliveira
- Laboratory of Endocrine Physiology and Metabology, Biomedical Institute, Universidade Federal Fluminense, Niterói 24210-130, Brazil.
| | - Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil.
| |
Collapse
|
227
|
Kamei N. Nose-to-Brain Delivery of Peptide Drugs Enhanced by Coadministration of Cell-penetrating Peptides: Therapeutic Potential for Dementia. YAKUGAKU ZASSHI 2017; 137:1247-1253. [PMID: 28966266 DOI: 10.1248/yakushi.17-00138] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent reports suggest that peptide drugs such as insulin have the potential to serve as therapeutics in neurodegenerative diseases such as Alzheimer's disease. However, the transport of these drugs to the therapeutic target, the brain, is significantly hindered by the blood-brain barrier (BBB). Intranasal administration appears to be an ideal solution for drug delivery to the brain, bypassing the BBB, however the entry of peptide drugs into neuronal and epithelial cells in the olfactory mucosa remains low. In this study, we therefore examined whether intranasal coadministration of cell-penetrating peptides (CPPs) could improve nose-to-brain drug transport. In both mice and rats, we found that direct transport of insulin into the brain was significantly facilitated when coadministered with amphipathic CPP penetratin, and eventually insulin reached the deeper regions of the brain such as the hippocampus. In the mouse line senescence-accelerated mouse prone-8 (SAMP8), spatial learning tests demonstrated that long-term intranasal coadministration of insulin with penetratin improved mild memory loss in the early stages of dementia. In contrast, the severe cognitive dysfunction in the aged SAMP8 mice was preserved despite intranasal coadministration of insulin with penetratin. The immunohistological examination of the hippocampus suggested that enhanced nose-to-brain delivery of insulin had a partial neuroprotective effect but unexpectedly increased amyloid β plaque deposition. In conclusion, intranasal coadministration of insulin with CPPs has the potential to serve as a therapeutic for mild cognitive dysfunction. To identify suitable pharmacotherapy for dementia with severe pathology, further studies of nose-to-brain delivery of molecularly appropriate biopharmaceuticals are necessary.
Collapse
Affiliation(s)
- Noriyasu Kamei
- Laboratory of Drug Delivery Systems, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
228
|
Vieira MNN, Lima-Filho RAS, De Felice FG. Connecting Alzheimer's disease to diabetes: Underlying mechanisms and potential therapeutic targets. Neuropharmacology 2017; 136:160-171. [PMID: 29129775 DOI: 10.1016/j.neuropharm.2017.11.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a risk factor for type 2 diabetes and vice versa, and a growing body of evidence indicates that these diseases are connected both at epidemiological, clinical and molecular levels. Recent studies have begun to reveal common pathogenic mechanisms shared by AD and type 2 diabetes. Impaired neuronal insulin signaling and endoplasmic reticulum (ER) stress are present in animal models of AD, similar to observations in peripheral tissue in T2D. These findings shed light into novel diabetes-related mechanisms leading to brain dysfunction in AD. Here, we review the literature on selected mechanisms shared between these diseases and discuss how the identification of such mechanisms may lead to novel therapeutic targets in AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Marcelo N N Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
229
|
Higgs S, Spetter MS, Thomas JM, Rotshtein P, Lee M, Hallschmid M, Dourish CT. Interactions between metabolic, reward and cognitive processes in appetite control: Implications for novel weight management therapies. J Psychopharmacol 2017; 31:1460-1474. [PMID: 29072515 PMCID: PMC5700796 DOI: 10.1177/0269881117736917] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Traditional models of appetite control have emphasised the role of parallel homeostatic and hedonic systems, but more recently the distinction between independent homeostatic and hedonic systems has been abandoned in favour of a framework that emphasises the cross talk between the neurochemical substrates of the two systems. In addition, evidence has emerged more recently, that higher level cognitive functions such as learning, memory and attention play an important role in everyday appetite control and that homeostatic signals also play a role in cognition. Here, we review this evidence and present a comprehensive model of the control of appetite that integrates cognitive, homeostatic and reward mechanisms. We discuss the implications of this model for understanding the factors that may contribute to disordered patterns of eating and suggest opportunities for developing more effective treatment approaches for eating disorders and weight management.
Collapse
Affiliation(s)
- Suzanne Higgs
- 1 School of Psychology, University of Birmingham, Birmingham, UK
| | | | - Jason M Thomas
- 2 Department of Psychology, Aston University, Birmingham, UK
| | - Pia Rotshtein
- 1 School of Psychology, University of Birmingham, Birmingham, UK
| | - Michelle Lee
- 3 Department of Psychology, Swansea University, Swansea, UK
| | - Manfred Hallschmid
- 4 Institute for Medical Psychology and Behavioural Neurobiology, University Tübingen, Tübingen, Germany
- 6 Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
230
|
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]
|
231
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
232
|
Mamik MK, Power C. Inflammasomes in neurological diseases: emerging pathogenic and therapeutic concepts. Brain 2017; 140:2273-2285. [PMID: 29050380 DOI: 10.1093/brain/awx133] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/15/2017] [Indexed: 12/23/2022] Open
Abstract
Inflammasome activation in the central nervous system occurs in both health and disease. Inflammasomes are cytosolic protein complexes that sense specific infectious or host stimuli and initiate inflammatory responses through caspase activation. Assembly of inflammasomes results in caspase-1-mediated proteolytic cleavage and release of the pro-inflammatory cytokines, interleukin-1β and interleukin-18, with initiation of pyroptosis, an inflammatory programmed cell death. Recent developments in the inflammasome field have uncovered novel molecular mechanisms that contribute to a broad range of neurological disorders including those associated with specific mutations in inflammasome genes as well as diseases modulated by inflammasome activation. This update focuses on recent developments in the field of inflammasome biology highlighting different inflammasome activators and pathways discovered in the nervous system. We also discuss targeted therapies that regulate inflammasomes and improve neurological outcomes.
Collapse
Affiliation(s)
- Manmeet K Mamik
- Department of Medicine (Division of Neurology), University of Alberta, Edmonton, AB, Canada
| | - Christopher Power
- Department of Medicine (Division of Neurology), University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
233
|
An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, Chia CW, Egan JM, Ferrucci L, Troncoso J, Levey AI, Lah J, Seyfried NT, Legido-Quigley C, O'Brien R, Thambisetty M. Evidence for brain glucose dysregulation in Alzheimer's disease. Alzheimers Dement 2017; 14:318-329. [PMID: 29055815 DOI: 10.1016/j.jalz.2017.09.011] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 07/05/2017] [Accepted: 09/07/2017] [Indexed: 10/18/2022]
Abstract
INTRODUCTION It is unclear whether abnormalities in brain glucose homeostasis are associated with Alzheimer's disease (AD) pathogenesis. METHODS Within the autopsy cohort of the Baltimore Longitudinal Study of Aging, we measured brain glucose concentration and assessed the ratios of the glycolytic amino acids, serine, glycine, and alanine to glucose. We also quantified protein levels of the neuronal (GLUT3) and astrocytic (GLUT1) glucose transporters. Finally, we assessed the relationships between plasma glucose measured before death and brain tissue glucose. RESULTS Higher brain tissue glucose concentration, reduced glycolytic flux, and lower GLUT3 are related to severity of AD pathology and the expression of AD symptoms. Longitudinal increases in fasting plasma glucose levels are associated with higher brain tissue glucose concentrations. DISCUSSION Impaired glucose metabolism due to reduced glycolytic flux may be intrinsic to AD pathogenesis. Abnormalities in brain glucose homeostasis may begin several years before the onset of clinical symptoms.
Collapse
Affiliation(s)
- Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Vijay R Varma
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | | | - Ramon Casanova
- Department of Biostatistical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Eric Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chee W Chia
- Translational Research and Medical Services Section, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Juan Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - James Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Richard O'Brien
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA.
| |
Collapse
|
234
|
Kulas JA, Puig KL, Combs CK. Amyloid precursor protein in pancreatic islets. J Endocrinol 2017; 235:49-67. [PMID: 28710249 PMCID: PMC6267436 DOI: 10.1530/joe-17-0122] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/13/2017] [Indexed: 01/04/2023]
Abstract
The amyloid precursor protein (APP) has been extensively investigated for its role in the production of amyloid beta (Aβ), a plaque-forming peptide in Alzheimer's disease (AD). Epidemiological evidence suggests type 2 diabetes is a risk factor for AD. The pancreas is an essential regulator of blood glucose levels through the secretion of the hormones insulin and glucagon. Pancreatic dysfunction is a well-characterized consequence of type 1 and type 2 diabetes. In this study, we have examined the expression and processing of pancreatic APP to test the hypothesis that APP may play a role in pancreatic function and the pathophysiology of diabetes. Our data demonstrate the presence of APP within the pancreas, including pancreatic islets in both mouse and human samples. Additionally, we report that the APP/PS1 mouse model of AD overexpresses APP within pancreatic islets, although this did not result in detectable levels of Aβ. We compared whole pancreas and islet culture lysates by Western blot from C57BL/6 (WT), APP-/- and APP/PS1 mice and observed APP-dependent differences in the total protein levels of GLUT4, IDE and BACE2. Immunohistochemistry for BACE2 detected high levels in pancreatic α cells. Additionally, both mouse and human islets processed APP to release sAPP into cell culture media. Moreover, sAPP stimulated insulin but not glucagon secretion from islet cultures. We conclude that APP and its metabolites are capable of influencing the basic physiology of the pancreas, possibly through the release of sAPP acting in an autocrine or paracrine manner.
Collapse
Affiliation(s)
- Joshua A Kulas
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health Sciences, Grand Forks, USA
| | - Kendra L Puig
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health Sciences, Grand Forks, USA
| | - Colin K Combs
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health Sciences, Grand Forks, USA
| |
Collapse
|
235
|
Intracranial IL-17A overexpression decreases cerebral amyloid angiopathy by upregulation of ABCA1 in an animal model of Alzheimer's disease. Brain Behav Immun 2017; 65:262-273. [PMID: 28526436 PMCID: PMC5537015 DOI: 10.1016/j.bbi.2017.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/18/2023] Open
Abstract
Neuroinflammation is a pervasive feature of Alzheimer's disease (AD) and characterized by activated microglia, increased proinflammatory cytokines and/or infiltrating immune cells. T helper 17 (Th17) cells are found in AD brain parenchyma and interleukin-17A (IL-17A) is identified around deposits of aggregated amyloid β protein (Aβ). However, the role of IL-17A in AD pathogenesis remains elusive. We overexpressed IL-17A in an AD mouse model via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated intracranial gene delivery. AD model mice subjected to injection of a vehicle (PBS) or rAAV5 carrying the lacZ gene served as controls. IL-17A did not exacerbate neuroinflammation in IL-17A-overexpressing mice. We found that IL-17A overexpression markedly improved glucose metabolism, decreased soluble Aβ levels in the hippocampus and cerebrospinal fluid, drastically reduced cerebral amyloid angiopathy, and modestly but significantly improved anxiety and learning deficits. Moreover, the ATP-binding cassette subfamily A member 1 (ABCA1), which can transport Aβ from the brain into the blood circulation, significantly increased in IL-17A-overexpressing mice. In vitro treatment of brain endothelial bEnd.3 cells with IL-17A induced a dose-dependent increase in protein expression of ABCA1 through ERK activation. Our study suggests that IL-17A may decrease Aβ levels in the brain by upregulating ABCA1 in blood-brain barrier endothelial cells.
Collapse
|
236
|
Alnajjar S, Jin HK, Kang JE, Park SH, Rhie SJ. Intranasal Insulin for Alzheimer's Disease and Amnestic Mild Cognitive Impairment: Systematic Review and Meta-analysis. ACTA ACUST UNITED AC 2017. [DOI: 10.24304/kjcp.2017.27.3.161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Sarah Alnajjar
- Division of Life and Pharmaceutical Sciences Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hye Kyung Jin
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ji Eun Kang
- Division of Life and Pharmaceutical Sciences Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
- Department of Pharmacy, National Medical Center, Seoul 04564, Republic of Korea
| | - So Hyun Park
- Division of Life and Pharmaceutical Sciences Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sandy Jeong Rhie
- Division of Life and Pharmaceutical Sciences Graduate School, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
237
|
Gunn‐Moore D, Kaidanovich‐Beilin O, Iradi MCG, Gunn‐Moore F, Lovestone S. Alzheimer's disease in humans and other animals: A consequence of postreproductive life span and longevity rather than aging. Alzheimers Dement 2017; 14:195-204. [DOI: 10.1016/j.jalz.2017.08.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/19/2017] [Accepted: 08/19/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Danièlle Gunn‐Moore
- University of Edinburgh Royal (Dick) School of Veterinary Studies and The Roslin Institute Easter Bush Campus Roslin UK
| | | | - María Carolina Gallego Iradi
- University of Florida, College of Medicine Department of Neuroscience, Center for Translational Research in Neurodegenerative Diseases Gainesville FL USA
| | | | | |
Collapse
|
238
|
Bloom GS, Lazo JS, Norambuena A. Reduced brain insulin signaling: A seminal process in Alzheimer's disease pathogenesis. Neuropharmacology 2017; 136:192-195. [PMID: 28965829 DOI: 10.1016/j.neuropharm.2017.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 01/12/2023]
Abstract
The synaptic dysfunction and death of neurons that mediate memory and cognition account together for the behavioral symptoms of Alzheimer's disease (AD). Reduced insulin signaling in the brain is a hallmark of AD patients, even in the absence of systemic type 1 or type 2 diabetes, prompting some researchers to refer to AD as brain-specific, or type 3 diabetes. A key question that arises about this signature feature of AD is "how, if at all, does the brain's impaired ability to utilize insulin contribute to the behavioral deficits associated with AD?" The fact that type 2 diabetes is a risk factor for AD suggests a causative role for impaired insulin responsiveness in AD pathogenesis, but how that might occur at a detailed molecular level had been elusive. Here we review recent findings that mechanistically link soluble forms of amyloid-β (Aβ) and tau, the respective building blocks of the amyloid plaques and neurofibrillary tangles that accumulate in the brains of AD patients, with neuronal decline that is associated with poor insulin responsiveness and may begin long before AD symptoms become evident. We discuss how Aβ and tau work coordinately to deprive neurons of functionally accessible insulin receptors and dysregulate normal signaling by the protein kinase, mTOR. Finally, we suggest how newly gained knowledge about pathogenic signaling caused by reduced brain insulin signaling might be exploited for improved early detection and therapeutic intervention for AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- George S Bloom
- Department of Biology, University of Virginia, Charlottesville, VA, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| | - John S Lazo
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA; Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Andrés Norambuena
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
239
|
Zhao N, Liu CC, Van Ingelgom AJ, Martens YA, Linares C, Knight JA, Painter MM, Sullivan PM, Bu G. Apolipoprotein E4 Impairs Neuronal Insulin Signaling by Trapping Insulin Receptor in the Endosomes. Neuron 2017; 96:115-129.e5. [PMID: 28957663 PMCID: PMC5621659 DOI: 10.1016/j.neuron.2017.09.003] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 06/05/2017] [Accepted: 08/24/2017] [Indexed: 12/18/2022]
Abstract
Diabetes and impaired brain insulin signaling are linked to the pathogenesis of Alzheimer's disease (AD). The association between diabetes and AD-associated amyloid pathology is stronger among carriers of the apolipoprotein E (APOE) ε4 gene allele, the strongest genetic risk factor for late-onset AD. Here we report that apoE4 impairs neuronal insulin signaling in human apoE-targeted replacement (TR) mice in an age-dependent manner. High-fat diet (HFD) accelerates these effects in apoE4-TR mice at middle age. In primary neurons, apoE4 interacts with insulin receptor and impairs its trafficking by trapping it in the endosomes, leading to impaired insulin signaling and insulin-stimulated mitochondrial respiration and glycolysis. In aging brains, the increased apoE4 aggregation and compromised endosomal function further exacerbate the inhibitory effects of apoE4 on insulin signaling and related functions. Together, our study provides novel mechanistic insights into the pathogenic mechanisms of apoE4 and insulin resistance in AD.
Collapse
Affiliation(s)
- Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | | | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Joshua A Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Meghan M Painter
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Patrick M Sullivan
- Departments of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; GRECC, Durham Veterans Affairs Medical Center, Durham, NC 27705, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
240
|
Maimaiti S, Frazier HN, Anderson KL, Ghoweri AO, Brewer LD, Porter NM, Thibault O. Novel calcium-related targets of insulin in hippocampal neurons. Neuroscience 2017; 364:130-142. [PMID: 28939258 DOI: 10.1016/j.neuroscience.2017.09.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 01/28/2023]
Abstract
Both insulin signaling disruption and Ca2+ dysregulation are closely related to memory loss during aging and increase the vulnerability to Alzheimer's disease (AD). In hippocampal neurons, aging-related changes in calcium regulatory pathways have been shown to lead to higher intracellular calcium levels and an increase in the Ca2+-dependent afterhyperpolarization (AHP), which is associated with cognitive decline. Recent studies suggest that insulin reduces the Ca2+-dependent AHP. Given the sensitivity of neurons to insulin and evidence that brain insulin signaling is reduced with age, insulin-mediated alterations in calcium homeostasis may underlie the beneficial actions of insulin in the brain. Indeed, increasing insulin signaling in the brain via intranasal delivery has yielded promising results such as improving memory in both clinical and animal studies. However, while several mechanisms have been proposed, few have focused on regulation on intracellular Ca2+. In the present study, we further examined the effects of acute insulin on calcium pathways in primary hippocampal neurons in culture. Using the whole-cell patch-clamp technique, we found that acute insulin delivery reduced voltage-gated calcium currents. Fura-2 imaging was used to also address acute insulin effects on spontaneous and depolarization-mediated Ca2+ transients. Results indicate that insulin reduced Ca2+ transients, which appears to have involved a reduction in ryanodine receptor function. Together, these results suggest insulin regulates pathways that control intracellular Ca2+ which may reduce the AHP and improve memory. This may be one mechanism contributing to improved memory recall in response to intranasal insulin therapy in the clinic.
Collapse
Affiliation(s)
- Shaniya Maimaiti
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Hilaree N Frazier
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Katie L Anderson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Adam O Ghoweri
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Lawrence D Brewer
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Nada M Porter
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, University of Kentucky Medical Center, UKMC, MS-310; 800 Rose Street, Lexington, KY 40536, United States.
| |
Collapse
|
241
|
Chen Y, Guo Z, Mao YF, Zheng T, Zhang B. Intranasal Insulin Ameliorates Cerebral Hypometabolism, Neuronal Loss, and Astrogliosis in Streptozotocin-Induced Alzheimer's Rat Model. Neurotox Res 2017; 33:716-724. [PMID: 28929339 DOI: 10.1007/s12640-017-9809-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 11/27/2022]
Abstract
Intracerebroventricular injection of streptozotocin (ICV-STZ) in rodents leads to cognitive impairments and several pathological changes like Alzheimer's disease (AD). However, there is hardly any research about the effect of ICV-STZ on regional cerebral glucose metabolism in rodents. Previous studies have demonstrated that intranasal insulin improves cognition in AD patients. However, the underlying mechanism remains elusive. Here, we treated the ICV-STZ rats with daily intranasal delivery of insulin (2 U/day) for 6 consecutive weeks, then monitored 18F-fluorodeoxyglucose (18F-FDG) uptake using a high-resolution small-animal positron emission tomography (microPET) and studied the expression of neuronal nuclei (NeuN) and glial fibrillary acidic protein (GFAP) using immunohistochemical staining. We observed that 18F-FDG uptake decreased significantly at the prefrontal cortex, cingulate cortex, striatum, hippocampus, and entorhinal cortex in ICV-STZ rats as compared with the control rats. Intranasal insulin restores the cerebral glucose metabolism in prefrontal and cingulate cortex and attenuates astroglia activation and neuronal loss in the hippocampus of ICV-STZ rats. These findings provide the mechanistic basis for treating AD patients with intranasal insulin.
Collapse
Affiliation(s)
- Yanxing Chen
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No.88 Jiefang Road, Zhejiang, Hangzhou, 310009, China
| | - Zhangyu Guo
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No.88 Jiefang Road, Zhejiang, Hangzhou, 310009, China
| | - Yan-Fang Mao
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou, 310016, China
| | - Tingting Zheng
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No.88 Jiefang Road, Zhejiang, Hangzhou, 310009, China
| | - Baorong Zhang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, No.88 Jiefang Road, Zhejiang, Hangzhou, 310009, China.
| |
Collapse
|
242
|
Abstract
The relationship between obesity and cognitive impairment is important given the globally ageing population in whom cognitive decline and neurodegenerative disorders will carry grave individual, societal and financial burdens. This review examines the evidence for the link between obesity and cognitive function in terms of both the immediate effects on cognitive performance, and effects on the trajectory of cognitive ageing and likelihood of dementia. In mid-life, there is a strong association between obesity and impaired cognitive function. Anthropometric measures of obesity are also associated with reduced neural integrity (e.g. grey and white matter atrophy). Increasing age coupled with the negative metabolic consequences of obesity (e.g. type 2 diabetes mellitus) are likely to significantly contribute to cognitive decline and incidence of dementia. Stress is identified as a potential risk factor promoting abdominal obesity and contributing to impaired cognitive function. However, the potentially protective effects of obesity against cognitive decline in older age require further examination. Finally, surgical and whole diet interventions, which address obesity may improve cognitive capacity and confer some protection against later cognitive decline. In conclusion, obesity and its comorbidities are associated with impaired cognitive performance, accelerated cognitive decline and neurodegenerative pathologies such as dementia in later life. Interventions targeting mid-life obesity may prove beneficial in reducing the cognitive risks associated with obesity.
Collapse
|
243
|
The diabetic brain and cognition. J Neural Transm (Vienna) 2017; 124:1431-1454. [PMID: 28766040 DOI: 10.1007/s00702-017-1763-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022]
Abstract
The prevalence of both Alzheimer's disease (AD) and vascular dementia (VaD) is increasing with the aging of the population. Studies from the last several years have shown that people with diabetes have an increased risk for dementia and cognitive impairment. Therefore, the authors of this consensus review tried to elaborate on the role of diabetes, especially diabetes type 2 (T2DM) in both AD and VaD. Based on the clinical and experimental work of scientists from 18 countries participating in the International Congress on Vascular Disorders and on literature search using PUBMED, it can be concluded that T2DM is a risk factor for both, AD and VaD, based on a pathology of glucose utilization. This pathology is the consequence of a disturbance of insulin-related mechanisms leading to brain insulin resistance. Although the underlying pathological mechanisms for AD and VaD are different in many aspects, the contribution of T2DM and insulin resistant brain state (IRBS) to cerebrovascular disturbances in both disorders cannot be neglected. Therefore, early diagnosis of metabolic parameters including those relevant for T2DM is required. Moreover, it is possible that therapeutic options utilized today for diabetes treatment may also have an effect on the risk for dementia. T2DM/IRBS contribute to pathological processes in AD and VaD.
Collapse
|
244
|
Insulin Treatment Prevents Neuroinflammation and Neuronal Injury with Restored Neurobehavioral Function in Models of HIV/AIDS Neurodegeneration. J Neurosci 2017; 36:10683-10695. [PMID: 27733618 DOI: 10.1523/jneurosci.1287-16.2016] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/26/2016] [Indexed: 12/17/2022] Open
Abstract
HIV-1 infection of the brain causes the neurodegenerative syndrome HIV-associated neurocognitive disorders (HAND), for which there is no specific treatment. Herein, we investigated the actions of insulin using ex vivo and in vivo models of HAND. Increased neuroinflammatory gene expression was observed in brains from patients with HIV/AIDS. The insulin receptor was detected on both neurons and glia, but its expression was unaffected by HIV-1 infection. Insulin treatment of HIV-infected primary human microglia suppressed supernatant HIV-1 p24 levels, reduced CXCL10 and IL-6 transcript levels, and induced peroxisome proliferator-activated receptor gamma (PPAR-γ) expression. Insulin treatment of primary human neurons prevented HIV-1 Vpr-mediated cell process retraction and death. In feline immunodeficiency virus (FIV) infected cats, daily intranasal insulin treatment (20.0 IU/200 μl for 6 weeks) reduced CXCL10, IL-6, and FIV RNA detection in brain, although PPAR-γ in glia was increased compared with PBS-treated FIV+ control animals. These molecular changes were accompanied by diminished glial activation in cerebral cortex and white matter of insulin-treated FIV+ animals, with associated preservation of cortical neurons. Neuronal counts in parietal cortex, striatum, and hippocampus were higher in the FIV+/insulin-treated group compared with the FIV+/PBS-treated group. Moreover, intranasal insulin treatment improved neurobehavioral performance, including both memory and motor functions, in FIV+ animals. Therefore, insulin exerted ex vivo and in vivo antiviral, anti-inflammatory, and neuroprotective effects in models of HAND, representing a new therapeutic option for patients with inflammatory or infectious neurodegenerative disorders including HAND. SIGNIFICANCE STATEMENT HIV-associated neurocognitive disorders (HAND) represent a spectrum disorder of neurocognitive dysfunctions resulting from HIV-1 infection. Although the exact mechanisms causing HAND are unknown, productive HIV-1 infection in the brain with associated neuroinflammation is a potential pathogenic mechanism resulting in neuronal damage and death. We report that, in HIV-infected microglia cultures, insulin treatment led to reduced viral replication and inflammatory gene expression. In addition, intranasal insulin treatment of experimentally feline immunodeficiency virus-infected animals resulted in improved motor and memory performances. We show that insulin restored expression of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), which is suppressed by HIV-1 replication. Our findings indicate a unique function for insulin in improving neurological outcomes in lentiviral infections, implicating insulin as a therapeutic intervention for HAND.
Collapse
|
245
|
D'Cunha NM, McKune AJ, Panagiotakos DB, Georgousopoulou EN, Thomas J, Mellor DD, Naumovski N. Evaluation of dietary and lifestyle changes as modifiers of S100β levels in Alzheimer's disease. Nutr Neurosci 2017; 22:1-18. [PMID: 28696163 DOI: 10.1080/1028415x.2017.1349032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is a significant body of research undertaken in order to elucidate the mechanisms underlying the pathology of Alzheimer's disease (AD), as well as to discover early detection biomarkers and potential therapeutic strategies. One such proposed biomarker is the calcium binding protein S100β, which, depending on its local concentration, is known to exhibit both neurotrophic and neuroinflammatory properties in the central nervous system. At present, relatively little is known regarding the effect of chronic S100β disruption in AD. Dietary intake has been identified as a modifiable risk factor for AD. Preliminary in vitro and animal studies have demonstrated an association between S100β expression and dietary intake which links to AD pathophysiology. This review describes the association of S100β to fatty acids, ketone bodies, insulin, and botanicals as well as the potential impact of physical activity as a lifestyle factor. We also discuss the prospective implications of these findings, including support of the use of a Mediterranean dietary pattern and/or the ketogenic diet as an approach to modify AD risk.
Collapse
Affiliation(s)
- Nathan M D'Cunha
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Andrew J McKune
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,c University of Canberra, Research Institute for Sport and Exercise , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,d Discipline of Biokinetics, Exercise and Leisure Sciences, School of Health Sciences , University of KwaZulu-Natal , Durban 4041 , South Africa
| | - Demosthenes B Panagiotakos
- e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Ekavi N Georgousopoulou
- b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia.,e Department of Nutrition-Dietetics, School of Health and Education , Harokopio University , Athens 176 71 , Greece
| | - Jackson Thomas
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Duane D Mellor
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| | - Nenad Naumovski
- a University of Canberra Health Research Institute (UCHRI) , University of Canberra , Locked Bag 1, Bruce , Canberra ACT 2601 , Australia.,b Collaborative Research in Bioactives and Biomarkers Group (CRIBB) , University of Canberra , Bruce , Canberra ACT 2601 , Australia
| |
Collapse
|
246
|
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
|
247
|
Bell GA, Fadool DA. Awake, long-term intranasal insulin treatment does not affect object memory, odor discrimination, or reversal learning in mice. Physiol Behav 2017; 174:104-113. [PMID: 28259806 PMCID: PMC5639911 DOI: 10.1016/j.physbeh.2017.02.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 02/23/2017] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
Intranasal insulin delivery is currently being used in clinical trials to test for improvement in human memory and cognition, and in particular, for lessening memory loss attributed to neurodegenerative diseases. Studies have reported the effects of short-term intranasal insulin treatment on various behaviors, but less have examined long-term effects. The olfactory bulb contains the highest density of insulin receptors in conjunction with the highest level of insulin transport within the brain. Previous research from our laboratory has demonstrated that acute insulin intranasal delivery (IND) enhanced both short- and long-term memory as well as increased two-odor discrimination in a two-choice paradigm. Herein, we investigated the behavioral and physiological effects of chronic insulin IND. Adult, male C57BL6/J mice were intranasally treated with 5μg/μl of insulin twice daily for 30 and 60days. Metabolic assessment indicated no change in body weight, caloric intake, or energy expenditure following chronic insulin IND, but an increase in the frequency of meal bouts selectively in the dark cycle. Unlike acute insulin IND, which has been shown to cause enhanced performance in odor habituation/dishabituation and two-odor discrimination tasks in mice, chronic insulin IND did not enhance olfactometry-based odorant discrimination or olfactory reversal learning. In an object memory recognition task, insulin IND-treated mice did not perform differently than controls, regardless of task duration. Biochemical analyses of the olfactory bulb revealed a modest 1.3 fold increase in IR kinase phosphorylation but no significant increase in Kv1.3 phosphorylation. Substrate phosphorylation of IR kinase downstream effectors (MAPK/ERK and Akt signaling) proved to be highly variable. These data indicate that chronic administration of insulin IND in mice fails to enhance olfactory ability, object memory recognition, or a majority of systems physiology metabolic factors - as reported to elicit a modulatory effect with acute administration. This leads to two alternative interpretations regarding long-term insulin IND in mice: 1) It causes an initial stage of insulin resistance to dampen the behaviors that would normally be modulated under acute insulin IND, but ability to clear a glucose challenge is still retained, or 2) There is a lack of behavioral modulation at high concentration of insulin attributed to the twice daily intervals of hyperinsulinemia caused by insulin IND administration without any insulin resistance, per se.
Collapse
Affiliation(s)
- Genevieve A Bell
- Department of Biological Science and Program in Neuroscience, The Florida State University, Tallahassee, FL 32306-4295, United States
| | - Debra Ann Fadool
- Department of Biological Science and Program in Neuroscience, The Florida State University, Tallahassee, FL 32306-4295, United States; Institute of Molecular Biophysics, The Florida State University, Tallahassee, FL 32306-4380, United States.
| |
Collapse
|
248
|
Chen Y, Dai CL, Wu Z, Iqbal K, Liu F, Zhang B, Gong CX. Intranasal Insulin Prevents Anesthesia-Induced Cognitive Impairment and Chronic Neurobehavioral Changes. Front Aging Neurosci 2017; 9:136. [PMID: 28539885 PMCID: PMC5424543 DOI: 10.3389/fnagi.2017.00136] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/24/2017] [Indexed: 11/26/2022] Open
Abstract
General anesthesia increases the risk for cognitive impairment post operation, especially in the elderly and vulnerable individuals. Recent animal studies on the impact of anesthesia on postoperative cognitive impairment have provided some valuable insights, but much remains to be understood. Here, by using mice of various ages and conditions, we found that anesthesia with propofol and sevoflurane caused significant deficits in spatial learning and memory, as tested using Morris Water Maze (MWM) 2–6 days after anesthesia exposure, in aged (17–18 months old) wild-type (WT) mice and in adult (7–8 months old) 3xTg-AD mice (a triple transgenic mouse model of Alzheimer’s disease (AD)), but not in adult WT mice. Anesthesia resulted in long-term neurobehavioral changes in the fear conditioning task carried out 65 days after exposure to anesthesia in 3xTg-AD mice. Importantly, daily intranasal administration of insulin (1.75 U/mouse/day) for only 3 days prior to anesthesia completely prevented the anesthesia-induced deficits in spatial learning and memory and the long-term neurobehavioral changes tested 65 days after exposure to anesthesia in 3xTg-AD mice. These results indicate that aging and AD-like brain pathology increase the vulnerability to cognitive impairment after anesthesia and that intranasal treatment with insulin can prevent anesthesia-induced cognitive impairment.
Collapse
Affiliation(s)
- Yanxing Chen
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, China.,Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA
| | - Chun-Ling Dai
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA
| | - Zhe Wu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA.,Department of Cell Biology and Genetics, School of Basic Medicine, Hubei University of Science and TechnologyXianning, China
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental DisabilitiesNew York, NY, USA
| |
Collapse
|
249
|
Mullins RJ, Diehl TC, Chia CW, Kapogiannis D. Insulin Resistance as a Link between Amyloid-Beta and Tau Pathologies in Alzheimer's Disease. Front Aging Neurosci 2017; 9:118. [PMID: 28515688 PMCID: PMC5413582 DOI: 10.3389/fnagi.2017.00118] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/11/2017] [Indexed: 12/19/2022] Open
Abstract
Current hypotheses and theories regarding the pathogenesis of Alzheimer’s disease (AD) heavily implicate brain insulin resistance (IR) as a key factor. Despite the many well-validated metrics for systemic IR, the absence of biomarkers for brain-specific IR represents a translational gap that has hindered its study in living humans. In our lab, we have been working to develop biomarkers that reflect the common mechanisms of brain IR and AD that may be used to follow their engagement by experimental treatments. We present two promising biomarkers for brain IR in AD: insulin cascade mediators probed in extracellular vesicles (EVs) enriched for neuronal origin, and two-dimensional magnetic resonance spectroscopy (MRS) measures of brain glucose. As further evidence for a fundamental link between brain IR and AD, we provide a novel analysis demonstrating the close spatial correlation between brain expression of genes implicated in IR (using Allen Human Brain Atlas data) and tau and beta-amyloid pathologies. We proceed to propose the bold hypotheses that baseline differences in the metabolic reliance on glycolysis, and the expression of glucose transporters (GLUT) and insulin signaling genes determine the vulnerability of different brain regions to Tau and/or Amyloid beta (Aβ) pathology, and that IR is a critical link between these two pathologies that define AD. Lastly, we provide an overview of ongoing clinical trials that target IR as an angle to treat AD, and suggest how biomarkers may be used to evaluate treatment efficacy and target engagement.
Collapse
Affiliation(s)
- Roger J Mullins
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH)Baltimore, MD, USA
| | - Thomas C Diehl
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH)Baltimore, MD, USA
| | - Chee W Chia
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH)Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health (NIA/NIH)Baltimore, MD, USA
| |
Collapse
|
250
|
Simó R, Ciudin A, Simó-Servat O, Hernández C. Cognitive impairment and dementia: a new emerging complication of type 2 diabetes-The diabetologist's perspective. Acta Diabetol 2017; 54:417-424. [PMID: 28210868 DOI: 10.1007/s00592-017-0970-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/30/2017] [Indexed: 12/24/2022]
Abstract
Type 2 diabetes mellitus (T2D) and Alzheimer's disease (AD) are two of the most common diseases of aging around the world. Given the frequency with which T2D and AD occur, the notion that people with T2D may be at increased risk for AD has large societal consequences, and understanding the mechanistic links between these diseases is imperative for the development of effective AD prevention and treatment strategies. Apart from being an accelerator of AD, T2D is associated with a progressive cognitive decline. Impaired insulin signaling, inflammation, the accumulation of advanced glycation end-products and oxidative stress all play an essential role in the pathogenesis of both AD and diabetic complications. Therefore, it is reasonable to postulate that these pathways are involved in the increased risk of dementia that occurs in the T2D population. The early diagnosis of cognitive impairment and the identification of the subset of patients at a higher risk of developing AD is a challenge for healthcare providers, and meeting it will permit us to implement a personalized medicine, which is an essential issue in diabetes care with significant therapeutic implications. The main gaps that should be filled to achieve this objective are examined.
Collapse
Affiliation(s)
- Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| | - Andreea Ciudin
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Olga Simó-Servat
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Hernández
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Pg. Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|