1
|
Kumaree KK, Brimson JM, Verma K, Chuchawankul S, Tencomnao T, Prasansuklab A. Agarwood leaf ethanol extract provides neuroprotective properties and promotes cholinergic differentiation of HT22 hippocampal neurons. Sci Rep 2025; 15:10230. [PMID: 40133398 PMCID: PMC11937462 DOI: 10.1038/s41598-025-93462-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Neurodegenerative diseases, characterized by the loss or damage of neurons, represent a growing global health concern. Plants are a rich source of naturally occurring compounds with immense therapeutic potential. Among them, Aquilaria crassna (commonly known as agarwood) is a precious fragrant plant extensively used in cosmetics, perfumes, and traditional Asian medicine. However, its neuroprotective role, particularly in neuroregeneration, has been minimally explored. This study aimed to investigate the therapeutic potential of agarwood leaves in promoting neuroregeneration, with a focus on cholinergic function and neural differentiation. To identify bioactive compounds, a comprehensive LC-MS analysis was conducted on agarwood ethanolic extract (AWE). The phytochemicals detected were further evaluated using in silico methods to predict their interaction with receptor proteins linked to neurodegenerative diseases. Virtual screening revealed that several compounds in AWE exhibited strong binding affinities to receptors such as sigma-1, TrkB, Nogo-66, and p75NTR, providing insights into the potential mechanisms underlying its neuroprotective effects. The in-silico findings were validated through in vitro experiments using HT-22 mouse hippocampal cells as a model. AWE treatment led to a dose-dependent increase in the expression of marker proteins associated with neural differentiation and regeneration, including neuronal nuclei (NeuN), growth-associated protein 43 (GAP43), synaptophysin (Syn), brain-derived neurotrophic factor (BDNF), and the sigma-1 receptor. Additionally, AWE enhanced the expression of specific markers for cholinergic neurons, demonstrating its influence on neuronal development and synaptic function. These findings provide compelling evidence of AWE's neuroprotective properties, highlighting its potential as a therapeutic agent for neurodegenerative diseases.
Collapse
Affiliation(s)
- Kishoree K Kumaree
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence on Natural Products for Neuroprotection and Anti-ageing (Neur-Age NatChula), Chulalongkorn University, Bangkok, 10330, Thailand
| | - James M Brimson
- Center of Excellence on Natural Products for Neuroprotection and Anti-ageing (Neur-Age NatChula), Chulalongkorn University, Bangkok, 10330, Thailand
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kanika Verma
- Center of Excellence on Natural Products for Neuroprotection and Anti-ageing (Neur-Age NatChula), Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Siriporn Chuchawankul
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tewin Tencomnao
- Center of Excellence on Natural Products for Neuroprotection and Anti-ageing (Neur-Age NatChula), Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Anchalee Prasansuklab
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence on Natural Products for Neuroprotection and Anti-ageing (Neur-Age NatChula), Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
2
|
Fisher A, Levey AI. CNS muscarinic receptors and muscarinic receptor agonists in Alzheimer disease treatment. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:161-184. [PMID: 40340060 DOI: 10.1016/b978-0-443-19088-9.00016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
This review explores the main aspects that form the basis of the cholinergic-oriented treatment of Alzheimer disease. Muscarinic acetylcholine receptor subtypes in the brain and periphery are discussed. It includes a new and updated overview of the involvement of muscarinic receptors in Alzheimer disease and the recent development of new and highly selective M1 muscarinic receptor agonists with disease-modifying potential. Activation of the M1 muscarinic receptor is a rational therapeutic strategy for the treatment of schizophrenia and Alzheimer disease, as this receptor plays a pivotal role in modulating cognitive deficits and the pathology of the disease. Such activation can be achieved through M1 allosteric and bitopic muscarinic agonists, M1 positive allosteric modulators (M1 PAMs), and direct-acting M1 muscarinic orthosteric agonists. The efficacy of M1 PAMs depends on acetylcholine, which declines in Alzheimer disease as postsynaptic neurons lose cholinergic input from the basal forebrain. On the other hand, the activity of M1 muscarinic orthosteric agonists is independent of the functional or anatomic integrity of presynaptic cholinergic terminals, and likely retain efficacy as the disease progresses, even after presynaptic degeneration of cholinergic fibers. Based on the acceptance criteria for a preferred M1 muscarinic agonist for the treatment of AD, aiming for efficacy, specificity, and safety in clinical use, few M1 muscarinic agonists fulfill these requirements, such as orthosteric M1 agonists-cevimeline (aka AF102B), the first FDA-approved M1 agonist, and NSC001 (aka AF267B). The pros and cons of various muscarinic agonists developed are critically discussed in comparison to these drugs. The review proposes new alternatives to cholinergic therapy, particularly selective M1 muscarinic drugs, that should be designed to amplify its clinical effect and supplement the disease-modifying effect of new treatments to slow down or arrest disease progression.
Collapse
Affiliation(s)
- Abraham Fisher
- ADPD: Advances in Science & Therapy, International Conference on Alzheimer's and Parkinson's Diseases and Related Neurological Disorders, Lisbon, Portugal.
| | - Allan I Levey
- Department of Neurology, Emory University, Atlanta, GA, United States
| |
Collapse
|
3
|
Rathee S, Sen D, Pandey V, Jain SK. Advances in Understanding and Managing Alzheimer's Disease: From Pathophysiology to Innovative Therapeutic Strategies. Curr Drug Targets 2024; 25:752-774. [PMID: 39039673 DOI: 10.2174/0113894501320096240627071400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder characterized by the presence of amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles, leading to cognitive and physical decline. Representing the majority of dementia cases, AD poses a significant burden on healthcare systems globally, with onset typically occurring after the age of 65. While most cases are sporadic, about 10% exhibit autosomal forms associated with specific gene mutations. Neurofibrillary tangles and Aβ plaques formed by misfolded tau proteins and Aβ peptides contribute to neuronal damage and cognitive impairment. Currently, approved drugs, such as acetylcholinesterase inhibitors and N-methyl D-aspartate receptor agonists, offer only partial symptomatic relief without altering disease progression. A promising development is using lecanemab, a humanized IgG1 monoclonal antibody, as an immune therapeutic approach. Lecanemab demonstrates selectivity for polymorphic Aβ variants and binds to large soluble Aβ aggregates, providing a potential avenue for targeted treatment. This shift in understanding the role of the adaptive immune response in AD pathogenesis opens new possibilities for therapeutic interventions aiming to address the disease's intricate mechanisms. This review aims to summarize recent advancements in understanding Alzheimer's disease pathophysiology and innovative therapeutic approaches, providing valuable insights for both researchers and clinicians.
Collapse
Affiliation(s)
- Sunny Rathee
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Debasis Sen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Vishal Pandey
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| |
Collapse
|
4
|
Moreno-Rodriguez M, Perez SE, Martinez-Gardeazabal J, Manuel I, Malek-Ahmadi M, Rodriguez-Puertas R, Mufson EJ. Frontal Cortex Lipid Alterations During the Onset of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1515-1532. [PMID: 38578893 DOI: 10.3233/jad-231485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Although sporadic Alzheimer's disease (AD) is a neurodegenerative disorder of unknown etiology, familial AD is associated with specific gene mutations. A commonality between these forms of AD is that both display multiple pathogenic events including cholinergic and lipid dysregulation. Objective We aimed to identify the relevant lipids and the activity of their related receptors in the frontal cortex and correlating them with cognition during the progression of AD. Methods MALDI-mass spectrometry imaging (MSI) and functional autoradiography was used to evaluate the distribution of phospholipids/sphingolipids and the activity of cannabinoid 1 (CB1), sphingosine 1-phosphate 1 (S1P1), and muscarinic M2/M4 receptors in the frontal cortex (FC) of people that come to autopsy with premortem clinical diagnosis of AD, mild cognitive impairment (MCI), and no cognitive impairment (NCI). Results MALDI-MSI revealed an increase in myelin-related lipids, such as diacylglycerol (DG) 36:1, DG 38:5, and phosphatidic acid (PA) 40:6 in the white matter (WM) in MCI compared to NCI, and a downregulation of WM phosphatidylinositol (PI) 38:4 and PI 38:5 levels in AD compared to NCI. Elevated levels of phosphatidylcholine (PC) 32:1, PC 34:0, and sphingomyelin 38:1 were observed in discrete lipid accumulations in the FC supragranular layers during disease progression. Muscarinic M2/M4 receptor activation in layers V-VI decreased in AD compared to MCI. CB1 receptor activity was upregulated in layers V-VI, while S1P1 was downregulated within WM in AD relative to NCI. Conclusions FC WM lipidomic alterations are associated with myelin dyshomeostasis in prodromal AD, suggesting WM lipid maintenance as a potential therapeutic target for dementia.
Collapse
Affiliation(s)
- Marta Moreno-Rodriguez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Ivan Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | | | - Rafael Rodriguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
5
|
Tiepolt S, Meyer PM, Patt M, Deuther-Conrad W, Hesse S, Barthel H, Sabri O. PET Imaging of Cholinergic Neurotransmission in Neurodegenerative Disorders. J Nucl Med 2022; 63:33S-44S. [PMID: 35649648 DOI: 10.2967/jnumed.121.263198] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Indexed: 12/13/2022] Open
Abstract
As a neuromodulator, the neurotransmitter acetylcholine plays an important role in cognitive, mood, locomotor, sleep/wake, and olfactory functions. In the pathophysiology of most neurodegenerative diseases, such as Alzheimer disease (AD) or Lewy body disorder (LBD), cholinergic receptors, transporters, or enzymes are involved and relevant as imaging targets. The aim of this review is to summarize current knowledge on PET imaging of cholinergic neurotransmission in neurodegenerative diseases. For PET imaging of presynaptic vesicular acetylcholine transporters (VAChT), (-)-18F-fluoroethoxybenzovesamicol (18F-FEOBV) was the first PET ligand that could be successfully translated to clinical application. Since then, the number of 18F-FEOBV PET investigations on patients with AD or LBD has grown rapidly and provided novel, important findings concerning the pathophysiology of AD and LBD. Regarding the α4β2 nicotinic acetylcholine receptors (nAChRs), various second-generation PET ligands, such as 18F-nifene, 18F-AZAN, 18F-XTRA, (-)-18F-flubatine, and (+)-18F-flubatine, were developed and successfully translated to human application. In neurodegenerative diseases such as AD and LBD, PET imaging of α4β2 nAChRs is of special value for monitoring disease progression and drugs directed to α4β2 nAChRs. For PET of α7 nAChR, 18F-ASEM and 11C-MeQAA were successfully applied in mild cognitive impairment and AD, respectively. The highest potential for α7 nAChR PET is seen in staging, in evaluating disease progression, and in therapy monitoring. PET of selective muscarinic acetylcholine receptors (mAChRs) is still in an early stage, as the development of subtype-selective radioligands is complicated. Promising radioligands to image mAChR subtypes M1 (11C-LSN3172176), M2 (18F-FP-TZTP), and M4 (11C-MK-6884) were developed and successfully translated to humans. PET imaging of mAChRs is relevant for the assessment and monitoring of therapies in AD and LBD. PET of acetylcholine esterase activity has been investigated since the 1990s. Many PET studies with 11C-PMP and 11C-MP4A demonstrated cortical cholinergic dysfunction in dementia associated with AD and LBD. Recent studies indicated a solid relationship between subcortical and cortical cholinergic dysfunction and noncognitive dysfunctions such as balance and gait in LBD. Taken together, PET of distinct components of cholinergic neurotransmission is of great interest for diagnosis, disease monitoring, and therapy monitoring and to gain insight into the pathophysiology of different neurodegenerative disorders.
Collapse
Affiliation(s)
- Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| | - Philipp M Meyer
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| | | | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; and
| |
Collapse
|
6
|
Kondak C, Riedel G, Harrington CR, Wischik CM, Klein J. Hydromethylthionine enhancement of central cholinergic signalling is blocked by rivastigmine and memantine. J Neurochem 2021; 160:172-184. [PMID: 34855998 DOI: 10.1111/jnc.15553] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/26/2021] [Accepted: 11/30/2021] [Indexed: 12/30/2022]
Abstract
The prevention of tau protein aggregations is a therapeutic goal for the treatment of Alzheimer's disease (AD), and hydromethylthionine (HMT) (also known as leucomethylthioninium-mesylate [LMTM]), is a potent inhibitor of tau aggregation in vitro and in vivo. In two Phase 3 clinical trials in AD, HMT had greater pharmacological activity on clinical endpoints in patients not receiving approved symptomatic treatments for AD (acetylcholinesterase (AChE) inhibitors and/or memantine) despite different mechanisms of action. To investigate this drug interaction in an animal model, we used tau-transgenic L1 and wild-type NMRI mice treated with rivastigmine or memantine prior to adding HMT, and measured changes in hippocampal acetylcholine (ACh) by microdialysis. HMT given alone doubled hippocampal ACh levels in both mouse lines and increased stimulated ACh release induced by exploration of the open field or by infusion of scopolamine. Rivastigmine increased ACh release in both mouse lines, whereas memantine was more active in tau-transgenic L1 mice. Importantly, our study revealed a negative interaction between HMT and symptomatic AD drugs: the HMT effect was completely eliminated in mice that had been pre-treated with either rivastigmine or memantine. Rivastigmine was found to inhibit AChE, whereas HMT and memantine had no effects on AChE or on choline acetyltransferase (ChAT). The interactions observed in this study demonstrate that HMT enhances cholinergic activity in mouse brain by a mechanism of action unrelated to AChE inhibition. Our findings establish that the drug interaction that was first observed clinically has a neuropharmacological basis and is not restricted to animals with tau aggregation pathology. Given the importance of the cholinergic system for memory function, the potential for commonly used AD drugs to interfere with the treatment effects of disease-modifying drugs needs to be taken into account in the design of clinical trials.
Collapse
Affiliation(s)
- Constantin Kondak
- Institute of Medical Sciences, Translational Neuroscience, University of Aberdeen, Aberdeen, Scotland.,Institute of Pharmacology and Clinical Pharmacy, Goethe University Frankfurt, Frankfurt, Germany
| | - Gernot Riedel
- Institute of Medical Sciences, Translational Neuroscience, University of Aberdeen, Aberdeen, Scotland
| | - Charles R Harrington
- Institute of Medical Sciences, Translational Neuroscience, University of Aberdeen, Aberdeen, Scotland.,TauRx Therapeutics Ltd, Aberdeen, Scotland
| | - Claude M Wischik
- Institute of Medical Sciences, Translational Neuroscience, University of Aberdeen, Aberdeen, Scotland.,TauRx Therapeutics Ltd, Aberdeen, Scotland
| | - Jochen Klein
- Institute of Pharmacology and Clinical Pharmacy, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
7
|
Boer CG, Hatzikotoulas K, Southam L, Stefánsdóttir L, Zhang Y, Coutinho de Almeida R, Wu TT, Zheng J, Hartley A, Teder-Laving M, Skogholt AH, Terao C, Zengini E, Alexiadis G, Barysenka A, Bjornsdottir G, Gabrielsen ME, Gilly A, Ingvarsson T, Johnsen MB, Jonsson H, Kloppenburg M, Luetge A, Lund SH, Mägi R, Mangino M, Nelissen RRGHH, Shivakumar M, Steinberg J, Takuwa H, Thomas LF, Tuerlings M, Babis GC, Cheung JPY, Kang JH, Kraft P, Lietman SA, Samartzis D, Slagboom PE, Stefansson K, Thorsteinsdottir U, Tobias JH, Uitterlinden AG, Winsvold B, Zwart JA, Davey Smith G, Sham PC, Thorleifsson G, Gaunt TR, Morris AP, Valdes AM, Tsezou A, Cheah KSE, Ikegawa S, Hveem K, Esko T, Wilkinson JM, Meulenbelt I, Lee MTM, van Meurs JBJ, Styrkársdóttir U, Zeggini E. Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell 2021; 184:4784-4818.e17. [PMID: 34450027 PMCID: PMC8459317 DOI: 10.1016/j.cell.2021.07.038] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/26/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis affects over 300 million people worldwide. Here, we conduct a genome-wide association study meta-analysis across 826,690 individuals (177,517 with osteoarthritis) and identify 100 independently associated risk variants across 11 osteoarthritis phenotypes, 52 of which have not been associated with the disease before. We report thumb and spine osteoarthritis risk variants and identify differences in genetic effects between weight-bearing and non-weight-bearing joints. We identify sex-specific and early age-at-onset osteoarthritis risk loci. We integrate functional genomics data from primary patient tissues (including articular cartilage, subchondral bone, and osteophytic cartilage) and identify high-confidence effector genes. We provide evidence for genetic correlation with phenotypes related to pain, the main disease symptom, and identify likely causal genes linked to neuronal processes. Our results provide insights into key molecular players in disease processes and highlight attractive drug targets to accelerate translation.
Collapse
Affiliation(s)
- Cindy G Boer
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Tian T Wu
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - April Hartley
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045, Japan
| | - Eleni Zengini
- 4(th) Psychiatric Department, Dromokaiteio Psychiatric Hospital, 12461 Athens, Greece
| | - George Alexiadis
- 1(st) Department of Orthopaedics, KAT General Hospital, 14561 Athens, Greece
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland; Department of Orthopedic Surgery, Akureyri Hospital, 600 Akureyri, Iceland
| | - Marianne B Johnsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway; Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, 0424 Oslo, Norway
| | - Helgi Jonsson
- Department of Medicine, Landspitali The National University Hospital of Iceland, 108 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Margreet Kloppenburg
- Departments of Rheumatology and Clinical Epidemiology, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Almut Luetge
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | | | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Rob R G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia Steinberg
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 1340, Australia
| | - Hiroshi Takuwa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan; Department of Orthopedic Surgery, Shimane University, Shimane 693-8501, Japan
| | - Laurent F Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; BioCore-Bioinformatics Core Facility, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - George C Babis
- 2(nd) Department of Orthopaedics, National and Kapodistrian University of Athens, Medical School, Nea Ionia General Hospital Konstantopouleio, 14233 Athens, Greece
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jae Hee Kang
- Department of Medicine, Brigham and Women's Hospital, 181 Longwood Ave, Boston, MA 02115, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Steven A Lietman
- Musculoskeletal Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Kari Stefansson
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Bendik Winsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - John-Anker Zwart
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Pak Chung Sham
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester M13 9LJ, UK
| | - Ana M Valdes
- Faculty of Medicine and Health Sciences, School of Medicine, University of Nottingham, Nottingham, Nottinghamshire NG5 1PB, UK
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa 411 10, Greece
| | - Kathryn S E Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - J Mark Wilkinson
- Department of Oncology and Metabolism and Healthy Lifespan Institute, University of Sheffield, Sheffield S10 2RX, UK
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA; Institute of Biomedical Sciences, Academia Sinica, 115 Taipei, Taiwan
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | | | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, 81675 Munich, Germany.
| |
Collapse
|
8
|
Bakker C, van der Aart J, Labots G, Liptrot J, Cross DM, Klaassen ES, Dickinson S, Tasker T, Groeneveld GJ. Safety and Pharmacokinetics of HTL0018318, a Novel M 1 Receptor Agonist, Given in Combination with Donepezil at Steady State: A Randomized Trial in Healthy Elderly Subjects. Drugs R D 2021; 21:295-304. [PMID: 34164794 PMCID: PMC8363684 DOI: 10.1007/s40268-021-00352-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 10/27/2022] Open
Abstract
INTRODUCTION HTL0018318 is a selective muscarinic M1 receptor partial agonist under development for the symptomatic treatment of dementias, including Alzheimer's disease. Clinically, HTL0018318 would likely be used alone or in conjunction with cholinesterase inhibitors (e.g. donepezil). OBJECTIVE We investigated the safety, tolerability, and pharmacokinetics of HTL0018318 given alone and in combination with donepezil. METHODS This was a randomized, double-blind, placebo-controlled trial in 42 (to deliver 36 with combination treatment) healthy elderly subjects investigating the effects of oral HTL0018318 15 and 25 mg given alone and combined with donepezil 10 mg at steady state on adverse events (AEs), vital signs, saliva production, sleep quality, pulmonary function, subjective feelings, and pharmacokinetics. RESULTS AEs were reported by lower percentages of subjects after HTL0018318 alone than after donepezil alone. There was no increase in the percentage of subjects reporting AEs after co-administration than after donepezil alone. Supine systolic blood pressure was 1.6 mmHg (95% confidence interval [CI] -3.1 to -0.1) lower after HTL0018318 alone than after combination treatment. This was comparable with results from placebo alone: 1.7 mmHg (95% CI -3.2 to 0.2) lower than with combination treatment. Supine pulse rate was 3.3 bpm (95% CI 1.5-5.1) higher after HTL0018318 alone than with co-administration. HTL0018318 and donepezil did not meaningfully affect each other's pharmacokinetics. CONCLUSION HTL0018318 was well tolerated when given alone and in combination with donepezil. HTL0018318 and donepezil do not demonstrate pharmacokinetic or pharmacodynamic interactions, indicating that HTL0018318 can be safely administered in combination with donepezil. CLINICAL TRIAL REGISTRATION Netherlands Trial register identifier NL5915, registered on 28 October 2016.
Collapse
Affiliation(s)
- Charlotte Bakker
- Centre for Human Drug Research (CDHR), Leiden, The Netherlands.
- Leids Universitair Medisch Centrum, Leiden, The Netherlands.
| | | | - Geert Labots
- Centre for Human Drug Research (CDHR), Leiden, The Netherlands
| | | | | | | | | | | | - Geert Jan Groeneveld
- Centre for Human Drug Research (CDHR), Leiden, The Netherlands
- Leids Universitair Medisch Centrum, Leiden, The Netherlands
| |
Collapse
|
9
|
Geula C, Dunlop SR, Ayala I, Kawles AS, Flanagan ME, Gefen T, Mesulam MM. Basal forebrain cholinergic system in the dementias: Vulnerability, resilience, and resistance. J Neurochem 2021; 158:1394-1411. [PMID: 34272732 PMCID: PMC8458251 DOI: 10.1111/jnc.15471] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 01/15/2023]
Abstract
The basal forebrain cholinergic neurons (BFCN) provide the primary source of cholinergic innervation of the human cerebral cortex. They are involved in the cognitive processes of learning, memory, and attention. These neurons are differentially vulnerable in various neuropathologic entities that cause dementia. This review summarizes the relevance to BFCN of neuropathologic markers associated with dementias, including the plaques and tangles of Alzheimer's disease (AD), the Lewy bodies of diffuse Lewy body disease, the tauopathy of frontotemporal lobar degeneration (FTLD-TAU) and the TDP-43 proteinopathy of FTLD-TDP. Each of these proteinopathies has a different relationship to BFCN and their corticofugal axons. Available evidence points to early and substantial degeneration of the BFCN in AD and diffuse Lewy body disease. In AD, the major neurodegenerative correlate is accumulation of phosphotau in neurofibrillary tangles. However, these neurons are less vulnerable to the tauopathy of FTLD. An intriguing finding is that the intracellular tau of AD causes destruction of the BFCN, whereas that of FTLD does not. This observation has profound implications for exploring the impact of different species of tauopathy on neuronal survival. The proteinopathy of FTLD-TDP shows virtually no abnormal inclusions within the BFCN. Thus, the BFCN are highly vulnerable to the neurodegenerative effects of tauopathy in AD, resilient to the neurodegenerative effect of tauopathy in FTLD and apparently resistant to the emergence of proteinopathy in FTLD-TDP and perhaps also in Pick's disease. Investigations are beginning to shed light on the potential mechanisms of this differential vulnerability and their implications for therapeutic intervention.
Collapse
Affiliation(s)
- Changiz Geula
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Sara R Dunlop
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Ivan Ayala
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Allegra S Kawles
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Margaret E Flanagan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine Chicago, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
10
|
Llorente-Ovejero A, Martínez-Gardeazabal J, Moreno-Rodríguez M, Lombardero L, González de San Román E, Manuel I, Giralt MT, Rodríguez-Puertas R. Specific Phospholipid Modulation by Muscarinic Signaling in a Rat Lesion Model of Alzheimer's Disease. ACS Chem Neurosci 2021; 12:2167-2181. [PMID: 34037379 PMCID: PMC9162383 DOI: 10.1021/acschemneuro.1c00169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
![]()
Alzheimer’s disease (AD) represents
the most common cause
of dementia worldwide and has been consistently associated with the
loss of basal forebrain cholinergic neurons (BFCNs) leading to impaired
cholinergic neurotransmission, aberrant synaptic function, and altered
structural lipid metabolism. In this sense, membrane phospholipids
(PLs) can be used for de novo synthesis of choline (Ch) for the further
obtaining of acetylcholine (ACh) when its availability is compromised.
Specific lipid species involved in the metabolism of Ch have been
identified as possible biomarkers of phenoconversion to AD. Using
a rat model of BFCN lesion, we have evaluated the lipid composition
and muscarinic signaling in brain areas related to cognitive processes.
The loss of BFCN resulted in alterations of varied lipid species related
to Ch metabolism at nucleus basalis magnocellularis (NMB) and cortical
projection areas. The activity of muscarinic receptors (mAChR) was
decreased in the NMB and increased in the hippocampus according to
the subcellular distribution of M1/M2 mAChR
which could explain the learning and memory impairment reported in
this AD rat model. These results suggest that the modulation of specific
lipid metabolic routes could represent an alternative therapeutic
strategy to potentiate cholinergic neurotransmission and preserve
cell membrane integrity in AD.
Collapse
Affiliation(s)
- Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Laura Lombardero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Estíbaliz González de San Román
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
- Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - María Teresa Giralt
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
- Neurodegenerative Diseases, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
11
|
Bakker C, Prins S, Liptrot J, Hart EP, Klaassen ES, Brown GA, Brown A, Congreve M, Weir M, Marshall FH, Stevens J, Cross DM, Tasker T, Nathan PJ, Groeneveld GJ. Safety, pharmacokinetics and pharmacodynamics of HTL0009936, a selective muscarinic M 1 -acetylcholine receptor agonist: A randomized cross-over trial. Br J Clin Pharmacol 2021; 87:4439-4449. [PMID: 33891333 PMCID: PMC8596821 DOI: 10.1111/bcp.14872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 12/01/2022] Open
Abstract
AIMS HTL0009936 is a selective M1 muscarinic receptor agonist in development for cognitive dysfunction in Alzheimer's disease. Safety, tolerability and pharmacokinetics and exploratory pharmacodynamic effects of HTL0009936 administered by continuous IV infusion at steady state were investigated in elderly subjects with below average cognitive functioning (BACF). METHODS Part A was a four-treatment open label sequential study in healthy elderly investigating 10-83 mg HTL0009936 (IV) and a 24 mg HTL0009936 single oral dose. Part B was a five-treatment randomized, double-blind, placebo and physostigmine controlled cross-over study with IV HTL0009936 in elderly subjects with BACF. Pharmacodynamic assessments were performed using neurocognitive and electrophysiological tests. RESULTS Pharmacokinetics of HTL0009936 showed dose-proportional increases in exposure with a mean half-life of 2.4 hours. HTL0009936 was well-tolerated with transient dose-related adverse events (AEs). Small increases in mean systolic blood pressure of 7.12 mmHg (95% CI [3.99-10.24]) and in diastolic of 5.32 mmHg (95% CI [3.18-7.47]) were noted at the highest dose in part B. Overall, there was suggestive, but no definitive, positive or negative pharmacodynamic effects. Statistically significant effects were observed on P300 with HTL0009936 and adaptive tracking with physostigmine. CONCLUSIONS HTL0009936 showed well-characterized pharmacokinetics and single doses were safe and generally well-tolerated in healthy elderly subjects. Due to physostigmine tolerability issues and subject burden, the study design was changed and some pharmacodynamic assessments (neurocognitive) were performed at suboptimal drug exposures. Therefore no clear conclusions can be made on pharmacodynamic effects of HTL0009936, although an effect on P300 is suggestive of central target engagement.
Collapse
Affiliation(s)
- Charlotte Bakker
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | - Samantha Prins
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ellen P Hart
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | | | | | | | | | - Fiona H Marshall
- Sosei Heptares, Cambridge, UK.,MSD Research Laboratories (Merck & Co), Kenilworth, New Jersey, USA
| | - Jasper Stevens
- Centre for Human Drug Research, Leiden, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Pradeep J Nathan
- Sosei Heptares, Cambridge, UK.,Department of Psychiatry, University of Cambridge, Cambridge, UK.,School of Psychological Sciences, Monash University, Australia
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Slater C, Wang Q. Alzheimer's disease: An evolving understanding of noradrenergic involvement and the promising future of electroceutical therapies. Clin Transl Med 2021; 11:e397. [PMID: 33931975 PMCID: PMC8087948 DOI: 10.1002/ctm2.397] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) poses a significant global health concern over the next several decades. Multiple hypotheses have been put forth that attempt to explain the underlying pathophysiology of AD. Many of these are briefly reviewed here, but to-date no disease-altering therapy has been achieved. Despite this, recent work expanding on the role of noradrenergic system dysfunction in both the pathogenesis and symptomatic exacerbation of AD has shown promise. The role norepinephrine (NE) plays in AD remains complicated but pre-tangle tau has consistently been shown to arise in the locus coeruleus (LC) of patients with AD decades before symptom onset. The current research reviewed here indicates NE can facilitate neuroprotective and memory-enhancing effects through β adrenergic receptors, while α2A adrenergic receptors may exacerbate amyloid toxicity through a contribution to tau hyperphosphorylation. AD appears to involve a disruption in the balance between these two receptors and their various subtypes. There is also a poorly characterized interplay between the noradrenergic and cholinergic systems. LC deterioration leads to maladaptation in the remaining LC-NE system and subsequently inhibits cholinergic neuron function, eventually leading to the classic cholinergic disruption seen in AD. Understanding AD as a dysfunctional noradrenergic system, provides new avenues for the use of advanced neural stimulation techniques to both study and therapeutically target the earliest stages of neuropathology. Direct LC stimulation and non-invasive vagus nerve stimulation (VNS) have both demonstrated potential use as AD therapeutics. Significant work remains, though, to better understand the role of the noradrenergic system in AD and how electroceuticals can provide disease-altering treatments.
Collapse
Affiliation(s)
- Cody Slater
- Department of Biomedical EngineeringColumbia UniversityNew YorkNew YorkUSA
- Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Qi Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
13
|
Chen J, Cho KE, Skwarzynska D, Clancy S, Conley NJ, Clinton SM, Li X, Lin L, Zhu JJ. The Property-Based Practical Applications and Solutions of Genetically Encoded Acetylcholine and Monoamine Sensors. J Neurosci 2021; 41:2318-2328. [PMID: 33627325 PMCID: PMC7984589 DOI: 10.1523/jneurosci.1062-19.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 12/26/2022] Open
Abstract
Neuromodulatory communication among various neurons and non-neuronal cells mediates myriad physiological and pathologic processes, yet defining regulatory and functional features of neuromodulatory transmission remains challenging because of limitations of available monitoring tools. Recently developed genetically encoded neuromodulatory transmitter sensors, when combined with superresolution and/or deconvolution microscopy, allow the first visualization of neuromodulatory transmission with nanoscale or microscale spatiotemporal resolution. In vitro and in vivo experiments have validated several high-performing sensors to have the qualities necessary for demarcating fundamental synaptic properties of neuromodulatory transmission, and initial analysis has unveiled unexpected fine control and precision of neuromodulation. These new findings underscore the importance of synaptic dynamics in synapse-, subcellular-, and circuit-specific neuromodulation, as well as the prospect of genetically encoded transmitter sensors in expanding our knowledge of various behaviors and diseases, including Alzheimer's disease, sleeping disorders, tumorigenesis, and many others.
Collapse
Affiliation(s)
- Jun Chen
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University
- Pharmaceutical Sciences Graduate Program, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Katriel E Cho
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908
- Tools for Modern Neurobiology Class of 2020, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Daria Skwarzynska
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908
- Tools for Modern Neurobiology Class of 2020, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Shaylyn Clancy
- Tools for Modern Neurobiology Class of 2020, University of Virginia School of Medicine, Charlottesville, Virginia 22908
- Cell and Developmental Biology Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Nicholas J Conley
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia 22908
- Tools for Modern Neurobiology Class of 2020, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061
| | - Xiaokun Li
- Pharmaceutical Sciences Graduate Program, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Li Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University
- Pharmaceutical Sciences Graduate Program, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - J Julius Zhu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
14
|
Vijayraghavan S, Everling S. Neuromodulation of Persistent Activity and Working Memory Circuitry in Primate Prefrontal Cortex by Muscarinic Receptors. Front Neural Circuits 2021; 15:648624. [PMID: 33790746 PMCID: PMC8005543 DOI: 10.3389/fncir.2021.648624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/11/2021] [Indexed: 12/31/2022] Open
Abstract
Neuromodulation by acetylcholine plays a vital role in shaping the physiology and functions of cerebral cortex. Cholinergic neuromodulation influences brain-state transitions, controls the gating of cortical sensory stimulus responses, and has been shown to influence the generation and maintenance of persistent activity in prefrontal cortex. Here we review our current understanding of the role of muscarinic cholinergic receptors in primate prefrontal cortex during its engagement in the performance of working memory tasks. We summarize the localization of muscarinic receptors in prefrontal cortex, review the effects of muscarinic neuromodulation on arousal, working memory and cognitive control tasks, and describe the effects of muscarinic M1 receptor stimulation and blockade on the generation and maintenance of persistent activity of prefrontal neurons encoding working memory representations. Recent studies describing the pharmacological effects of M1 receptors on prefrontal persistent activity demonstrate the heterogeneity of muscarinic actions and delineate unexpected modulatory effects discovered in primate prefrontal cortex when compared with studies in rodents. Understanding the underlying mechanisms by which muscarinic receptors regulate prefrontal cognitive control circuitry will inform the search of muscarinic-based therapeutic targets in the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Susheel Vijayraghavan
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada
| | - Stefan Everling
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Robarts Research Institute, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
15
|
Bakker C, Tasker T, Liptrot J, Hart EP, Klaassen ES, Prins S, van der Doef TF, Brown GA, Brown A, Congreve M, Weir M, Marshall FH, Cross DM, Groeneveld GJ, Nathan PJ. First-in-man study to investigate safety, pharmacokinetics and exploratory pharmacodynamics of HTL0018318, a novel M 1 -receptor partial agonist for the treatment of dementias. Br J Clin Pharmacol 2021; 87:2945-2955. [PMID: 33351971 PMCID: PMC8359307 DOI: 10.1111/bcp.14710] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/09/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022] Open
Abstract
AIMS HTL0018318 is a selective M1 receptor partial agonist currently under development for the symptomatic treatment of cognitive and behavioural symptoms in Alzheimer's disease and other dementias. We investigated safety, tolerability, pharmacokinetics and exploratory pharmacodynamics (PD) of HTL0018318 following single ascending doses. METHODS This randomized, double-blind, placebo-controlled study in 40 healthy younger adult and 57 healthy elderly subjects, investigated oral doses of 1-35 mg HTL0018318. Pharmacodynamic assessments were performed using a battery of neurocognitive tasks and electrophysiological measurements. Cerebrospinal fluid concentrations of HTL0018318 and food effects on pharmacokinetics of HTL0018318 were investigated in an open label and partial cross-over design in 14 healthy subjects. RESULTS Pharmacokinetics of HTL0018318 were well-characterized showing dose proportional increases in exposure from 1-35 mg. Single doses of HTL0018318 were associated with mild dose-related adverse events of low incidence in both younger adult and elderly subjects. The most frequently reported cholinergic AEs included hyperhidrosis and increases in blood pressure up to 10.3 mmHg in younger adults (95% CI [4.2-16.3], 35-mg dose) and up to 11.9 mmHg in elderly subjects (95% CI [4.9-18.9], 15-mg dose). There were no statistically significant effects on cognitive function but the study was not powered to detect small to moderate effect sizes of clinical relevance. CONCLUSION HTL0018318 showed well-characterized pharmacokinetics and following single doses were generally well tolerated in the dose range studied. These provide encouraging data in support of the development for HTL0018318 for Alzheimer's disease and other dementias.
Collapse
Affiliation(s)
| | | | | | - Ellen P Hart
- Centre for Human Drug Research (CDHR), Leiden, Netherlands
| | | | - Samantha Prins
- Centre for Human Drug Research (CDHR), Leiden, Netherlands
| | | | | | | | | | | | | | | | - Geert Jan Groeneveld
- Centre for Human Drug Research (CDHR), Leiden, Netherlands.,Leiden University Medical Centre, Leiden, The Netherlands
| | - Pradeep J Nathan
- Sosei Heptares, Cambridge, UK.,Department of Psychiatry, University of Cambridge, UK.,School of Psychological Sciences, Monash University, Australia
| |
Collapse
|
16
|
Se Thoe E, Fauzi A, Tang YQ, Chamyuang S, Chia AYY. A review on advances of treatment modalities for Alzheimer's disease. Life Sci 2021; 276:119129. [PMID: 33515559 DOI: 10.1016/j.lfs.2021.119129] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/10/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which is mainly characterized by progressive impairment in cognition, emotion, language and memory in older population. Considering the impact of AD, formulations of pharmaceutical drugs and cholinesterase inhibitors have been widely propagated, receiving endorsement by FDA as a form of AD treatment. However, these medications were gradually discovered to be ineffective in removing the root of AD pathogenesis but merely targeting the symptoms so as to improve a patient's cognitive outcome. Hence, a search for better disease-modifying alternatives is put into motion. Having a clear understanding of the neuroprotective mechanisms and diverse properties undertaken by specific genes, antibodies and nanoparticles is central towards designing novel therapeutic agents. In this review, we provide a brief introduction on the background of Alzheimer's disease, the biology of blood-brain barrier, along with the potentials and drawbacks associated with current therapeutic treatment avenues pertaining to gene therapy, immunotherapy and nanotherapy for better diagnosis and management of Alzheimer's disease.
Collapse
Affiliation(s)
- Ewen Se Thoe
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Ayesha Fauzi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Yin Quan Tang
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Sunita Chamyuang
- School of Science, Mae Fah Luang University, Chaing Rai 57100, Thailand; Microbial Products and Innovation Research Group, Mae Fah Luang University, Chaing Rai 57100, Thailand
| | - Adeline Yoke Yin Chia
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, 47500 Selangor, Malaysia.
| |
Collapse
|
17
|
Vermeiren Y, Van Dam D, de Vries M, De Deyn PP. Psychiatric Disorders in Dementia. PET AND SPECT IN PSYCHIATRY 2021:317-385. [DOI: 10.1007/978-3-030-57231-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Przybyłowska M, Dzierzbicka K, Kowalski S, Chmielewska K, Inkielewicz-Stepniak I. Therapeutic Potential of Multifunctional Derivatives of Cholinesterase Inhibitors. Curr Neuropharmacol 2021; 19:1323-1344. [PMID: 33342413 PMCID: PMC8719290 DOI: 10.2174/1570159x19666201218103434] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/07/2020] [Accepted: 11/29/2020] [Indexed: 11/22/2022] Open
Abstract
The aim of this work is to review tacrine analogues from the last three years, which were not included in the latest review work, donepezil and galantamine hybrids from 2015 and rivastigmine derivatives from 2014. In this account, we summarize the efforts toward the development and characterization of non-toxic inhibitors of cholinesterases based on mentioned drugs with various interesting additional properties such as antioxidant, decreasing β-amyloid plaque aggregation, nitric oxide production, pro-inflammatory cytokines release, monoamine oxidase-B activity, cytotoxicity and oxidative stress in vitro and in animal model that classify these hybrids as potential multifunctional therapeutic agents for Alzheimer's disease. Moreover, herein, we have described the cholinergic hypothesis, mechanisms of neurodegeneration and current pharmacotherapy of Alzheimer's disease based on the restoration of cholinergic function through blocking enzymes that break down acetylcholine.
Collapse
Affiliation(s)
- Maja Przybyłowska
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Krystyna Dzierzbicka
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Szymon Kowalski
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Klaudia Chmielewska
- Department of Organic Chemistry, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
19
|
Lin L, Gupta S, Zheng WS, Si K, Zhu JJ. Genetically encoded sensors enable micro- and nano-scopic decoding of transmission in healthy and diseased brains. Mol Psychiatry 2021; 26:443-455. [PMID: 33277628 PMCID: PMC7850973 DOI: 10.1038/s41380-020-00960-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/06/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022]
Abstract
Neural communication orchestrates a variety of behaviors, yet despite impressive effort, delineating transmission properties of neuromodulatory communication remains a daunting task due to limitations of available monitoring tools. Recently developed genetically encoded neurotransmitter sensors, when combined with superresolution and deconvolution microscopic techniques, enable the first micro- and nano-scopic visualization of neuromodulatory transmission. Here we introduce this image analysis method by presenting its biophysical foundation, practical solutions, biological validation, and broad applicability. The presentation illustrates how the method resolves fundamental synaptic properties of neuromodulatory transmission, and the new data unveil unexpected fine control and precision of rodent and human neuromodulation. The findings raise the prospect of rapid advances in the understanding of neuromodulatory transmission essential for resolving the physiology or pathogenesis of various behaviors and diseases.
Collapse
Affiliation(s)
- Li Lin
- Department of Neurosurgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China. .,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Smriti Gupta
- grid.27755.320000 0000 9136 933XDepartment of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908 USA
| | - W. Sharon Zheng
- grid.27755.320000 0000 9136 933XDepartment of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908 USA ,grid.27755.320000 0000 9136 933XBiomedical Engineering Class of 2021, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Ke Si
- grid.13402.340000 0004 1759 700XCollege of Optical Science and Engineering, Zhejiang University, Hangzhou, 310027 China ,grid.13402.340000 0004 1759 700XSchool of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310027 China
| | - J. Julius Zhu
- grid.27755.320000 0000 9136 933XDepartment of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908 USA
| |
Collapse
|
20
|
Vishwas S, Awasthi A, Corrie L, Kumar Singh S, Gulati M. Multiple target-based combination therapy of galantamine, memantine and lycopene for the possible treatment of Alzheimer’s disease. Med Hypotheses 2020; 143:109879. [DOI: 10.1016/j.mehy.2020.109879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022]
|
21
|
Zhu PK, Zheng WS, Zhang P, Jing M, Borden PM, Ali F, Guo K, Feng J, Marvin JS, Wang Y, Wan J, Gan L, Kwan AC, Lin L, Looger LL, Li Y, Zhang Y. Nanoscopic Visualization of Restricted Nonvolume Cholinergic and Monoaminergic Transmission with Genetically Encoded Sensors. NANO LETTERS 2020; 20:4073-4083. [PMID: 32396366 PMCID: PMC7519949 DOI: 10.1021/acs.nanolett.9b04877] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
How neuromodulatory transmitters diffuse into the extracellular space remains an unsolved fundamental biological question, despite wide acceptance of the volume transmission model. Here, we report development of a method combining genetically encoded fluorescent sensors with high-resolution imaging and analysis algorithms which permits the first direct visualization of neuromodulatory transmitter diffusion at various neuronal and non-neuronal cells. Our analysis reveals that acetylcholine and monoamines diffuse at individual release sites with a spread length constant of ∼0.75 μm. These transmitters employ varied numbers of release sites, and when spatially close-packed release sites coactivate they can spillover into larger subcellular areas. Our data indicate spatially restricted (i.e., nonvolume) neuromodulatory transmission to be a prominent intercellular communication mode, reshaping current thinking of control and precision of neuromodulation crucial for understanding behaviors and diseases.
Collapse
Affiliation(s)
- Paula K. Zhu
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Math, Engineering & Science Academy Class of 2020, Albemarle High School, Charlottesville, VA 22901
- Summer Secondary School Neurobiology Class of 2019, Harvard University, Cambridge, MA 02138
- Current address: Undergraduate Class of 2024, Harvard College, Cambridge, MA 02138
| | - W. Sharon Zheng
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
- Department of Biomedical Engineering Class of 2021, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Peng Zhang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Miao Jing
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing 100871, China
| | - Philip M. Borden
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
- Current address: LifeEDIT, Research Triangle Park, NC 27709
| | - Farhan Ali
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511
| | - Kaiming Guo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jonathan S. Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Yali Wang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Li Gan
- Helen and Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine College, New York, NY 10065
| | - Alex C. Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Loren L. Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Yulong Li
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yajun Zhang
- State Key Laboratory of Membrane Biology and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| |
Collapse
|
22
|
Yi JH, Whitcomb DJ, Park SJ, Martinez-Perez C, Barbati SA, Mitchell SJ, Cho K. M1 muscarinic acetylcholine receptor dysfunction in moderate Alzheimer's disease pathology. Brain Commun 2020; 2:fcaa058. [PMID: 32766549 PMCID: PMC7391992 DOI: 10.1093/braincomms/fcaa058] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/03/2020] [Accepted: 04/13/2020] [Indexed: 01/08/2023] Open
Abstract
Aggregation of amyloid beta and loss of cholinergic innervation in the brain are predominant components of Alzheimer’s disease pathology and likely underlie cognitive impairment. Acetylcholinesterase inhibitors are one of the few treatment options for Alzheimer’s disease, where levels of available acetylcholine are enhanced to counteract the cholinergic loss. However, these inhibitors show limited clinical efficacy. One potential explanation for this is a concomitant dysregulation of cholinergic receptors themselves as a consequence of the amyloid beta pathology. We tested this hypothesis by examining levels of M1 muscarinic acetylcholine receptors in the temporal cortex from seven Alzheimer’s disease and seven non-disease age-matched control brain tissue samples (control: 85 ± 2.63 years old, moderate Alzheimer’s disease: 84 ± 2.32 years old, P-value = 0.721; eight female and six male patients). The samples were categorized into two groups: ‘control’ (Consortium to Establish a Registry for Alzheimer’s Disease diagnosis of ‘No Alzheimer’s disease’, and Braak staging pathology of I–II) and ‘moderate Alzheimer’s disease’ (Consortium to Establish a Registry for Alzheimer’s Disease diagnosis of ‘possible/probable Alzheimer’s disease’, and Braak staging pathology of IV). We find that in comparison to age-matched controls, there is a loss of M1 muscarinic acetylcholine receptors in moderate Alzheimer’s disease tissue (control: 2.17 ± 0.27 arbitrary units, n = 7, Mod-AD: 0.83 ± 0.16 arbitrary units, n = 7, two-tailed t-test, t = 4.248, P = 0.00113). Using a functional rat cortical brain slice model, we find that postsynaptic muscarinic acetylcholine receptor function is dysregulated by aberrant amyloid beta-mediated activation of metabotropic glutamate receptor 5. Crucially, blocking metabotropic glutamate receptor 5 restores muscarinic acetylcholine receptor function and object recognition memory in 5XFAD transgenic mice. This indicates that the amyloid beta-mediated activation of metabotropic glutamate receptor 5 negatively regulates muscarinic acetylcholine receptor and illustrates the importance of muscarinic acetylcholine receptors as a potential disease-modifying target in the moderate pathological stages of Alzheimer’s disease.
Collapse
Affiliation(s)
- Jee Hyun Yi
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS1 3NY, UK
| | - Daniel J Whitcomb
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS1 3NY, UK
| | - Se Jin Park
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee East-West Pharmaceutical Research Institute, College of Pharmacy, Kyung Hee University, Seoul 130-701, Korea
| | - Celia Martinez-Perez
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS1 3NY, UK
| | - Saviana A Barbati
- UK Dementia Research Institute at King's College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Scott J Mitchell
- UK Dementia Research Institute at King's College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Kwangwook Cho
- Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol BS1 3NY, UK.,UK Dementia Research Institute at King's College London, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| |
Collapse
|
23
|
Jakubik J, El-Fakahany EE. Current Advances in Allosteric Modulation of Muscarinic Receptors. Biomolecules 2020; 10:biom10020325. [PMID: 32085536 PMCID: PMC7072599 DOI: 10.3390/biom10020325] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/07/2023] Open
Abstract
Allosteric modulators are ligands that bind to a site on the receptor that is spatially separated from the orthosteric binding site for the endogenous neurotransmitter. Allosteric modulators modulate the binding affinity, potency, and efficacy of orthosteric ligands. Muscarinic acetylcholine receptors are prototypical allosterically-modulated G-protein-coupled receptors. They are a potential therapeutic target for the treatment of psychiatric, neurologic, and internal diseases like schizophrenia, Alzheimer’s disease, Huntington disease, type 2 diabetes, or chronic pulmonary obstruction. Here, we reviewed the progress made during the last decade in our understanding of their mechanisms of binding, allosteric modulation, and in vivo actions in order to understand the translational impact of studying this important class of pharmacological agents. We overviewed newly developed allosteric modulators of muscarinic receptors as well as new spin-off ideas like bitopic ligands combining allosteric and orthosteric moieties and photo-switchable ligands based on bitopic agents.
Collapse
Affiliation(s)
- Jan Jakubik
- Department of Neurochemistry, Institute of Physiology CAS, 142 20 Prague, Czech Republic
- Correspondence: (J.J.); (E.E.E.-F.)
| | - Esam E. El-Fakahany
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
- Correspondence: (J.J.); (E.E.E.-F.)
| |
Collapse
|
24
|
Sanchez-Mejias E, Nuñez-Diaz C, Sanchez-Varo R, Gomez-Arboledas A, Garcia-Leon JA, Fernandez-Valenzuela JJ, Mejias-Ortega M, Trujillo-Estrada L, Baglietto-Vargas D, Moreno-Gonzalez I, Davila JC, Vitorica J, Gutierrez A. Distinct disease-sensitive GABAergic neurons in the perirhinal cortex of Alzheimer's mice and patients. Brain Pathol 2019; 30:345-363. [PMID: 31491047 PMCID: PMC7064898 DOI: 10.1111/bpa.12785] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/02/2019] [Indexed: 12/29/2022] Open
Abstract
Neuronal loss is the best neuropathological substrate that correlates with cortical atrophy and dementia in Alzheimer's disease (AD). Defective GABAergic neuronal functions may lead to cortical network hyperactivity and aberrant neuronal oscillations and in consequence, generate a detrimental alteration in memory processes. In this study, using immunohistochemical and stereological approaches, we report that the two major and non-overlapping groups of inhibitory interneurons (SOM-cells and PV-cells) displayed distinct vulnerability in the perirhinal cortex of APP/PS1 mice and AD patients. SOM-positive neurons were notably sensitive and exhibited a dramatic decrease in the perirhinal cortex of 6-month-old transgenic mice (57% and 61% in areas 36 and 35, respectively) and, most importantly, in AD patients (91% in Braak V-VI cases). In addition, this interneuron degenerative process seems to occur in parallel, and closely related, with the progression of the amyloid pathology. However, the population expressing PV was unaffected in APP/PS1 mice while in AD brains suffered a pronounced and significant loss (69%). As a key component of cortico-hippocampal networks, the perirhinal cortex plays an important role in memory processes, especially in familiarity-based memory recognition. Therefore, disrupted functional connectivity of this cortical region, as a result of the early SOM and PV neurodegeneration, might contribute to the altered brain rhythms and cognitive failures observed in the initial clinical phase of AD patients. Finally, these findings highlight the failure of amyloidogenic AD models to fully recapitulate the selective neuronal degeneration occurring in humans.
Collapse
Affiliation(s)
- Elisabeth Sanchez-Mejias
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Cristina Nuñez-Diaz
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Raquel Sanchez-Varo
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Angela Gomez-Arboledas
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Antonio Garcia-Leon
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Jose Fernandez-Valenzuela
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marina Mejias-Ortega
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Laura Trujillo-Estrada
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - David Baglietto-Vargas
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ines Moreno-Gonzalez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jose Carlos Davila
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Vitorica
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS)-Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Antonia Gutierrez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
25
|
Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, Khachaturian AS, Vergallo A, Cavedo E, Snyder PJ, Khachaturian ZS. The cholinergic system in the pathophysiology and treatment of Alzheimer's disease. Brain 2019; 141:1917-1933. [PMID: 29850777 DOI: 10.1093/brain/awy132] [Citation(s) in RCA: 1052] [Impact Index Per Article: 175.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Cholinergic synapses are ubiquitous in the human central nervous system. Their high density in the thalamus, striatum, limbic system, and neocortex suggest that cholinergic transmission is likely to be critically important for memory, learning, attention and other higher brain functions. Several lines of research suggest additional roles for cholinergic systems in overall brain homeostasis and plasticity. As such, the brain's cholinergic system occupies a central role in ongoing research related to normal cognition and age-related cognitive decline, including dementias such as Alzheimer's disease. The cholinergic hypothesis of Alzheimer's disease centres on the progressive loss of limbic and neocortical cholinergic innervation. Neurofibrillary degeneration in the basal forebrain is believed to be the primary cause for the dysfunction and death of forebrain cholinergic neurons, giving rise to a widespread presynaptic cholinergic denervation. Cholinesterase inhibitors increase the availability of acetylcholine at synapses in the brain and are one of the few drug therapies that have been proven clinically useful in the treatment of Alzheimer's disease dementia, thus validating the cholinergic system as an important therapeutic target in the disease. This review includes an overview of the role of the cholinergic system in cognition and an updated understanding of how cholinergic deficits in Alzheimer's disease interact with other aspects of disease pathophysiology, including plaques composed of amyloid-β proteins. This review also documents the benefits of cholinergic therapies at various stages of Alzheimer's disease and during long-term follow-up as visualized in novel imaging studies. The weight of the evidence supports the continued value of cholinergic drugs as a standard, cornerstone pharmacological approach in Alzheimer's disease, particularly as we look ahead to future combination therapies that address symptoms as well as disease progression.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | - M-Marsel Mesulam
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
| | - Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ezio Giacobini
- Department of Internal Medicine, Rehabilitation and Geriatrics, University of Geneva Hospitals, Geneva, Switzerland
| | - George T Grossberg
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, St Louis, MO, USA
| | - Ara S Khachaturian
- The Campaign to Prevent Alzheimer's Disease by 2020 (PAD2020), Potomac, MD, USA
| | - Andrea Vergallo
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | - Enrica Cavedo
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | - Peter J Snyder
- Department of Neurology, Alpert Medical School of Brown University, Providence, RI USA.,Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | | |
Collapse
|
26
|
Muramatsu I, Uwada J, Yoshiki H, Sada K, Lee K, Yazawa T, Taniguchi T, Nishio M, Ishibashi T, Masuoka T. Novel regulatory systems for acetylcholine release in rat striatum and anti‐Alzheimer's disease drugs. J Neurochem 2019; 149:605-623. [DOI: 10.1111/jnc.14701] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 02/20/2019] [Accepted: 03/27/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Ikunobu Muramatsu
- Department of Pharmacology School of Medicine Kanazawa Medical University Uchinada, Ishikawa Japan
- Division of Genomic Science and Microbiology School of Medicine University of Fukui Eiheiji Fukui Japan
- Kimura Hospital Awara Fukui Japan
| | - Junsuke Uwada
- Division of Cellular Signal Transduction Department of Biochemistry Asahikawa Medical University Asahikawa Hokkaido Japan
| | - Hatsumi Yoshiki
- Division of Genomic Science and Microbiology School of Medicine University of Fukui Eiheiji Fukui Japan
| | - Kiyonao Sada
- Division of Genomic Science and Microbiology School of Medicine University of Fukui Eiheiji Fukui Japan
| | - Kung‐Shing Lee
- Division of Genomic Science and Microbiology School of Medicine University of Fukui Eiheiji Fukui Japan
- Department of Surgery Kaohsiung Medical University Kaohsiung Taiwan
| | - Takashi Yazawa
- Division of Cellular Signal Transduction Department of Biochemistry Asahikawa Medical University Asahikawa Hokkaido Japan
| | - Takanobu Taniguchi
- Division of Cellular Signal Transduction Department of Biochemistry Asahikawa Medical University Asahikawa Hokkaido Japan
| | - Matomo Nishio
- Department of Pharmacology School of Medicine Kanazawa Medical University Uchinada, Ishikawa Japan
| | - Takaharu Ishibashi
- Department of Pharmacology School of Medicine Kanazawa Medical University Uchinada, Ishikawa Japan
| | - Takayoshi Masuoka
- Department of Pharmacology School of Medicine Kanazawa Medical University Uchinada, Ishikawa Japan
| |
Collapse
|
27
|
Zink N, Bensmann W, Arning L, Stock AK, Beste C. CHRM2 Genotype Affects Inhibitory Control Mechanisms During Cognitive Flexibility. Mol Neurobiol 2019; 56:6134-6141. [PMID: 30729426 DOI: 10.1007/s12035-019-1521-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/30/2019] [Indexed: 12/30/2022]
Abstract
The cholinergic system is one of the most important neurotransmitter systems, but knowledge about the relevance of the cholinergic muscarinergic receptor system for cognitive functions is still scarce. Evidence suggests that the cholinergic muscarinic 2 receptor (CHRM2) plays an important role in the processing of cueing/prior information that help to increase the efficacy of lower-level attentional processes. In the current study, we investigated whether this is also the case for higher-level cognitive flexibility mechanisms. To this end, we tested N = 210 healthy adults with a backward inhibition task, in which prior information needs to be used to guide cognitive flexibility mechanisms. Testing different polymorphisms of the CHRM2 gene, we found that variation in this gene play a role in cognitive flexibility. It could be demonstrated that rs8191992 TT genotype carriers are better able to suppress no longer relevant information and to use prior information for cognitive flexibility, compared to A allele carriers. We further found that rs2350780 GG genotype carriers performed worse than A allele carriers. The results broaden the relevance of the CHRM2 system for cognitive functions beyond attentional selection processes. Corroborating recent theories on the relevance of the cholinergic system for cognitive processes, these results suggest that CHRM2 is important to process of "prior information" needed to inform subsequent cognitive operations. Considering the importance of prior information for adaptive behavioral control, it is possible that CHRM2 also modulates other instances of higher-level cognitive processes as long as these require the processing of "prior information."
Collapse
Affiliation(s)
- Nicolas Zink
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Schubertstraße 42, 01309, Dresden, Germany
| | - Wiebke Bensmann
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Schubertstraße 42, 01309, Dresden, Germany
| | - Larissa Arning
- Department of Human Genetics, Ruhr-University Bochum, Bochum, Germany
| | - Ann-Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Schubertstraße 42, 01309, Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Schubertstraße 42, 01309, Dresden, Germany.
| |
Collapse
|
28
|
Zhang J, Chen SR, Chen H, Pan HL. RE1-silencing transcription factor controls the acute-to-chronic neuropathic pain transition and Chrm2 receptor gene expression in primary sensory neurons. J Biol Chem 2018; 293:19078-19091. [PMID: 30327427 DOI: 10.1074/jbc.ra118.005846] [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] [Received: 09/12/2018] [Revised: 10/12/2018] [Indexed: 12/15/2022] Open
Abstract
Neuropathic pain is associated with persistent changes in gene expression in primary sensory neurons, but the underlying epigenetic mechanisms that cause these changes remain unclear. The muscarinic cholinergic receptors (mAChRs), particularly the M2 subtype (encoded by the cholinergic receptor muscarinic 2 (Chrm2) gene), are critically involved in the regulation of spinal nociceptive transmission. However, little is known about how Chrm2 expression is transcriptionally regulated. Here we show that nerve injury persistently increased the expression of RE1-silencing transcription factor (REST, also known as neuron-restrictive silencing factor [NRSF]), a gene-silencing transcription factor, in the dorsal root ganglion (DRG). Remarkably, nerve injury-induced chronic but not acute pain hypersensitivity was attenuated in mice with Rest knockout in DRG neurons. Also, siRNA-mediated Rest knockdown reversed nerve injury-induced chronic pain hypersensitivity in rats. Nerve injury persistently reduced Chrm2 expression in the DRG and diminished the analgesic effect of muscarine. The RE1 binding site on the Chrm2 promoter is required for REST-mediated Chrm2 repression, and nerve injury increased the enrichment of REST in the Chrm2 promoter in the DRG. Furthermore, Rest knockdown or genetic ablation in DRG neurons normalized Chrm2 expression and augmented muscarine's analgesic effect on neuropathic pain and fully reversed the nerve injury-induced reduction in the inhibitory effect of muscarine on glutamatergic input to spinal dorsal horn neurons. Our findings indicate that nerve injury-induced REST up-regulation in DRG neurons plays an important role in the acute-to-chronic pain transition and is essential for the transcriptional repression of Chrm2 in neuropathic pain.
Collapse
Affiliation(s)
- Jixiang Zhang
- From the Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Shao-Rui Chen
- From the Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hong Chen
- From the Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hui-Lin Pan
- From the Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
29
|
Hu Z, Wang L, Ma S, Kirisci L, Feng Z, Xue Y, Klunk WE, Kamboh MI, Sweet RA, Becker J, Lv Q, Lopez OL, Xie XQ. Synergism of antihypertensives and cholinesterase inhibitors in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:542-555. [PMID: 30386819 PMCID: PMC6205113 DOI: 10.1016/j.trci.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION We investigated the effect of antihypertensive (aHTN) medications and cholinesterase inhibitors (ChEIs) on the cognitive decline in patients with Alzheimer's disease (AD) and analyzed synergism by chemogenomics systems pharmacology mapping. METHODS We compared the effect of aHTN drugs on Mini-Mental State Examination scores in 617 AD patients with hypertension, and studied the synergistic effects. RESULTS The combination of diuretics, calcium channel blockers, and renin-angiotensin-aldosterone system blockers showed slower cognitive decline compared with other aHTN groups (Δβ = +1.46, P < .0001). aHTN medications slow down cognitive decline in ChEI users (Δβ = +0.56, P = .006), but not in non-ChEI users (Δβ = -0.31, P = .53). DISCUSSION aHTN and ChEI drugs showed synergistic effects. A combination of diuretics, renin-angiotensin-aldosterone system blockers, and calcium channel blockers had the slowest cognitive decline. The chemogenomics systems pharmacology-identified molecular targets provide system pharmacology interpretation of the synergism of the drugs in clinics. The results suggest that improving vascular health is essential for AD treatment and provide a novel direction for AD drug development.
Collapse
Affiliation(s)
- Ziheng Hu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lirong Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shifan Ma
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Levent Kirisci
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Xue
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Becker
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Jing M, Zhang P, Wang G, Feng J, Mesik L, Zeng J, Jiang H, Wang S, Looby JC, Guagliardo NA, Langma LW, Lu J, Zuo Y, Talmage DA, Role LW, Barrett PQ, Zhang LI, Luo M, Song Y, Zhu JJ, Li Y. A genetically encoded fluorescent acetylcholine indicator for in vitro and in vivo studies. Nat Biotechnol 2018; 36:726-737. [PMID: 29985477 PMCID: PMC6093211 DOI: 10.1038/nbt.4184] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023]
Abstract
The neurotransmitter acetylcholine (ACh) regulates a diverse array of physiological processes throughout the body. Despite its importance, cholinergic transmission in the majority of tissues and organs remains poorly understood owing primarily to the limitations of available ACh-monitoring techniques. We developed a family of ACh sensors (GACh) based on G-protein-coupled receptors that has the sensitivity, specificity, signal-to-noise ratio, kinetics and photostability suitable for monitoring ACh signals in vitro and in vivo. GACh sensors were validated with transfection, viral and/or transgenic expression in a dozen types of neuronal and non-neuronal cells prepared from multiple animal species. In all preparations, GACh sensors selectively responded to exogenous and/or endogenous ACh with robust fluorescence signals that were captured by epifluorescence, confocal, and/or two-photon microscopy. Moreover, analysis of endogenous ACh release revealed firing-pattern-dependent release and restricted volume transmission, resolving two long-standing questions about central cholinergic transmission. Thus, GACh sensors provide a user-friendly, broadly applicable tool for monitoring cholinergic transmission underlying diverse biological processes.
Collapse
Affiliation(s)
- Miao Jing
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871,
China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Peng Zhang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Guangfu Wang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
- Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin
150001, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871,
China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Lukas Mesik
- Zilkha Neurogenetic Institute, Department of Physiology & Neuroscience, Keck School of Medicine,
University of Southern California, Los Angeles, CA, 90033
| | - Jianzhi Zeng
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871,
China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Huoqing Jiang
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871,
China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Shaohua Wang
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794
| | - Jess C. Looby
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
- Undergraduate Class of 2019, University of Virginia College of Arts and Sciences, Charlottesville, VA
22908
| | - Nick A. Guagliardo
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Linda W. Langma
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Ju Lu
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Yi Zuo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA
95064
| | - David A. Talmage
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794
| | - Lorna W. Role
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794
| | - Paula Q. Barrett
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Department of Physiology & Neuroscience, Keck School of Medicine,
University of Southern California, Los Angeles, CA, 90033
| | - Minmin Luo
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yan Song
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - J. Julius Zhu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22908
- School of Medicine, Ningbo University, Ningbo, 315010, China
- Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, 6525 EN, Nijmegen,
Netherlands
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science
and Technology, Wuhan 430030, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871,
China
- PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| |
Collapse
|
31
|
Romberg C, Bartko S, Wess J, Saksida LM, Bussey TJ. Impaired object-location learning and recognition memory but enhanced sustained attention in M2 muscarinic receptor-deficient mice. Psychopharmacology (Berl) 2018; 235:3495-3508. [PMID: 30327842 PMCID: PMC6267149 DOI: 10.1007/s00213-018-5065-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/03/2018] [Indexed: 11/30/2022]
Abstract
RATIONALE Muscarinic acetylcholine receptors are known to play key roles in mediating cognitive processes, and impaired muscarinic cholinergic neurotransmission is associated with normal ageing processes and Alzheimer's disease. However, the specific contributions of the individual muscarinic receptor subtypes (M1-M5) to cognition are presently not well understood. OBJECTIVES The aim of this study was to investigate the contribution of M2-type muscarinic receptor signalling to sustained attention, executive control and learning and memory. METHODS M2 receptor-deficient (M2-/-) mice were tested on a touchscreen-operated task battery testing visual discrimination, behavioural flexibility, object-location associative learning, attention and response control. Spontaneous recognition memory for real-world objects was also assessed. RESULTS We found that M2-/- mice showed an enhancement of attentional performance, but significant deficits on some tests of learning and memory. Executive control and visual discrimination were unaffected by M2-depletion. CONCLUSIONS These findings suggest that M2 activation has heterogeneous effects across cognitive domains, and provide insights into how acetylcholine may support multiple specific cognitive processes through functionally distinct cholinergic receptor subtypes. They also suggest that therapeutics involving M2 receptor-active compounds should be assessed across a broad range of cognitive domains, as they may enhance some cognitive functions, but impair others.
Collapse
Affiliation(s)
- Carola Romberg
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK. .,Wellcome Trust and MRC Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB, UK. .,Department of Psychology, Research Unit Biological Psychology, Ludwig-Maximilians-University, Munich, Germany.
| | - Susan Bartko
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB UK
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Lisa M. Saksida
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB UK ,Wellcome Trust and MRC Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Department of Psychology, Research Unit Biological Psychology, Ludwig-Maximilians-University, Munich, Germany ,Brain and Mind Institute, Western University, London, ON Canada
| | - Timothy J. Bussey
- Department of Psychology, University of Cambridge, Cambridge, CB2 3EB UK ,Wellcome Trust and MRC Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, CB2 3EB UK ,Brain and Mind Institute, Western University, London, ON Canada ,Molecular Medicine Research Laboratories, Robarts Research Institute & Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON Canada
| |
Collapse
|
32
|
Pepeu G, Grazia Giovannini M. The fate of the brain cholinergic neurons in neurodegenerative diseases. Brain Res 2017; 1670:173-184. [PMID: 28652219 DOI: 10.1016/j.brainres.2017.06.023] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 01/03/2023]
Abstract
The aims of this review are: 1) to describe which cholinergic neurons are affected in brain neurodegenerative diseases leading to dementia; 2) to discuss the possible causes of the degeneration of the cholinergic neurons, 3) to summarize the functional consequences of the cholinergic deficit. The brain cholinergic system is basically constituted by three populations of phenotypically similar neurons forming a series of basal forebrain nuclei, the midpontine nuclei and a large population of striatal interneurons. In Alzheimer's disease there is an extensive loss of forebrain cholinergic neurons accompanied by a reduction of the cholinergic fiber network of the cortical mantel and hippocampus. The midpontine cholinergic nuclei are spared. The same situation occurs in the corticobasal syndrome and dementia following alcohol abuse and traumatic brain injury. Conversely, in Parkinson's disease, the midpontine nuclei degenerate, together with the dopaminergic nuclei, reducing the cholinergic input to thalamus and forebrain whereas the forebrain cholinergic neurons are spared. In Parkinson's disease with dementia, Lewis Body Dementia and Parkinsonian syndromes both groups of forebrain and midpontine cholinergic nuclei degenerate. In Huntington's disease a dysfunction of the striatal cholinergic interneurons without cell loss takes place. The formation and accumulation of misfolded proteins such as β-amyloid oligomers and plaques, tau protein tangles and α-synuclein clumps, and aggregated mutated huntingtin play a crucial role in the neuronal degeneration by direct cellular toxicity of the misfolded proteins and through the toxic compounds resulting from an extensive inflammatory reaction. Evidences indicate that β-amyloid disrupts NGF metabolism causing the degeneration of the cholinergic neurons which depend on NGF for their survival, namely the forebrain cholinergic neurons, sparing the midpontine and striatal neurons which express no specific NGF receptors. It is feasible that the latter cholinergic neurons may be damaged by direct toxicity of tau, α-synuclein and inflammations products through mechanisms not fully understood. Attention and learning and memory impairment are the functional consequences of the forebrain cholinergic neuron dysfunction, whereas the loss of midpontine cholinergic neurons results primarily in motor and sleep disturbances.
Collapse
Affiliation(s)
- Giancarlo Pepeu
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy.
| |
Collapse
|
33
|
Risher JF, Tucker P. Alkyl Mercury-Induced Toxicity: Multiple Mechanisms of Action. REVIEWS OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2017; 240:105-149. [PMID: 27161558 PMCID: PMC10508330 DOI: 10.1007/398_2016_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
There are a number of mechanisms by which alkylmercury compounds cause toxic action in the body. Collectively, published studies reveal that there are some similarities between the mechanisms of the toxic action of the mono-alkyl mercury compounds methylmercury (MeHg) and ethylmercury (EtHg). This paper represents a summary of some of the studies regarding these mechanisms of action in order to facilitate the understanding of the many varied effects of alkylmercurials in the human body. The similarities in mechanisms of toxicity for MeHg and EtHg are presented and compared. The difference in manifested toxicity of MeHg and EtHg are likely the result of the differences in exposure, metabolism, and elimination from the body, rather than differences in mechanisms of action between the two.
Collapse
Affiliation(s)
- John F Risher
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA.
| | - Pamela Tucker
- Division of Toxicology and Human Health Sciences, Agency for Toxic Substances and Disease Registry, 1600 Clifton Road (MS F-58), Atlanta, GA, 30333, USA
| |
Collapse
|
34
|
Bradley SJ, Bourgognon JM, Sanger HE, Verity N, Mogg AJ, White DJ, Butcher AJ, Moreno JA, Molloy C, Macedo-Hatch T, Edwards JM, Wess J, Pawlak R, Read DJ, Sexton PM, Broad LM, Steinert JR, Mallucci GR, Christopoulos A, Felder CC, Tobin AB. M1 muscarinic allosteric modulators slow prion neurodegeneration and restore memory loss. J Clin Invest 2016; 127:487-499. [PMID: 27991860 PMCID: PMC5272187 DOI: 10.1172/jci87526] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 11/03/2016] [Indexed: 11/17/2022] Open
Abstract
The current frontline symptomatic treatment for Alzheimer's disease (AD) is whole-body upregulation of cholinergic transmission via inhibition of acetylcholinesterase. This approach leads to profound dose-related adverse effects. An alternative strategy is to selectively target muscarinic acetylcholine receptors, particularly the M1 muscarinic acetylcholine receptor (M1 mAChR), which was previously shown to have procognitive activity. However, developing M1 mAChR-selective orthosteric ligands has proven challenging. Here, we have shown that mouse prion disease shows many of the hallmarks of human AD, including progressive terminal neurodegeneration and memory deficits due to a disruption of hippocampal cholinergic innervation. The fact that we also show that muscarinic signaling is maintained in both AD and mouse prion disease points to the latter as an excellent model for testing the efficacy of muscarinic pharmacological entities. The memory deficits we observed in mouse prion disease were completely restored by treatment with benzyl quinolone carboxylic acid (BQCA) and benzoquinazoline-12 (BQZ-12), two highly selective positive allosteric modulators (PAMs) of M1 mAChRs. Furthermore, prolonged exposure to BQCA markedly extended the lifespan of diseased mice. Thus, enhancing hippocampal muscarinic signaling using M1 mAChR PAMs restored memory loss and slowed the progression of mouse prion disease, indicating that this ligand type may have clinical benefit in diseases showing defective cholinergic transmission, such as AD.
Collapse
Affiliation(s)
- Sophie J. Bradley
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | - Helen E. Sanger
- Eli Lilly and Co., Neuroscience, Windlesham, Surrey, United Kingdom
| | - Nicholas Verity
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Adrian J. Mogg
- Eli Lilly and Co., Neuroscience, Windlesham, Surrey, United Kingdom
| | - David J. White
- Central Research Facility, University of Leicester, Leicester, United Kingdom
| | - Adrian J. Butcher
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Julie A. Moreno
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Colin Molloy
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | - Jurgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Robert Pawlak
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - David J. Read
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Patrick M. Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Lisa M. Broad
- Eli Lilly and Co., Neuroscience, Windlesham, Surrey, United Kingdom
| | - Joern R. Steinert
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
| | - Giovanna R. Mallucci
- MRC Toxicology Unit, University of Leicester, Leicester, United Kingdom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Christian C. Felder
- Eli Lilly and Co., Neuroscience, Lilly Corporate Center, Indianapolis, Indiana, USA
| | - Andrew B. Tobin
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
35
|
Gao J, Midde N, Zhu J, Terry AV, McInnes C, Chapman JM. Synthesis and biological evaluation of ranitidine analogs as multiple-target-directed cognitive enhancers for the treatment of Alzheimer's disease. Bioorg Med Chem Lett 2016; 26:5573-5579. [PMID: 27769620 PMCID: PMC5185470 DOI: 10.1016/j.bmcl.2016.09.072] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/02/2016] [Accepted: 09/28/2016] [Indexed: 11/17/2022]
Abstract
Using molecular modeling and rationally designed structural modifications, the multi-target structure-activity relationship for a series of ranitidine analogs has been investigated. Incorporation of a variety of isosteric groups indicated that appropriate aromatic moieties provide optimal interactions with the hydrophobic and π-π interactions with the peripheral anionic site of the AChE active site. The SAR of a series of cyclic imides demonstrated that AChE inhibition is increased by additional aromatic rings, where 1,8-naphthalimide derivatives were the most potent analogs and other key determinants were revealed. In addition to improving AChE activity and chemical stability, structural modifications allowed determination of binding affinities and selectivities for M1-M4 receptors and butyrylcholinesterase (BuChE). These results as a whole indicate that the 4-nitropyridazine moiety of the JWS-USC-75IX parent ranitidine compound (JWS) can be replaced with other chemotypes while retaining effective AChE inhibition. These studies allowed investigation into multitargeted binding to key receptors and warrant further investigation into 1,8-naphthalimide ranitidine derivatives for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Jie Gao
- Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA; Department of Pharmacology and Toxicology, Augusta University, Health Sciences Campus, CB-3530, 1459 Laney Walker Blvd., Augusta, GA 30912, USA
| | - Narasimha Midde
- Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Jun Zhu
- Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Augusta University, Health Sciences Campus, CB-3530, 1459 Laney Walker Blvd., Augusta, GA 30912, USA
| | - Campbell McInnes
- Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.
| | - James M Chapman
- Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
36
|
Lee JH, Francis PT, Ballard CG, Aarsland D, Kalaria RN, Wong PTH, Chen CP, Lai MK. Muscarinic M1 Receptor Coupling to G-protein is Intact in Parkinson’s Disease Dementia. JOURNAL OF PARKINSONS DISEASE 2016; 6:733-739. [DOI: 10.3233/jpd-160932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Jasinda H. Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Paul T. Francis
- Wolfson Centre for Age-Related Diseases, King’s College London, London, UK
| | - Clive G. Ballard
- Wolfson Centre for Age-Related Diseases, King’s College London, London, UK
| | - Dag Aarsland
- Department Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Raj N. Kalaria
- Institute for Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Peter T.-H. Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Memory, Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| | - Mitchell K.P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Kent Ridge, Singapore
- Wolfson Centre for Age-Related Diseases, King’s College London, London, UK
- Memory, Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| |
Collapse
|
37
|
Higher levels of different muscarinic receptors in the cortex and hippocampus from subjects with Alzheimer’s disease. J Neural Transm (Vienna) 2016; 124:273-284. [DOI: 10.1007/s00702-016-1625-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/15/2016] [Indexed: 12/27/2022]
|
38
|
Guerram M, Zhang LY, Jiang ZZ. G-protein coupled receptors as therapeutic targets for neurodegenerative and cerebrovascular diseases. Neurochem Int 2016; 101:1-14. [PMID: 27620813 DOI: 10.1016/j.neuint.2016.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 12/24/2022]
Abstract
Neurodegenerative and cerebrovascular diseases are frequent in elderly populations and comprise primarily of dementia (mainly Alzheimer's disease) Parkinson's disease and stroke. These neurological disorders (NDs) occur as a result of neurodegenerative processes and represent one of the most frequent causes of death and disability worldwide with a significant clinical and socio-economic impact. Although NDs have been characterized for many years, the exact molecular mechanisms that govern these pathologies or why they target specific individuals and specific neuronal populations remain unclear. As research progresses, many similarities appear which relate these diseases to one another on a subcellular level. Discovering these similarities offers hope for therapeutic advances that could ameliorate the conditions of many diseases simultaneously. G-protein coupled receptors (GPCRs) are the most abundant receptor type in the central nervous system and are linked to complex downstream pathways, manipulation of which may have therapeutic application in many NDs. This review will highlight the potential use of neurotransmitter GPCRs as emerging therapeutic targets for neurodegenerative and cerebrovascular diseases.
Collapse
Affiliation(s)
- Mounia Guerram
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Faculty of Exact Sciences and Nature and Life Sciences, Department of Biology, Larbi Ben M'hidi University, Oum El Bouaghi 04000, Algeria
| | - Lu-Yong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen-Zhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
39
|
Imaging α4β2 Nicotinic Acetylcholine Receptors (nAChRs) in Baboons with [18F]XTRA, a Radioligand with Improved Specific Binding in Extra-Thalamic Regions. Mol Imaging Biol 2016; 19:280-288. [DOI: 10.1007/s11307-016-0999-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
40
|
Chauhan PS, Misra UK, Kalita J, Chandravanshi LP, Khanna VK. Memory and learning seems to be related to cholinergic dysfunction in the JE rat model. Physiol Behav 2016; 156:148-55. [DOI: 10.1016/j.physbeh.2016.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 10/12/2015] [Accepted: 01/09/2016] [Indexed: 10/22/2022]
|
41
|
Newman LA, Gold PE. Attenuation in rats of impairments of memory by scopolamine, a muscarinic receptor antagonist, by mecamylamine, a nicotinic receptor antagonist. Psychopharmacology (Berl) 2016; 233:925-32. [PMID: 26660295 PMCID: PMC4752895 DOI: 10.1007/s00213-015-4174-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/23/2015] [Indexed: 01/08/2023]
Abstract
RATIONALE Scopolamine, a muscarinic antagonist, impairs learning and memory for many tasks, supporting an important role for the cholinergic system in these cognitive functions. The findings are most often interpreted to indicate that a decrease in postsynaptic muscarinic receptor activation mediates the memory impairments. However, scopolamine also results in increased release of acetylcholine in the brain as a result of blocking presynaptic muscarinic receptors. OBJECTIVES The present experiments assess whether scopolamine-induced increases in acetylcholine release may impair memory by overstimulating postsynaptic cholinergic nicotinic receptors, i.e., by reaching the high end of a nicotinic receptor activation inverted-U dose-response function. RESULTS Rats tested in a spontaneous alternation task showed dose-dependent working memory deficits with systemic injections of mecamylamine and scopolamine. When an amnestic dose of scopolamine (0.15 mg/kg) was co-administered with a subamnestic dose of mecamylamine (0.25 mg/kg), this dose of mecamylamine significantly attenuated the scopolamine-induced memory impairments. We next assessed the levels of acetylcholine release in the hippocampus in the presence of scopolamine and mecamylamine. Mecamylamine injections resulted in decreased release of acetylcholine, while scopolamine administration caused a large increase in acetylcholine release. CONCLUSIONS These findings indicate that a nicotinic antagonist can attenuate impairments in memory produced by a muscarinic antagonist. The nicotinic antagonist may block excessive activation of nicotinic receptors postsynaptically or attenuate increases in acetylcholine release presynaptically. Either effect of a nicotinic antagonist-to decrease scopolamine-induced increases in acetylcholine output or to decrease postsynaptic acetylcholine receptor activation-may mediate the negative effects on memory of muscarinic antagonists.
Collapse
Affiliation(s)
- L A Newman
- Department of Biology, Syracuse University, 114 Life Sciences Complex, Syracuse, NY, 13244, USA
| | - P E Gold
- Department of Biology, Syracuse University, 114 Life Sciences Complex, Syracuse, NY, 13244, USA.
| |
Collapse
|
42
|
Kandimalla R, Vallamkondu J, Corgiat EB, Gill KD. Understanding Aspects of Aluminum Exposure in Alzheimer's Disease Development. Brain Pathol 2016; 26:139-54. [PMID: 26494454 PMCID: PMC8028870 DOI: 10.1111/bpa.12333] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/12/2015] [Indexed: 01/21/2023] Open
Abstract
Aluminum is a ubiquitously abundant nonessential element. Aluminum has been associated with neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis, and dialysis encephalopathy. Many continue to regard aluminum as controversial although increasing evidence supports the implications of aluminum in the pathogenesis of AD. Aluminum causes the accumulation of tau protein and Aβ protein in the brain of experimental animals. Aluminum induces neuronal apoptosis in vivo and in vitro, either by endoplasmic stress from the unfolded protein response, by mitochondrial dysfunction, or a combination of them. Some, people who are exposed chronically to aluminum, either from through water and/or food, have not shown any AD pathology, apparently because their gastrointestinal barrier is more effective. This article is written keeping in mind mechanisms of action of aluminum neurotoxicity with respect to AD.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Department of BiochemistryPost Graduate Institute of Medical Education and ResearchChandigarhIndia
- Radiation Oncology, Emory UniversityAtlantaGA
| | | | - Edwin B Corgiat
- Genetics and Molecular Biology ProgramEmory University Laney Graduate SchoolAtlantaGA
| | - Kiran Dip Gill
- Department of BiochemistryPost Graduate Institute of Medical Education and ResearchChandigarhIndia
| |
Collapse
|
43
|
Insight into the Molecular Imaging of Alzheimer's Disease. Int J Biomed Imaging 2016; 2016:7462014. [PMID: 26880871 PMCID: PMC4736963 DOI: 10.1155/2016/7462014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease is a complex neurodegenerative disease affecting millions of individuals worldwide. Earlier it was diagnosed only via clinical assessments and confirmed by postmortem brain histopathology. The development of validated biomarkers for Alzheimer's disease has given impetus to improve diagnostics and accelerate the development of new therapies. Functional imaging like positron emission tomography (PET), single photon emission computed tomography (SPECT), functional magnetic resonance imaging (fMRI), and proton magnetic resonance spectroscopy provides a means of detecting and characterising the regional changes in brain blood flow, metabolism, and receptor binding sites that are associated with Alzheimer's disease. Multimodal neuroimaging techniques have indicated changes in brain structure and metabolic activity, and an array of neurochemical variations that are associated with neurodegenerative diseases. Radiotracer-based PET and SPECT potentially provide sensitive, accurate methods for the early detection of disease. This paper presents a review of neuroimaging modalities like PET, SPECT, and selected imaging biomarkers/tracers used for the early diagnosis of AD. Neuroimaging with such biomarkers and tracers could achieve a much higher diagnostic accuracy for AD and related disorders in the future.
Collapse
|
44
|
Hirao K, Pontone GM, Smith GS. Molecular imaging of neuropsychiatric symptoms in Alzheimer's and Parkinson's disease. Neurosci Biobehav Rev 2015; 49:157-70. [PMID: 25446948 PMCID: PMC4806385 DOI: 10.1016/j.neubiorev.2014.11.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 10/27/2014] [Accepted: 11/12/2014] [Indexed: 01/14/2023]
Abstract
Neuropsychiatric symptoms (NPS) are very common in neurodegenerative diseases and are a major contributor to disability and caregiver burden. There is accumulating evidence that NPS may be a prodrome and/or a "risk factor" of neurodegenerative diseases. The medications used to treat these symptoms in younger patients are not very effective in patients with neurodegenerative disease and may have serious side effects. An understanding of the neurobiology of NPS is critical for the development of more effective intervention strategies. Targeting these symptoms may also have implications for prevention of cognitive or motor decline. Molecular brain imaging represents a bridge between basic and clinical observations and provides many opportunities for translation from animal models and human post-mortem studies to in vivo human studies. Molecular brain imaging studies in Alzheimer's disease (AD) and Parkinson's disease (PD) are reviewed with a primary focus on positron emission tomography studies of NPS. Future directions for the field of molecular imaging in AD and PD to understand the neurobiology of NPS will be discussed.
Collapse
Affiliation(s)
- Kentaro Hirao
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA; Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Gregory M Pontone
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA
| | - Gwenn S Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, USA.
| |
Collapse
|
45
|
Xuan AG, Pan XB, Wei P, Ji WD, Zhang WJ, Liu JH, Hong LP, Chen WL, Long DH. Valproic acid alleviates memory deficits and attenuates amyloid-β deposition in transgenic mouse model of Alzheimer's disease. Mol Neurobiol 2014; 51:300-12. [PMID: 24854198 DOI: 10.1007/s12035-014-8751-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 05/13/2014] [Indexed: 12/12/2022]
Abstract
In the brains of patients with Alzheimer's disease (AD) and transgenic AD mouse models, astrocytes and microglia activated by amyloid-β (Aβ) contribute to the inflammatory process that develops around injury in the brain. Valproic acid (VPA) has been shown to have anti-inflammatory function. The present study intended to explore the therapeutic effect of VPA on the neuropathology and memory deficits in APPswe/PS1ΔE9 (APP/PS1) transgenic mice. Here, we report that VPA-treated APP/PS1 mice markedly improved memory deficits and decreased Aβ deposition compared with the vehicle-treated APP/PS1 mice. Moreover, the extensive astrogliosis and microgliosis as well as the increased expression in interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in the hippocampus and cortex of APP/PS1 transgenic mice were significantly reduced following administration of VPA, which attenuated neuronal degeneration. Concomitantly, VPA alleviated the levels of p65 NF-κB phosphorylation and enhanced the levels of acetyl-H3, Bcl-2, and phospho-glycogen synthase kinase (GSK)-3β that occurred in the hippocampus of APP/PS1 transgenic mice. These results demonstrate that VPA could significantly ameliorate spatial memory impairment and Aβ deposition at least in part via the inhibition of inflammation, suggesting that administration of VPA could provide a therapeutic approach for AD.
Collapse
Affiliation(s)
- Ai-Guo Xuan
- Department of Anatomy, Guangzhou Medical University, Guangzhou, 510182, Guangdong, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vermeiren Y, Van Dam D, De Deyn PP. Psychiatric Disorders in Dementia. PET AND SPECT IN PSYCHIATRY 2014:271-324. [DOI: 10.1007/978-3-642-40384-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
47
|
Mesulam MM. Cholinergic circuitry of the human nucleus basalis and its fate in Alzheimer's disease. J Comp Neurol 2013; 521:4124-44. [PMID: 23852922 PMCID: PMC4175400 DOI: 10.1002/cne.23415] [Citation(s) in RCA: 256] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/11/2013] [Accepted: 06/28/2013] [Indexed: 01/15/2023]
Abstract
The nucleus basalis is located at the confluence of the limbic and reticular activating systems. It receives dopaminergic input from the ventral tegmental area/substantia nigra, serotonergic input from the raphe nuclei, and noradrenergic input from the nucleus locus coeruleus. Its cholinergic contingent, known as Ch4, provides the principal source of acetylcholine for the cerebral cortex and amygdala. More than half of presynaptic varicosities along its cholinergic axons make traditional synaptic contacts with cortical neurons. Limbic and paralimbic cortices of the brain receive the heaviest cholinergic input from Ch4 and are also the principal sources of reciprocal cortical projections back to the nucleus basalis. This limbic affiliation explains the role of the nucleus basalis in modulating the impact and memorability of incoming sensory information. The anatomical continuity of the nucleus basalis with other basomedial limbic structures may underlie its early and high vulnerability to the tauopathy and neurofibrillary degeneration of Alzheimer's disease. The tauopathy in Ch4 eventually leads to the degeneration of the cholinergic axons that it sends to the cerebral cortex. The early involvement of Ch4 has a magnifying effect on Alzheimer's pathology, because neurofibrillary degeneration in a small number of neurons can perturb neurotransmission in all cortical areas. Although the exact contribution of the Ch4 lesion to the cognitive changes of Alzheimer's disease remains poorly understood, the cholinergic circuitry of the nucleus basalis is emerging as one of the most strategically positioned and behaviorally consequential modulatory systems of the human cerebral cortex. J. Comp. Neurol. 521:4124-4144, 2013. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M.-Marsel Mesulam
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University Medical School, Chicago, Illinois 60611
| |
Collapse
|
48
|
Holland JP, Liang SH, Rotstein BH, Collier TL, Stephenson NA, Greguric I, Vasdev N. Alternative approaches for PET radiotracer development in Alzheimer's disease: imaging beyond plaque. J Labelled Comp Radiopharm 2013; 57:323-31. [PMID: 24327420 DOI: 10.1002/jlcr.3158] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/29/2013] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) and related dementias show increasing clinical prevalence, yet our understanding of the etiology and pathobiology of disease-related neurodegeneration remains limited. In this regard, noninvasive imaging with radiotracers for positron emission tomography (PET) presents a unique tool for quantifying spatial and temporal changes in characteristic biological markers of brain disease and for assessing potential drug efficacy. PET radiotracers targeting different protein markers are being developed to address questions pertaining to the molecular and/or genetic heterogeneity of AD and related dementias. For example, radiotracers including [(11) C]-PiB and [(18) F]-AV-45 (Florbetapir) are being used to measure the density of Aβ-plaques in AD patients and to interrogate the biological mechanisms of disease initiation and progression. Our focus is on the development of novel PET imaging agents, targeting proteins beyond Aβ-plaques, which can be used to investigate the broader mechanism of AD pathogenesis. Here, we present the chemical basis of various radiotracers which show promise in preclinical or clinical studies for use in evaluating the phenotypic or biochemical characteristics of AD. Radiotracers for PET imaging neuroinflammation, metal ion association with Aβ-plaques, tau protein, cholinergic and cannabinoid receptors, and enzymes including glycogen-synthase kinase-3β and monoamine oxidase B amongst others, and their connection to AD are highlighted.
Collapse
Affiliation(s)
- Jason P Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Department of Radiology, Harvard Medical School, 55 Fruit St., White 427, Boston, Massachusetts, 02114, USA; Life Sciences, Australian Nuclear Science and Technology Organisation, Kirrawee, New South Wales, 2232, Australia
| | | | | | | | | | | | | |
Collapse
|
49
|
Laursen B, Mørk A, Plath N, Kristiansen U, Bastlund JF. Impaired hippocampal acetylcholine release parallels spatial memory deficits in Tg2576 mice subjected to basal forebrain cholinergic degeneration. Brain Res 2013; 1543:253-62. [PMID: 24231553 DOI: 10.1016/j.brainres.2013.10.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 01/22/2023]
Abstract
The Alzheimer's disease (AD) mouse model Tg2576 overexpresses an AD associated mutant variant of human APP and accumulates amyloid beta (Aβ) in an age-dependent manner. Using the selective cholinergic immunotoxin mu p75-saporin (SAP), we induced a partial basal forebrain cholinergic degeneration (BFCD) in 3 months old male Tg2576 mice to co-express cholinergic degeneration with Aβ overexpression as these characteristics constitutes key hallmarks of AD. At 9 months, SAP lesioned Tg2576 mice were cognitively impaired in two spatial paradigms addressing working memory and mid to long-term memory. Conversely, there was no deterioration of cognitive functioning in sham lesioned Tg2576 mice or wild type littermates (wt) receiving the immunotoxin. At 10 months of age, release of acetylcholine (ACh) was addressed by microdialysis in conscious mice. Scopolamine-induced increases in hippocampal ACh efflux was significantly reduced in SAP lesioned Tg2576 mice compared to sham lesioned Tg2576 mice. Intriguingly, there was no significant difference in ACh efflux between wt treatment groups. Following SAP treatment, choline acetyltransferase activity was reduced in the hippocampus and frontal cortex and the reduction was comparable between groups. Our results suggest that partial BFCD acts collectively with increased levels of Aβ to induce cognitive decline and to compromise cholinergic release. Tg2576 mice with BFCD may constitute a new and suitable AD mouse model to study the interrelations between cholinergic deficits and amsyloid deposition.
Collapse
Affiliation(s)
- Bettina Laursen
- H. Lundbeck A/S, Synaptic Transmission 1, Ottiliavej 9, 2500 Valby, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Arne Mørk
- H. Lundbeck A/S, Synaptic Transmission 1, Ottiliavej 9, 2500 Valby, Denmark
| | - Niels Plath
- H. Lundbeck A/S, Synaptic Transmission 1, Ottiliavej 9, 2500 Valby, Denmark
| | - Uffe Kristiansen
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | | |
Collapse
|
50
|
Boulos J, Jakubik J, Randakova A, Avila C. Synthesis ofN-Substituted Piperidine Salts as Potential Muscarinic Ligands for Alzheimer's Applications. J Heterocycl Chem 2013. [DOI: 10.1002/jhet.1742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- John Boulos
- Department of Physical Sciences; Barry University; Miami Shores Florida 33161 USA
| | - Jan Jakubik
- Department of Neurochemistry; Institute of Physiology of the Academy of Sciences of the Czech Republic; v.v.i., 142 20 Prague Czech Republic
| | - Alena Randakova
- Department of Neurochemistry; Institute of Physiology of the Academy of Sciences of the Czech Republic; v.v.i., 142 20 Prague Czech Republic
| | - Cristina Avila
- Department of Physical Sciences; Barry University; Miami Shores Florida 33161 USA
| |
Collapse
|