1
|
Ongnok B, Prathumsap N, Chunchai T, Pantiya P, Arunsak B, Chattipakorn N, Chattipakorn SC. Nicotinic and Muscarinic Acetylcholine Receptor Agonists Counteract Cognitive Impairment in a Rat Model of Doxorubicin-Induced Chemobrain via Attenuation of Multiple Programmed Cell Death Pathways. Mol Neurobiol 2024; 61:8831-8850. [PMID: 38568417 DOI: 10.1007/s12035-024-04145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 03/21/2024] [Indexed: 10/23/2024]
Abstract
Chemotherapy causes undesirable long-term neurological sequelae, chemotherapy-induced cognitive impairment (CICI), or chemobrain in cancer survivors. Activation of programmed cell death (PCD) has been proposed to implicate in the development and progression of chemobrain. Neuronal apoptosis has been extensively recognized in experimental models of chemobrain, but little is known about alternative forms of PCD in response to chemotherapy. Activation of acetylcholine receptors (AChRs) is emerging as a promising target in attenuating a wide variety of the neuronal death associated with neurodegeneration. Thus, this study aimed to investigate the therapeutic capacity of AChR agonists on cognitive function and molecular hallmarks of multiple PCD against chemotherapy neurotoxicity. To establish the chemobrain model, male Wistar rats were assigned to receive six doses of doxorubicin (DOX: 3 mg/kg) via intraperitoneal injection. The DOX-treated rats received either an a7nAChR agonist (PNU-282987: 3 mg/kg/day), mAChR agonists (bethanechol: 12 mg/kg/day), or the two as a combined treatment. DOX administration led to impaired cognitive function via neuroinflammation, glial activation, reduced synaptic/blood-brain barrier integrity, defective mitochondrial ROS-detoxifying capacity, and dynamic imbalance. DOX insult also mediated hyperphosphorylation of Tau and simultaneously induced various PCD, including apoptosis, necroptosis, and pyroptosis in the hippocampus. Concomitant treatment with either PNU-282987, bethanechol, or a combination of the two potently attenuated neuroinflammation, mitochondrial dyshomeostasis, and Tau hyperphosphorylation, thereby suppressing excessive apoptosis, necroptosis, and pyroptosis and improving cognitive function in DOX-treated rats. Our findings suggest that activation of AChRs using their agonists effectively protected against DOX-induced neuronal death and chemobrain.
Collapse
Affiliation(s)
- Benjamin Ongnok
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Nanthip Prathumsap
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Titikorn Chunchai
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Patcharapong Pantiya
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Busarin Arunsak
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand.
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Mueang Chiang Mai, 50200, Chiang Mai, Thailand.
| |
Collapse
|
2
|
Yohn SE, Harvey PD, Brannan SK, Horan WP. The potential of muscarinic M 1 and M 4 receptor activators for the treatment of cognitive impairment associated with schizophrenia. Front Psychiatry 2024; 15:1421554. [PMID: 39483736 PMCID: PMC11525114 DOI: 10.3389/fpsyt.2024.1421554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cognitive impairment is a core symptom of schizophrenia and a major determinant of poor long-term functional outcomes. Despite considerable efforts, we do not yet have any approved pharmacological treatments for cognitive impairment associated with schizophrenia (CIAS). A combination of advances in pre-clinical research and recent clinical trial findings have led to a resurgence of interest in the cognition-enhancing potential of novel muscarinic acetylcholine receptor (mAChR) agonists in schizophrenia. This article provides an overview of the scientific rationale for targeting M1 and M4 mAChRs. We describe the evolution of neuroscience research on these receptors since early drug discovery efforts focused on the mAChR agonist xanomeline. This work has revealed that M1 and M4 mAChRs are highly expressed in brain regions that are implicated in cognition. The functional significance of M1 and M4 mAChRs has been extensively characterized in animal models via use of selective receptor subtype compounds through neuronal and non-neuronal mechanisms. Recent clinical trials of a dual M1/M4 mAChR agonist show promising, replicable evidence of potential pro-cognitive effects in schizophrenia, with several other mAChR agonists in clinical development.
Collapse
Affiliation(s)
| | - Phillip D. Harvey
- Division of Psychology, University of Miami, Miami, FL, United States
| | | | - William P. Horan
- Bristol Myers Squibb, Princeton, NJ, United States
- Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
3
|
Belov V, Guehl NJ, Duvvuri S, Iredale P, Moon SH, Dhaynaut M, Chakilam S, MacDonagh AC, Rice PA, Yokell DL, Renger JJ, El Fakhri G, Normandin MD. PET imaging of M4 muscarinic acetylcholine receptors in rhesus macaques using [ 11C]MK-6884: Quantification with kinetic modeling and receptor occupancy by CVL-231 (emraclidine), a novel positive allosteric modulator. J Cereb Blood Flow Metab 2024; 44:1329-1342. [PMID: 38477292 PMCID: PMC11342722 DOI: 10.1177/0271678x241238820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024]
Abstract
Stimulation of the M4 muscarinic acetylcholine receptor reduces striatal hyperdopaminergia, suggesting its potential as a therapeutic target for schizophrenia. Emraclidine (CVL-231) is a novel, highly selective, positive allosteric modulator (PAM) of M4 muscarinic acetylcholine receptors i.e. acts as a modulator that increases the response of these receptors. First, we aimed to further characterize the positron emission tomography (PET) imaging and quantification performance of a recently developed M4 PAM radiotracer, [11C]MK-6884, in non-human primates (NHPs). Second, we applied these results to determine the receptor occupancy of CVL-231 as a function of dose. Using paired baseline-blocking PET scans, we quantified total volume of distribution, binding potential, and receptor occupancy. Both blood-based and reference region-based methods quantified M4 receptor levels across brain regions. The 2-tissue 4-parameter kinetic model best fitted regional [11C]MK-6884-time activity curves. Only the caudate nucleus and putamen displayed statistically significant [11C]MK-6884 uptake and dose-dependent blocking by CVL-231. For binding potential and receptor occupancy quantification, the simplified reference tissue model using the grey cerebellum as a reference region was employed. CVL-231 demonstrated dose-dependent M4 receptor occupancy in the striatum of the NHP brain and shows promise for further development in clinical trials.
Collapse
Affiliation(s)
- Vasily Belov
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicolas J Guehl
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Sung-Hyun Moon
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Alexander C MacDonagh
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter A Rice
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel L Yokell
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Monaco M, Trebesova H, Grilli M. Muscarinic Receptors and Alzheimer's Disease: New Perspectives and Mechanisms. Curr Issues Mol Biol 2024; 46:6820-6835. [PMID: 39057049 PMCID: PMC11276210 DOI: 10.3390/cimb46070407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases on a global scale. Historically, this pathology has been linked to cholinergic transmission, and despite the scarcity of effective therapies, numerous alternative processes and targets have been proposed as potential avenues for comprehending this complex illness. Nevertheless, the fundamental pathophysiological mechanisms underpinning AD remain largely enigmatic, with a growing body of evidence advocating for the significance of muscarinic receptors in modulating the brain's capacity to adapt and generate new memories. This review summarizes the current state of the art in the field of muscarinic receptors' involvement in AD. A specific key factor was the relationship between comorbidity and the emergence of new mechanisms.
Collapse
Affiliation(s)
- Martina Monaco
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (H.T.)
| | - Hanna Trebesova
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (H.T.)
| | - Massimo Grilli
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genoa, Italy; (M.M.); (H.T.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 16148 Genoa, Italy
| |
Collapse
|
5
|
Tsolias A, Zhou Y, Mojica CA, Sakharkar M, Tsolias MZ, Moore TL, Rosene DL, Medalla M. Neuroanatomical Substrates of Circuit-Specific Cholinergic Modulation across the Primate Anterior Cingulate Cortex. J Neurosci 2024; 44:e0953232024. [PMID: 38719447 PMCID: PMC11170673 DOI: 10.1523/jneurosci.0953-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024] Open
Abstract
Acetylcholine is a robust neuromodulator of the limbic system and a critical regulator of arousal and emotions. The anterior cingulate cortex (ACC) and the amygdala (AMY) are key limbic structures that are both densely innervated by cholinergic afferents and interact with each other for emotional regulation. The ACC is composed of functionally distinct dorsal (A24), rostral (A32), and ventral (A25) areas that differ in their connections with the AMY. The structural substrates of cholinergic modulation of distinct ACC microcircuits and outputs to AMY are thought to depend on the laminar and subcellular localization of cholinergic receptors. The present study examines the distribution of muscarinic acetylcholine receptors, m1 and m2, on distinct excitatory and inhibitory neurons and on AMY-targeting projection neurons within ACC areas, via immunohistochemistry and injections of neural tracers into the basolateral AMY in adult rhesus monkeys of both sexes. We found that laminar densities of m1+ and m2+ expressing excitatory and inhibitory neurons depended on area and cell type. Among the ACC areas, ventral subgenual ACC A25 exhibited greater m2+ localization on presynaptic inhibitory axon terminals and greater density of m1+ and m2+ expressing AMY-targeting (tracer+) pyramidal neurons. These patterns suggest robust cholinergic disinhibition and potentiation of amygdalar outputs from the limbic ventral ACC, which may be linked to the hyperexcitability of this subgenual ACC area in depression. These findings reveal the anatomical substrate of diverse cholinergic modulation of specific ACC microcircuits and amygdalar outputs that mediate cognitive-emotional integration and dysfunctions underlying stress and affective disorders.
Collapse
Affiliation(s)
- Alexandra Tsolias
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
| | - Yuxin Zhou
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
| | - Chromewell A Mojica
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
| | - Mitali Sakharkar
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
| | - Marianna Z Tsolias
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
| | - Tara L Moore
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts 02215
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts 02215
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts 02118
- Center for Systems Neuroscience, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
6
|
Duko B, Bedaso A, Dachew BA, Newnham E, Gebremedhin AT, Tessema G, Einarsdottir K, Alati R, Pereira G. The effect of maternal prenatal tobacco smoking on offspring academic achievement: A systematic review and meta-analysis. Addict Behav 2024; 153:107985. [PMID: 38367506 DOI: 10.1016/j.addbeh.2024.107985] [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: 08/25/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Previous epidemiological studies examining the prospective association between maternal prenatal tobacco smoking and offspring academic achievement have reported conflicting results. Therefore, this systematic review and meta-analysis was conducted to examine the magnitude and consistency of association reported by those studies. METHODS This systematic review and meta-analysis was guided by the PRISMA protocol. Relevant epidemiological studies on the topic were extracted from four main databases (PubMed/Medline, Embase, PsycINFO, and Scopus). The Newcastle-Ottawa Scale (NOS) was used to appraise the methodological quality of the included studies. We conducted a narrative assessment of the studies that did not report effect estimates. Inverse variance-weighted random effect meta-analysis was used to combine studies reporting effect sizes to estimate pooled adjusted odds ratio with 95% confidence intervals (95% CI). The review was prospectively registered in PROSPERO (CRD42022350901). RESULTS Nineteen observational studies, published between 1973 and 2021 with a total of 1.25 million study participants were included in the final review. Of these, fifteen studies (79 %) reported reduced academic achievement in offspring exposed to maternal prenatal tobacco smoking. The eight primary studies (sample size = 723,877) included in the meta-analysis together suggested a 49 % higher risk of reduced academic achievement in offspring exposed to maternal prenatal tobacco smoking when compared to non-exposed offspring (Pooled odds ratio = 1.49, 95 % CI:1.17-1.91). CONCLUSION Our review found a positive association between maternal prenatal tobacco smoking and offspring reduced academic achievement. However, variation in the adjustment of potential confounders and significant heterogeneity across included studies limited more conclusive inference. Mechanistic studies to identify causal pathways and specific academic impacts are needed to inform targeted developmental programs to assist child learning and academic performance.
Collapse
Affiliation(s)
- Bereket Duko
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; Australian Centre for Precision Health, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia.
| | - Asres Bedaso
- The Daffodil Centre, The University of Sydney, a joint venture with Cancer Council, New South Wales, Australia
| | - Berihun Assefa Dachew
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia
| | - Elizabeth Newnham
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, WA 6102, Australia; FXB Center for Health and Human Rights, Harvard University, Boston, MA, USA
| | | | - Gizachew Tessema
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, WA 6102, Australia
| | - Kristjana Einarsdottir
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; School of Health Sciences, University of Iceland, Vatnsmýrarvegur 16, 101 Reykjavík, Iceland
| | - Rosa Alati
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, WA 6102, Australia; Institute for Social Sciences Research, The University of Queensland, 80 Meier's Rd, Indooroopilly, Qld 4068 Australia
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Kent Street, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, WA 6102, Australia; Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
7
|
Satchell M, Fry B, Noureddine Z, Simmons A, Ognjanovski NN, Aton SJ, Zochowski MR. Neuromodulation via muscarinic acetylcholine pathway can facilitate distinct, complementary, and sequential roles for NREM and REM states during sleep-dependent memory consolidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.19.541465. [PMID: 38293183 PMCID: PMC10827095 DOI: 10.1101/2023.05.19.541465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Across vertebrate species, sleep consists of repeating cycles of NREM followed by REM. However, the respective functions of NREM, REM, and their stereotypic cycling pattern are not well understood. Using a simplified biophysical network model, we show that NREM and REM sleep can play differential and critical roles in memory consolidation primarily regulated, based on state-specific changes in cholinergic signaling. Within this network, decreasing and increasing muscarinic acetylcholine (ACh) signaling during bouts of NREM and REM, respectively, differentially alters neuronal excitability and excitatory/inhibitory balance. During NREM, deactivation of inhibitory neurons leads to network-wide disinhibition and bursts of synchronized activity led by firing in engram neurons. These features strengthen connections from the original engram neurons to less-active network neurons. In contrast, during REM, an increase in network inhibition suppresses firing in all but the most-active excitatory neurons, leading to competitive strengthening/pruning of the memory trace. We tested the predictions of the model against in vivo recordings from mouse hippocampus during active sleep-dependent memory storage. Consistent with modeling results, we find that functional connectivity between CA1 neurons changes differentially at transition from NREM to REM sleep during learning. Returning to the model, we find that an iterative sequence of state-specific activations during NREM/REM cycling is essential for memory storage in the network, serving a critical role during simultaneous consolidation of multiple memories. Together these results provide a testable mechanistic hypothesis for the respective roles of NREM and REM sleep, and their universal relative timing, in memory consolidation. Significance statement Using a simplified computational model and in vivo recordings from mouse hippocampus, we show that NREM and REM sleep can play differential roles in memory consolidation. The specific neurophysiological features of the two sleep states allow for expansion of memory traces (during NREM) and prevention of overlap between different memory traces (during REM). These features are likely essential in the context of storing more than one new memory simultaneously within a brain network.
Collapse
|
8
|
Park R, Kang MS, Heo G, Shin YC, Han DW, Hong SW. Regulated Behavior in Living Cells with Highly Aligned Configurations on Nanowrinkled Graphene Oxide Substrates: Deep Learning Based on Interplay of Cellular Contact Guidance. ACS NANO 2024; 18:1325-1344. [PMID: 38099607 DOI: 10.1021/acsnano.2c09815] [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: 01/17/2024]
Abstract
Micro-/nanotopographical cues have emerged as a practical and promising strategy for controlling cell fate and reprogramming, which play a key role as biophysical regulators in diverse cellular processes and behaviors. Extracellular biophysical factors can trigger intracellular physiological signaling via mechanotransduction and promote cellular responses such as cell adhesion, migration, proliferation, gene/protein expression, and differentiation. Here, we engineered a highly ordered nanowrinkled graphene oxide (GO) surface via the mechanical deformation of an ultrathin GO film on an elastomeric substrate to observe specific cellular responses based on surface-mediated topographical cues. The ultrathin GO film on the uniaxially prestrained elastomeric substrate through self-assembly and subsequent compressive force produced GO nanowrinkles with periodic amplitude. To examine the acute cellular behaviors on the GO-based cell interface with nanostructured arrays of wrinkles, we cultured L929 fibroblasts and HT22 hippocampal neuronal cells. As a result, our developed cell-culture substrate obviously provided a directional guidance effect. In addition, based on the observed results, we adapted a deep learning (DL)-based data processing technique to precisely interpret the cell behaviors on the nanowrinkled GO surfaces. According to the learning/transfer learning protocol of the DL network, we detected cell boundaries, elongation, and orientation and quantitatively evaluated cell velocity, traveling distance, displacement, and orientation. The presented experimental results have intriguing implications such that the nanotopographical microenvironment could engineer the living cells' morphological polarization to assemble them into useful tissue chips consisting of multiple cell types.
Collapse
Affiliation(s)
- Rowoon Park
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Gyeonghwa Heo
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Yong Cheol Shin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Ohio 44195, United States
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan 46241, Republic of Korea
- Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
9
|
Mao LM, Young L, Chu XP, Wang JQ. Regulation of Src family kinases by muscarinic acetylcholine receptors in heterologous cells and neurons. Front Mol Neurosci 2024; 16:1340725. [PMID: 38273940 PMCID: PMC10808654 DOI: 10.3389/fnmol.2023.1340725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Five muscarinic acetylcholine (mACh) receptor subtypes are divided into two classes: the M1 class (M1, M3, and M5) and the M2 class (M2 and M4). The former is coupled to Gq proteins, while the latter is coupled to Gi/o proteins. Accumulating evidence indicates that mACh receptors play a significant role in the regulation of the Src family kinase (SFK), a subfamily of non-receptor tyrosine kinases. mACh receptors exert their roles in a subtype-dependent fashion and preferentially target Src and Fyn, two members of SFKs that are expressed in the brain and enriched at synaptic sites. While the M1 receptor positively modulates SFK activity, the M4 receptor inhibits it. By modulating SFKs, mACh receptors are actively involved in the regulation of expression and function of a variety of receptors, structural proteins, and signaling molecules. In particular, the M4 receptor and the dopamine D1 receptor are coexpressed in striatonigral projection neurons of the striatum. Gi/o-coupled M4 and Gq-coupled D1 receptors antagonistically regulate SFK activity, thereby forming a dynamic balance controlling glutamate receptor activity, excitability of neurons, and synaptic plasticity. In summary, mACh receptors play a crucial role in regulating SFK activity in heterologous cells and neurons.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Lexi Young
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - John Q. Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
10
|
Sabbir MG. Cholinergic Receptor Muscarinic 1 Co-Localized with Mitochondria in Cultured Dorsal Root Ganglion Neurons, and Its Deletion Disrupted Mitochondrial Ultrastructure in Peripheral Neurons: Implications in Alzheimer's Disease. J Alzheimers Dis 2024; 98:247-264. [PMID: 38427478 DOI: 10.3233/jad-230883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) has been linked to the pathogenesis of Alzheimer's disease (AD). Our recent study found significantly lower CHRM1 protein levels in AD patient cortices, linked to reduced survival. Furthermore, using knockout mice (Chrm1-/-) we demonstrated that deletion of Chrm1 alters cortical mitochondrial structure and function, directly establishing a connection between its loss and mitochondrial dysfunction in the context of AD. While CHRM1's role in the brain has been extensively investigated, its impact on peripheral neurons in AD remains a crucial area of research, especially considering reported declines in peripheral nerve conduction among AD patients. Objective The objective was to characterize Chrm1 localization and mitochondrial deficits in Chrm1-/- dorsal root ganglion (DRG) neurons. Methods Recombinant proteins tagged with Green or Red Fluorescent Protein (GFP/RFP) were transiently expressed to investigate the localization of Chrm1 and mitochondria, as well as mitochondrial movement in the neurites of cultured primary mouse DRG neurons, using confocal time-lapse live cell imaging. Transmission electron microscopy was performed to examine the ultrastructure of mitochondria in both wild-type and Chrm1-/- DRGs. Results Fluorescence imaging revealed colocalization and comigration of N-terminal GFP-tagged Chrm1 and mitochondrial localization signal peptide-tagged RFP-labelled mitochondria in the DRGs neurons. A spectrum of mitochondrial structural abnormalities, including disruption and loss of cristae was observed in 87% neurons in Chrm1-/- DRGs. Conclusions This study suggests that Chrm1 may be localized in the neuronal mitochondria and loss of Chrm1 in peripheral neurons causes sever mitochondrial structural aberrations resembling AD pathology.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Department of Psychology and Neuroscience, Collegeof Psychology, Nova Southeastern University, Fort Lauderdale, FL, USA
- Alzo Biosciences Inc., San Diego, CA, USA
| |
Collapse
|
11
|
Yamanishi K, Hata M, Gamachi N, Watanabe Y, Yamanishi C, Okamura H, Matsunaga H. Molecular Mechanisms of IL18 in Disease. Int J Mol Sci 2023; 24:17170. [PMID: 38139000 PMCID: PMC10743479 DOI: 10.3390/ijms242417170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Interleukin 18 (IL18) was originally identified as an inflammation-induced cytokine that is secreted by immune cells. An increasing number of studies have focused on its non-immunological functions, with demonstrated functions for IL18 in energy homeostasis and neural stability. IL18 is reportedly required for lipid metabolism in the liver and brown adipose tissue. Furthermore, IL18 (Il18) deficiency in mice leads to mitochondrial dysfunction in hippocampal cells, resulting in depressive-like symptoms and cognitive impairment. Microarray analyses of Il18-/- mice have revealed a set of genes with differential expression in liver, brown adipose tissue, and brain; however, the impact of IL18 deficiency in these tissues remains uncertain. In this review article, we discuss these genes, with a focus on their relationships with the phenotypic disease traits of Il18-/- mice.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Masaki Hata
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Naomi Gamachi
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Chiaki Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Haruki Okamura
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| |
Collapse
|
12
|
Zhang JJ, Fu H, Lin R, Zhou J, Haider A, Fang W, Elghazawy NH, Rong J, Chen J, Li Y, Ran C, Collier TL, Chen Z, Liang SH. Imaging Cholinergic Receptors in the Brain by Positron Emission Tomography. J Med Chem 2023; 66:10889-10916. [PMID: 37583063 PMCID: PMC10461233 DOI: 10.1021/acs.jmedchem.3c00573] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Indexed: 08/17/2023]
Abstract
Cholinergic receptors represent a promising class of diagnostic and therapeutic targets due to their significant involvement in cognitive decline associated with neurological disorders and neurodegenerative diseases as well as cardiovascular impairment. Positron emission tomography (PET) is a noninvasive molecular imaging tool that has helped to shed light on the roles these receptors play in disease development and their diverse functions throughout the central nervous system (CNS). In recent years, there has been a notable advancement in the development of PET probes targeting cholinergic receptors. The purpose of this review is to provide a comprehensive overview of the recent progress in the development of these PET probes for cholinergic receptors with a specific focus on ligand structure, radiochemistry, and pharmacology as well as in vivo performance and applications in neuroimaging. The review covers the structural design, pharmacological properties, radiosynthesis approaches, and preclinical and clinical evaluations of current state-of-the-art PET probes for cholinergic receptors.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Hualong Fu
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ruofan Lin
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Jingyin Zhou
- Key
Laboratory of Radiopharmaceuticals, Ministry of Education, College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ahmed Haider
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Weiwei Fang
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Nehal H. Elghazawy
- Department
of Pharmaceutical, Chemistry, Faculty of Pharmacy & Biotechnology, German University in Cairo, 11835 Cairo, Egypt
| | - Jian Rong
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jiahui Chen
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Yinlong Li
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chongzhao Ran
- Athinoula
A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02114, United States
| | - Thomas L. Collier
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Zhen Chen
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Provincial Key Lab for the Chemistry and Utilization
of Agro-Forest Biomass, Jiangsu Key Lab of Biomass-Based Green Fuels
and Chemicals, International Innovation Center for Forest Chemicals
and Materials, College of Chemical Engineering, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Steven H. Liang
- Division
of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital
& Department of Radiology, Harvard Medical
School, Boston, Massachusetts 02114, United States
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
13
|
Zheng F, Wess J, Alzheimer C. Long-Term-But Not Short-Term-Plasticity at the Mossy Fiber-CA3 Pyramidal Cell Synapse in Hippocampus Is Altered in M1/M3 Muscarinic Acetylcholine Receptor Double Knockout Mice. Cells 2023; 12:1890. [PMID: 37508553 PMCID: PMC10378318 DOI: 10.3390/cells12141890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Muscarinic acetylcholine receptors are well-known for their crucial involvement in hippocampus-dependent learning and memory, but the exact roles of the various receptor subtypes (M1-M5) are still not fully understood. Here, we studied how M1 and M3 receptors affect plasticity at the mossy fiber (MF)-CA3 pyramidal cell synapse. In hippocampal slices from M1/M3 receptor double knockout (M1/M3-dKO) mice, the signature short-term plasticity of the MF-CA3 synapse was not significantly affected. However, the rather unique NMDA receptor-independent and presynaptic form of long-term potentiation (LTP) of this synapse was much larger in M1/M3-deficient slices compared to wild-type slices in both field potential and whole-cell recordings. Consistent with its presynaptic origin, induction of MF-LTP strongly enhanced the excitatory drive onto single CA3 pyramidal cells, with the effect being more pronounced in M1/M3-dKO cells. In an earlier study, we found that the deletion of M2 receptors in mice disinhibits MF-LTP in a similar fashion, suggesting that endogenous acetylcholine employs both M1/M3 and M2 receptors to constrain MF-LTP. Importantly, such synergism was not observed for MF long-term depression (LTD). Low-frequency stimulation, which reliably induced LTD of MF synapses in control slices, failed to do so in M1/M3-dKO slices and gave rise to LTP instead. In striking contrast, loss of M2 receptors augmented LTD when compared to control slices. Taken together, our data demonstrate convergence of M1/M3 and M2 receptors on MF-LTP, but functional divergence on MF-LTD, with the net effect resulting in a well-balanced bidirectional plasticity of the MF-CA3 pyramidal cell synapse.
Collapse
Affiliation(s)
- Fang Zheng
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Biological Chemistry, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
14
|
Nag S, Arakawa R, Jia Z, Lachapelle E, Zhang L, Maresca K, Chen L, Jahan M, Mccarthy T, Halldin C. Characterization of a Novel M4 PAM PET Radioligand [11C]PF06885190 in Nonhuman Primates (NHP). Molecules 2023; 28:4612. [PMID: 37375167 DOI: 10.3390/molecules28124612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Muscarinic acetylcholine receptors (mAChR), including M4, draw attention as therapeutic targets for several neurodegenerative diseases including Alzheimer's disease (AD). PET imaging of M4 positive allosteric modulator (PAM) allows qualification of the distribution as well as the expression of this receptor under physiological conditions and thereby helps to assess the receptor occupancy (RO) of a drug candidate. In this study, our aims were (a) to synthesize a novel M4 PAM PET radioligand [11C]PF06885190 (b) to evaluate the brain distribution of [11C]PF06885190 in nonhuman primates (NHP) and (c) to analyze its radiometabolites in the blood plasma of NHP. Radiolabeling of [11C]PF06885190 was accomplished via N-methylation of the precursor. Six PET measurements were performed using two male cynomolgus monkeys, where three PET measurements were at baseline, two after pretreatment with a selective M4 PAM compound CVL-231 and one after pretreatment with donepezil. The total volume of distribution (VT) of [11C]PF06885190 was examined using Logan graphical analysis with arterial input function. Radiometabolites were analyzed in monkey blood plasma using gradient HPLC system. Radiolabeling of [11C]PF06885190 was successfully accomplished and the radioligand was found to be stable in the formulation, with radiochemical purity exceeding 99% 1 h after the end of the synthesis. [11C]PF06885190 was characterized in the cynomolgus monkey brain where a moderate brain uptake was found at the baseline condition. However, it showed fast wash-out as it dropped to half of the peak at around 10 min. Change of VT from baseline was around -10% after pretreatment with a M4 PAM, CVL-231. Radiometabolite studies showed relatively fast metabolism. Although sufficient brain uptake of [11C]PF06885190 was observed, these data suggest that [11C]PF06885190 might have too low specific binding in the NHP brain to be further applied in PET imaging.
Collapse
Affiliation(s)
- Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 17164 Stockholm, Sweden
| | - Ryosuke Arakawa
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 17164 Stockholm, Sweden
| | - Zhisheng Jia
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 17164 Stockholm, Sweden
| | - Erik Lachapelle
- Worldwide Research, Development and Medical, Pfizer Inc., Groton, CT 06340, USA
| | - Lei Zhang
- Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Kevin Maresca
- Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Laigao Chen
- Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Mahabuba Jahan
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 17164 Stockholm, Sweden
| | - Timothy Mccarthy
- Worldwide Research, Development and Medical, Pfizer Inc., Cambridge, MA 02139, USA
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, 17164 Stockholm, Sweden
| |
Collapse
|
15
|
Sabbir MG, Swanson M, Speth RC, Albensi BC. Hippocampal versus cortical deletion of cholinergic receptor muscarinic 1 in mice differentially affects post-translational modifications and supramolecular assembly of respiratory chain-associated proteins, mitochondrial ultrastructure, and respiration: implications in Alzheimer's disease. Front Cell Dev Biol 2023; 11:1179252. [PMID: 37293125 PMCID: PMC10246746 DOI: 10.3389/fcell.2023.1179252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction: In a previous retrospective study using postmortem human brain tissues, we demonstrated that loss of Cholinergic Receptor Muscarinic 1 (CHRM1) in the temporal cortex of a subset of Alzheimer's patients was associated with poor survival, whereas similar loss in the hippocampus showed no such association. Mitochondrial dysfunction underlies Alzheimer's pathogenesis. Therefore, to investigate the mechanistic basis of our findings, we evaluated cortical mitochondrial phenotypes in Chrm1 knockout (Chrm1-/-) mice. Cortical Chrm1 loss resulted in reduced respiration, reduced supramolecular assembly of respiratory protein complexes, and caused mitochondrial ultrastructural abnormalities. These mouse-based findings mechanistically linked cortical CHRM1 loss with poor survival of Alzheimer's patients. However, evaluation of the effect of Chrm1 loss on mouse hippocampal mitochondrial characteristics is necessary to fully understand our retrospective human tissue-based observations. This is the objective of this study. Methods: Enriched hippocampal and cortical mitochondrial fractions (EHMFs/ECMFs, respectively) derived from wild-type and Chrm1-/- mice were used to measure respiration by quantifying real-time oxygen consumption, supramolecular assembly of oxidative phosphorylation (OXPHOS)-associated proteins by blue native polyacrylamide gel electrophoresis, post-translational modifications (PTMs) by isoelectric focusing (IEF), and mitochondrial ultrastructure by electron microscopy. Results: In contrast to our previous observations in Chrm1-/- ECMFs, EHMFs of Chrm1-/- mice significantly increased respiration with a concomitant increase in the supramolecular assembly of OXPHOS-associated proteins, specifically Atp5a and Uqcrc2, with no mitochondrial ultrastructural alterations. IEF of ECMFs and EHMFs from Chrm1-/- mice showed a decrease and an increase, respectively in a negatively charged (pH∼3) fraction of Atp5a relative to the wild-type mice, with a corresponding decrease or increase in the supramolecular assembly of Atp5a and respiration indicating a tissue-specific signaling effect. Discussion: Our findings indicate that loss of Chrm1 in the cortex causes structural, and physiological alterations to mitochondria that compromise neuronal function, whereas Chrm1 loss in the hippocampus may benefit neuronal function by enhancing mitochondrial function. This brain region-specific differential effect of Chrm1 deletion on mitochondrial function supports our human brain region-based findings and Chrm1-/- mouse behavioral phenotypes. Furthermore, our study indicates that Chrm1-mediated brain region-specific differential PTMs of Atp5a may alter complex-V supramolecular assembly which in turn regulates mitochondrial structure-function.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Alzo Biosciences Inc., San Diego, CA, United States
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Mamiko Swanson
- Alzo Biosciences Inc., San Diego, CA, United States
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Robert C. Speth
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, United States
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
16
|
Sabbir MG, Swanson M, Albensi BC. Loss of cholinergic receptor muscarinic 1 impairs cortical mitochondrial structure and function: implications in Alzheimer's disease. Front Cell Dev Biol 2023; 11:1158604. [PMID: 37274741 PMCID: PMC10233041 DOI: 10.3389/fcell.2023.1158604] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction: Cholinergic Receptor Muscarinic 1 (CHRM1) is a G protein-coupled acetylcholine (ACh) receptor predominantly expressed in the cerebral cortex. In a retrospective postmortem brain tissues-based study, we demonstrated that severely (≥50% decrease) reduced CHRM1 proteins in the temporal cortex of Alzheimer's patients significantly correlated with poor patient outcomes. The G protein-mediated CHRM1 signal transduction cannot sufficiently explain the mechanistic link between cortical CHRM1 loss and the appearance of hallmark Alzheimer's pathophysiologies, particularly mitochondrial structural and functional abnormalities. Therefore, the objective of this study was to analyze the molecular, ultrastructural, and functional properties of cortical mitochondria using CHRM1 knockout (Chrm1-/-) and wild-type mice to identify mitochondrial abnormalities. Methods: Isolated and enriched cortical mitochondrial fractions derived from wild-type and Chrm1-/- mice were assessed for respiratory deficits (oxygen consumption) following the addition of different substrates. The supramolecular assembly of mitochondrial oxidative phosphorylation (OXPHOS)-associated protein complexes (complex I-V) and cortical mitochondrial ultrastructure were investigated by blue native polyacrylamide gel electrophoresis and transmission electron microscopy (TEM), respectively. A cocktail of antibodies, specific to Ndufb8, Sdhb, Uqcrc2, Mtco1, and Atp5a proteins representing different subunits of complexes I-V, respectively was used to characterize different OXPHOS-associated protein complexes. Results: Loss of Chrm1 led to a significant reduction in cortical mitochondrial respiration (oxygen consumption) concomitantly associated with reduced oligomerization of ATP synthase (complex V) and supramolecular assembly of complexes I-IV (Respirasome). Overexpression of Chrm1 in transformed cells (lacking native Chrm1) significantly increased complex V oligomerization and respirasome assembly leading to enhanced respiration. TEM analysis revealed that Chrm1 loss led to mitochondrial ultrastructural defects and alteration in the tinctorial properties of cortical neurons causing a significant increase in the abundance of dark cortical neurons (Chrm1-/- 85% versus wild-type 2%). Discussion: Our findings indicate a hitherto unknown effect of Chrm1 deletion in cortical neurons affecting mitochondrial function by altering multiple interdependent factors including ATP synthase oligomerization, respirasome assembly, and mitochondrial ultrastructure. The appearance of dark neurons in Chrm1-/- cortices implies potentially enhanced glutamatergic signaling in pyramidal neurons under Chrm1 loss condition. The findings provide novel mechanistic insights into Chrm1 loss with the appearance of mitochondrial pathophysiological deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Alzo Biosciences Inc, SanDiego, CA, United States
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Mamiko Swanson
- Alzo Biosciences Inc, SanDiego, CA, United States
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
- Barry & Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
17
|
Hassani S, Neumann A, Russell J, Jones C, Womelsdorf T. M 1-selective muscarinic allosteric modulation enhances cognitive flexibility and effective salience in nonhuman primates. Proc Natl Acad Sci U S A 2023; 120:e2216792120. [PMID: 37104474 PMCID: PMC10161096 DOI: 10.1073/pnas.2216792120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 03/21/2023] [Indexed: 04/28/2023] Open
Abstract
Acetylcholine (ACh) in cortical neural circuits mediates how selective attention is sustained in the presence of distractors and how flexible cognition adjusts to changing task demands. The cognitive domains of attention and cognitive flexibility might be differentially supported by the M1 muscarinic acetylcholine receptor (mAChR) subtype. Understanding how M1 mAChR mechanisms support these cognitive subdomains is of highest importance for advancing novel drug treatments for conditions with altered attention and reduced cognitive control including Alzheimer's disease or schizophrenia. Here, we tested this question by assessing how the subtype-selective M1 mAChR positive allosteric modulator (PAM) VU0453595 affects visual search and flexible reward learning in nonhuman primates. We found that allosteric potentiation of M1 mAChRs enhanced flexible learning performance by improving extradimensional set shifting, reducing latent inhibition from previously experienced distractors and reducing response perseveration in the absence of adverse side effects. These procognitive effects occurred in the absence of apparent changes of attentional performance during visual search. In contrast, nonselective ACh modulation using the acetylcholinesterase inhibitor (AChEI) donepezil improved attention during visual search at doses that did not alter cognitive flexibility and that already triggered gastrointestinal cholinergic side effects. These findings illustrate that M1 mAChR positive allosteric modulation enhances cognitive flexibility without affecting attentional filtering of distraction, consistent with M1 activity boosting the effective salience of relevant over irrelevant objects specifically during learning. These results suggest that M1 PAMs are versatile compounds for enhancing cognitive flexibility in disorders spanning schizophrenia and Alzheimer's diseases.
Collapse
Affiliation(s)
- Seyed A. Hassani
- Department of Psychology, Vanderbilt University, Nashville, TN37240
| | - Adam Neumann
- Department of Psychology, Vanderbilt University, Nashville, TN37240
| | - Jason Russell
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN37240
| | - Carrie K. Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN37240
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN37240
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, TN37240
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN37240
| |
Collapse
|
18
|
Mahmoudi R, Novella JL, Laurent-Badr S, Boulahrouz S, Tran D, Morrone I, Jaïdi Y. Cholinergic Antagonists and Behavioral Disturbances in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:ijms24086921. [PMID: 37108085 PMCID: PMC10138684 DOI: 10.3390/ijms24086921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Cholinergic antagonists interfere with synaptic transmission in the central nervous system and are involved in pathological processes in patients with neurocognitive disorders (NCD), such as behavioral and psychological symptoms of dementia (BPSD). In this commentary, we will briefly review the current knowledge on the impact of cholinergic burden on BPSD in persons with NCD, including the main pathophysiological mechanisms. Given the lack of clear consensus regarding symptomatic management of BPSD, special attention must be paid to this preventable, iatrogenic condition in patients with NCD, and de-prescription of cholinergic antagonists should be considered in patients with BPSD.
Collapse
Affiliation(s)
- Rachid Mahmoudi
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
- UR 3797 Vieillissement, Fragilité (VieFra), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
| | - Jean Luc Novella
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
- UR 3797 Vieillissement, Fragilité (VieFra), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
| | - Sarah Laurent-Badr
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
| | - Sarah Boulahrouz
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
- UR 3797 Vieillissement, Fragilité (VieFra), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
| | - David Tran
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
| | - Isabella Morrone
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
- Cognition Health and Society Laboratory (C2S-EA 6291), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
| | - Yacine Jaïdi
- Department of Geriatric and Internal Medicine, Reims University Hospitals, Maison Blanche Hospital, 51092 Reims, France
- UR 3797 Vieillissement, Fragilité (VieFra), Faculty of Medicine, University of Reims Champagne-Ardenne, 51687 Reims, France
| |
Collapse
|
19
|
Sumi T, Harada K. Muscarinic acetylcholine receptor-dependent and NMDA receptor-dependent LTP and LTD share the common AMPAR trafficking pathway. iScience 2023; 26:106133. [PMID: 36866246 PMCID: PMC9972575 DOI: 10.1016/j.isci.2023.106133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/30/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The forebrain cholinergic system promotes higher brain function in part by signaling through the M1 muscarinic acetylcholine receptor (mAChR). Long-term potentiation (LTP) and long-term depression (LTD) of excitatory synaptic transmission in the hippocampus are also induced by mAChR. An AMPA receptor (AMPAR) trafficking model for hippocampal neurons has been proposed to simulate N-methyl-D-aspartate receptor (NMDAR)-dependent synaptic plasticity in the early phase. In this study, we demonstrated the validity of the hypothesis that the mAChR-dependent LTP/LTD shares a common AMPAR trafficking pathway associated with NMDAR-dependent LTP/LTD. However, unlike NMDAR, Ca2+ influx into the spine cytosol occurs owing to the Ca2+ stored inside the ER and is induced via the activation of inositol 1,4,5-trisphosphate (IP3) receptors during M1 mAChR activation. Moreover, the AMPAR trafficking model implies that alterations in LTP and LTD observed in Alzheimer's disease could be attributed to age-dependent reductions in AMPAR expression levels.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Corresponding author
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Tempaku-cho, Toyohashi 441-8580, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Tempaku-cho, Toyohashi, 441-8580, Japan
| |
Collapse
|
20
|
Yao R, Yamada K, Kito T, Aizu N, Iwata D, Izawa S, Nishii K, Sawada H, Chihara T. Novel shaking exercises for hippocampal and medial prefrontal cortex functioning maintain spatial working memory. Exp Gerontol 2023; 171:112024. [PMID: 36372283 DOI: 10.1016/j.exger.2022.112024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
INTRODUCTION The decline in spatial working memory is one of the earliest signs of normal brain aging. OBJECTIVE We developed a novel physical exercise method, termed the "shaking exercise," to slow down this process. METHODS The experimental protocol included administering the shaking exercise for 8-32 weeks in male senescence-accelerated mouse prone 10 (SAMP-10). They were subjected to the T-maze test, followed by immunohistochemical analysis, to assess the influence of the shaking exercise on the M1 muscarinic acetylcholine receptor (CHRM1) and α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) of the dorsal hippocampus and medial prefrontal cortex (dHC-mPFC). RESULTS The T-maze test demonstrated that the shaking group had less hesitation in the face of selecting direction at week 24. In the immunohistochemical analysis, more CHRM1s were in the CA3 subregion and more AMPARs were in the subiculum. CHRM1s and AMPARs were maintained in the CA1 region and the mPFC. The CHRM1s seem to have a positive effect on the AMPAR in the dentate gyrus (DG) region and the CA3 region. In the CA1 region, CHRM1s were negatively correlated with AMPARs. In addition, high-density neurons were expressed in the shaking group in the upstream DG, the middle part and the distal part of CA3, the distal part of CA1, and the mPFC. CONCLUSIONS Our results raise the possibility that maintenance of the spatial working memory effect observed with the shaking exercise is driven in part by the uneven affection of CHRM1s and AMPARs in the dHC-mPFC circuit system and significantly maintains the neuronal expression in the dHC-mPFC.
Collapse
Affiliation(s)
- Runhong Yao
- Department of Physical Therapy, School of Health Sciences, Japan University of Health Sciences, Satte, Saitama 340-0145, Japan; Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Kouji Yamada
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Takumi Kito
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan; Department of Physical Therapy, Faculty of Health Sciences, Kinjo University, Hakusan, Ishikawa 924-8511, Japan
| | - Naoki Aizu
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Daiki Iwata
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Sho Izawa
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Kazuhiro Nishii
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan.
| | - Hirohide Sawada
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan.
| | - Takeshi Chihara
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Mie 512-8045, Japan.
| |
Collapse
|
21
|
Structure-activity relationship of pyrazol-4-yl-pyridine derivatives and identification of a radiofluorinated probe for imaging the muscarinic acetylcholine receptor M 4. Acta Pharm Sin B 2023; 13:213-226. [PMID: 36815036 PMCID: PMC9939360 DOI: 10.1016/j.apsb.2022.07.008] [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: 01/22/2022] [Revised: 05/13/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
There is an accumulating body of evidence implicating the muscarinic acetylcholine receptor 4 (M4) in schizophrenia and dementia with Lewy bodies, however, a clinically validated M4 positron emission tomography (PET) radioligand is currently lacking. As such, the aim of this study was to develop a suitable M4 PET ligand that allows the non-invasive visualization of M4 in the brain. Structure-activity relationship studies of pyrazol-4-yl-pyridine derivates led to the discovery of target compound 12 - a subtype-selective positive allosteric modulator (PAM). The radiofluorinated analogue, [18F]12, was synthesized in 28 ± 10% radiochemical yield, >37 GBq/μmol and an excellent radiochemical purity >99%. Initial in vitro autoradiograms on rodent brain sections were performed in the absence of carbachol and showed moderate specificity as well as a low selectivity of [18F]12 for the M4-rich striatum. However, in the presence of carbachol, a significant increase in tracer binding was observed in the rat striatum, which was reduced by >60% under blocking conditions, thus indicating that orthosteric ligand interaction is required for efficient binding of [18F]12 to the allosteric site. Remarkably, however, the presence of carbachol was not required for high specific binding in the non-human primate (NHP) and human striatum, and did not further improve the specificity and selectivity of [18F]12 in higher species. These results pointed towards significant species-differences and paved the way for a preliminary PET study in NHP, where peak brain uptake of [18F]12 was found in the putamen and temporal cortex. In conclusion, we report on the identification and preclinical development of the first radiofluorinated M4 PET radioligand with promising attributes. The availability of a clinically validated M4 PET radioligand harbors potential to facilitate drug development and provide a useful diagnostic tool for non-invasive imaging.
Collapse
|
22
|
Szczurowska E, Szánti-Pintér E, Chetverikov N, Randáková A, Kudová E, Jakubík J. Modulation of Muscarinic Signalling in the Central Nervous System by Steroid Hormones and Neurosteroids. Int J Mol Sci 2022; 24:ijms24010507. [PMID: 36613951 PMCID: PMC9820491 DOI: 10.3390/ijms24010507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Muscarinic acetylcholine receptors expressed in the central nervous system mediate various functions, including cognition, memory, or reward. Therefore, muscarinic receptors represent potential pharmacological targets for various diseases and conditions, such as Alzheimer's disease, schizophrenia, addiction, epilepsy, or depression. Muscarinic receptors are allosterically modulated by neurosteroids and steroid hormones at physiologically relevant concentrations. In this review, we focus on the modulation of muscarinic receptors by neurosteroids and steroid hormones in the context of diseases and disorders of the central nervous system. Further, we propose the potential use of neuroactive steroids in the development of pharmacotherapeutics for these diseases and conditions.
Collapse
Affiliation(s)
- Ewa Szczurowska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
| | - Eszter Szánti-Pintér
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
| | - Nikolai Chetverikov
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Alena Randáková
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Eva Kudová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
- Correspondence: (E.K.); (J.J.)
| | - Jan Jakubík
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
- Correspondence: (E.K.); (J.J.)
| |
Collapse
|
23
|
Qian XH, Liu XL, Chen G, Chen SD, Tang HD. Injection of amyloid-β to lateral ventricle induces gut microbiota dysbiosis in association with inhibition of cholinergic anti-inflammatory pathways in Alzheimer's disease. J Neuroinflammation 2022; 19:236. [PMID: 36171620 PMCID: PMC9520842 DOI: 10.1186/s12974-022-02599-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease and its pathogenesis is still unclear. There is dysbiosis of gut microbiota in AD patients. More importantly, dysbiosis of the gut microbiota has been observed not only in AD patients, but also in patients with mild cognitive impairment (MCI). However, the mechanism of gut microbiota dysbiosis in AD is poorly understood. Cholinergic anti-inflammatory pathway is an important pathway for the central nervous system (CNS) regulation of peripheral immune homeostasis, especially in the gut. Therefore, we speculated that dysfunction of cholinergic anti-inflammatory pathway is a potential pathway for dysbiosis of the gut microbiota in AD. METHODS In this study, we constructed AD model mice by injecting Aβ1-42 into the lateral ventricle, and detected the cognitive level of mice by the Morris water maze test. In addition, 16S rDNA high-throughput analysis was used to detect the gut microbiota abundance of each group at baseline, 2 weeks and 4 weeks after surgery. Furthermore, immunofluorescence and western blot were used to detect alteration of intestinal structure of mice, cholinergic anti-inflammatory pathway, and APP process of brain and colon in each group. RESULTS Aβ1-42 i.c.v induced cognitive impairment and neuron damage in the brain of mice. At the same time, Aβ1-42 i.c.v induced alteration of gut microbiota at 4 weeks after surgery, while there was no difference at the baseline and 2 weeks after surgery. In addition, changes in colon structure and increased levels of pro-inflammatory factors were detected in Aβ1-42 treatment group, accompanied by inhibition of cholinergic anti-inflammatory pathways. Amyloidogenic pathways in both the brain and colon were accelerated in Aβ1-42 treatment group. CONCLUSIONS The present findings suggested that Aβ in the CNS can induce gut microbiota dysbiosis, alter intestinal structure and accelerate the amyloidogenic pathways, which were related to inhibiting cholinergic anti-inflammatory pathways.
Collapse
Affiliation(s)
- Xiao-Hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406, China
| | - Guang Chen
- The Second Hospital of Anhui Medical University, Anhui, 230601, China
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Medical Center on Aging of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
24
|
Sabbir MG, Speth RC, Albensi BC. Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) Protein in the Hippocampus and Temporal Cortex of a Subset of Individuals with Alzheimer’s Disease, Parkinson’s Disease, or Frontotemporal Dementia: Implications for Patient Survival. J Alzheimers Dis 2022; 90:727-747. [DOI: 10.3233/jad-220766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Dysfunction of cholinergic neurotransmission is a hallmark of Alzheimer’s disease (AD); forming the basis for using acetylcholine (ACh) esterase (AChE) inhibitors to mitigate symptoms of ACh deficiency in AD. The Cholinergic Receptor Muscarinic 1 (CHRM1) is highly expressed in brain regions impaired by AD. Previous analyses of postmortem AD brains revealed unaltered CHRM1 mRNA expression compared to normal brains. However, the CHRM1 protein level in AD and other forms of dementia has not been extensively studied. Reduced expression of CHRM1 in AD patients may explain the limited clinical efficacy of AChE inhibitors. Objective: To quantify CHRM1 protein in the postmortem hippocampus and temporal cortex of AD, Parkinson’s disease (PD), and frontotemporal dementia (FTD) patients. Methods: Western blotting was performed on postmortem hippocampus (N = 19/73/7/9: unaffected/AD/FTD/PD) and temporal cortex (N = 9/74/27: unaffected/AD/PD) using a validated anti-CHRM1 antibody. Results: Quantification based on immunoblotting using a validated anti-CHRM1 antibody revealed a significant loss of CHRM1 protein level (<50%) in the hippocampi (78% AD, 66% PD, and 85% FTD) and temporal cortices (56% AD and 42% PD) of dementia patients. Loss of CHRM1 in the temporal cortex was significantly associated with early death (<65–75 years) for both AD and PD patients. Conclusion: Severe reduction of CHRM1 in a subset of AD and PD patients can explain the reported low efficacy of AChE inhibitors as a mitigating treatment for dementia patients. Based on this study, it can be suggested that future research should prioritize therapeutic restoration of CHRM1 protein levels in cholinergic neurons.
Collapse
Affiliation(s)
- Mohammad Golam Sabbir
- Alzo Biosciences Inc., San Diego, CA, USA
- St. Boniface Hospital Albrechtsen Research Centre, Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Manitoba, Canada
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
| | - Robert C. Speth
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC, USA
| | - Benedict C. Albensi
- Nova Southeastern University, College of Pharmacy, Davie, FL, USA
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, Manitoba, Canada
- University of Manitoba, College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
25
|
Dwomoh L, Tejeda G, Tobin A. Targeting the M1 muscarinic acetylcholine receptor in Alzheimer's disease. Neuronal Signal 2022; 6:NS20210004. [PMID: 35571495 PMCID: PMC9069568 DOI: 10.1042/ns20210004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) remains a major cause of morbidity and mortality worldwide, and despite extensive research, only a few drugs are available for management of the disease. One strategy has been to up-regulate cholinergic neurotransmission to improve cognitive function, but this approach has dose-limiting adverse effects. To avoid these adverse effects, new drugs that target specific receptor subtypes of the cholinergic system are needed, and the M1 subtype of muscarinic acetylcholine receptor (M1-mAChR) has been shown to be a good target for this approach. By using several strategies, M1-mAChR ligands have been developed and trialled in preclinical animal models and in human studies, with varying degrees of success. This article reviews the different approaches to targeting the M1-mAChR in AD and discusses the advantages and limitations of these strategies. The factors to consider in targeting the M1-mAChR in AD are also discussed.
Collapse
Affiliation(s)
- Louis Dwomoh
- 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
| | - Gonzalo S. Tejeda
- 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
| | - 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
|
26
|
Drug Design Targeting the Muscarinic Receptors and the Implications in Central Nervous System Disorders. Biomedicines 2022; 10:biomedicines10020398. [PMID: 35203607 PMCID: PMC8962391 DOI: 10.3390/biomedicines10020398] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
There is substantial evidence that cholinergic system function impairment plays a significant role in many central nervous system (CNS) disorders. During the past three decades, muscarinic receptors (mAChRs) have been implicated in various pathologies and have been prominent targets of drug-design efforts. However, due to the high sequence homology of the orthosteric binding site, many drug candidates resulted in limited clinical success. Although several advances in treating peripheral pathologies have been achieved, targeting CNS pathologies remains challenging for researchers. Nevertheless, significant progress has been made in recent years to develop functionally selective orthosteric and allosteric ligands targeting the mAChRs with limited side effect profiles. This review highlights past efforts and focuses on recent advances in drug design targeting these receptors for Alzheimer’s disease (AD), schizophrenia (SZ), and depression.
Collapse
|
27
|
Tsolias A, Medalla M. Muscarinic Acetylcholine Receptor Localization on Distinct Excitatory and Inhibitory Neurons Within the ACC and LPFC of the Rhesus Monkey. Front Neural Circuits 2022; 15:795325. [PMID: 35087381 PMCID: PMC8786743 DOI: 10.3389/fncir.2021.795325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/09/2021] [Indexed: 12/14/2022] Open
Abstract
Acetylcholine (ACh) can act on pre- and post-synaptic muscarinic receptors (mAChR) in the cortex to influence a myriad of cognitive processes. Two functionally-distinct regions of the prefrontal cortex-the lateral prefrontal cortex (LPFC) and the anterior cingulate cortex (ACC)-are differentially innervated by ascending cholinergic pathways yet, the nature and organization of prefrontal-cholinergic circuitry in primates are not well understood. Using multi-channel immunohistochemical labeling and high-resolution microscopy, we found regional and laminar differences in the subcellular localization and the densities of excitatory and inhibitory subpopulations expressing m1 and m2 muscarinic receptors, the two predominant cortical mAChR subtypes, in the supragranular layers of LPFC and ACC in rhesus monkeys (Macaca mulatta). The subset of m1+/m2+ expressing SMI-32+ pyramidal neurons labeled in layer 3 (L3) was denser in LPFC than in ACC, while m1+/m2+ SMI-32+ neurons co-expressing the calcium-binding protein, calbindin (CB) was greater in ACC. Further, we found between-area differences in laminar m1+ dendritic expression, and m2+ presynaptic localization on cortico-cortical (VGLUT1+) and sub-cortical inputs (VGLUT2+), suggesting differential cholinergic modulation of top-down vs. bottom-up inputs in the two areas. While almost all inhibitory interneurons-identified by their expression of parvalbumin (PV+), CB+, and calretinin (CR+)-expressed m1+, the localization of m2+ differed by subtype and area. The ACC exhibited a greater proportion of m2+ inhibitory neurons compared to the LPFC and had a greater density of presynaptic m2+ localized on inhibitory (VGAT+) inputs targeting proximal somatodendritic compartments and axon initial segments of L3 pyramidal neurons. These data suggest a greater capacity for m2+-mediated cholinergic suppression of inhibition in the ACC compared to the LPFC. The anatomical localization of muscarinic receptors on ACC and LPFC micro-circuits shown here contributes to our understanding of diverse cholinergic neuromodulation of functionally-distinct prefrontal areas involved in goal-directed behavior, and how these interactions maybe disrupted in neuropsychiatric and neurological conditions.
Collapse
Affiliation(s)
- Alexandra Tsolias
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| |
Collapse
|
28
|
OUP accepted manuscript. Brain 2022; 145:2250-2275. [DOI: 10.1093/brain/awac096] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
|
29
|
Suo WZ. GRK5 Deficiency Causes Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 85:1399-1410. [PMID: 34958040 DOI: 10.3233/jad-215379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Prevention of Alzheimer's disease (AD) is a high priority mission while searching for a disease modifying therapy for AD, a devastating major public health crisis. Clinical observations have identified a prodromal stage of AD for which the patients have mild cognitive impairment (MCI) though do not yet meet AD diagnostic criteria. As an identifiable transitional stage before the onset of AD, MCI should become the high priority target for AD prevention, assuming successful prevention of MCI and/or its conversion to AD also prevents the subsequent AD. By pulling this string, one demonstrated cause of amnestic MCI appears to be the deficiency of G protein-coupled receptor-5 (GRK5). The most compelling evidence is that GRK5 knockout (GRK5KO) mice naturally develop into aMCI during aging. Moreover, GRK5 deficiency was reported to occur during prodromal stage of AD in CRND8 transgenic mice. When a GRK5KO mouse was crossbred with Tg2576 Swedish amyloid precursor protein transgenic mouse, the resulted double transgenic GAP mice displayed exaggerated behavioral and pathological changes across the spectrum of AD pathogenesis. Therefore, the GRK5 deficiency possesses unique features and advantage to serve as a prophylactic therapeutic target for MCI due to AD.
Collapse
Affiliation(s)
- William Z Suo
- Laboratory for Alzheimer's Disease & Aging Research, VA Medical Center, Kansas City, MO, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,The University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| |
Collapse
|
30
|
Biased M1 muscarinic receptor mutant mice show accelerated progression of prion neurodegenerative disease. Proc Natl Acad Sci U S A 2021; 118:2107389118. [PMID: 34893539 PMCID: PMC8685681 DOI: 10.1073/pnas.2107389118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/14/2023] Open
Abstract
The M1 muscarinic acetylcholine receptor (M1-receptor) plays a crucial role in learning and memory and is a validated drug target for the treatment of Alzheimer’s disease (AD). Furthermore, M1-receptor ligands have been demonstrated to display disease-modifying effects in preclinical models of neurodegenerative disease. By employing a genetic mouse model expressing a G protein–biased M1-receptor in combination with a mouse model of terminal neurodegenerative disease, we demonstrate here that the M1-receptor exerts an inherent neuroprotective activity that is dependent on its phosphorylation status. Thus, in AD drug development programs, M1-receptor ligands that maintain the receptor phosphorylation status will be more likely to lead to beneficial neuroprotective outcomes. There are currently no treatments that can slow the progression of neurodegenerative diseases, such as Alzheimer’s disease (AD). There is, however, a growing body of evidence that activation of the M1 muscarinic acetylcholine receptor (M1-receptor) can not only restore memory loss in AD patients but in preclinical animal models can also slow neurodegenerative disease progression. The generation of an effective medicine targeting the M1-receptor has however been severely hampered by associated cholinergic adverse responses. By using genetically engineered mouse models that express a G protein–biased M1-receptor, we recently established that M1-receptor mediated adverse responses can be minimized by ensuring activating ligands maintain receptor phosphorylation/arrestin-dependent signaling. Here, we use these same genetic models in concert with murine prion disease, a terminal neurodegenerative disease showing key hallmarks of AD, to establish that phosphorylation/arrestin-dependent signaling delivers neuroprotection that both extends normal animal behavior and prolongs the life span of prion-diseased mice. Our data point to an important neuroprotective property inherent to the M1-receptor and indicate that next generation M1-receptor ligands designed to drive receptor phosphorylation/arrestin-dependent signaling would potentially show low adverse responses while delivering neuroprotection that will slow disease progression.
Collapse
|
31
|
Ayano G, Betts K, Dachew BA, Alati R. Maternal smoking during pregnancy and poor academic performance in adolescent offspring: A registry data-based cohort study. Addict Behav 2021; 123:107072. [PMID: 34364108 DOI: 10.1016/j.addbeh.2021.107072] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/08/2021] [Accepted: 07/28/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Previous studies have suggested associations between maternal smoking during pregnancy (MSDP) and a range of adverse outcomes in offspring. However, evidence reporting adverse effects on poor academic performance in adolescence is scant. METHODS This register-based cohort study used linked data obtained from New South Wales (NSW) educational and health registries in Australia. MSDP was assessed using self-reports of smoking during pregnancy. Offspring's educational performance was assessed using the National Assessment Program for Literacy and numeracy (NAPLAN), when students were in grade 9 and approximately aged 14 years. We used multivariable logistic regression models to explore associations. RESULTS Adolescent offspring exposed to MSDP were at an increased risk of substandard academic performance in all domains, with the highest odds for spelling [OR, 3.12 (95%CI 2.98-3.26)] followed by writing [OR, 2.97 (95%CI 2.84-3.11)], reading [OR, 2.49 (95%CI 2.37-2.62)], and numeracy [OR, 2.43 (95%CI 2.30-2.58)]. In our sex-stratified analysis, MSDP displayed stronger effects on the academic performance of female offspring in all domains. CONCLUSIONS Our findings showed that MSDP was associated with an increased risk of reduced academic performance in adolescent offspring. The different effects of MSDP on the academic performance of male and female offspring is a new finding, which needs further investigation.
Collapse
|
32
|
Kurimoto E, Yamada R, Hirakawa T, Kimura H. Therapeutic potential of TAK-071, a muscarinic M 1 receptor positive allosteric modulator with low cooperativity, for the treatment of cognitive deficits and negative symptoms associated with schizophrenia. Neurosci Lett 2021; 764:136240. [PMID: 34509568 DOI: 10.1016/j.neulet.2021.136240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 08/27/2021] [Accepted: 09/07/2021] [Indexed: 11/27/2022]
Abstract
The selective activation of the muscarinic M1 receptor (M1R) may be a promising approach for treating cognitive impairment associated with cholinergic dysfunction. We previously reported that low cooperativity (α-value) is associated with a favorable cholinergic side effect profile of M1R positive allosteric modulators (M1 PAMs), as well as being a crucial factor for the cognitive improvement observed after combining M1 PAMs with donepezil, in rodents. In this study, we preclinically characterized TAK-071, a novel M1 PAM with low cooperativity (α-value = 199), as a new therapy for schizophrenia. We tested TAK-071 in the offspring of polyriboinosinic-polyribocytidylic acid-treated dams, which is a maternal immune activation model of schizophrenia. TAK-071 improved sociability deficits and working memory in this model. In a genetic mouse model of schizophrenia, miR-137 transgenic (Tg) mice, TAK-071 improved deficits in working memory, recognition memory, sociability, and sensorimotor gating. Patients with schizophrenia usually take several antipsychotics to treat positive symptoms. Thus, we also investigated the combined effects of TAK-071 with currently prescribed antipsychotics. Among the 10 antipsychotics tested, only olanzapine and quetiapine showed M1R antagonistic effects, which were counteracted by TAK-071 at possible effective concentrations for cognitive improvement in vitro. Moreover, haloperidol did not affect the ability of TAK-071 to improve working memory in miR-137 Tg mice, suggesting a low risk of losing efficacy when combined with dopamine D2 receptor antagonists. In conclusion, TAK-071 can exert beneficial effects on social behavior and cognitive function and could be a new therapy for schizophrenia.
Collapse
Affiliation(s)
- Emi Kurimoto
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Ryuji Yamada
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Takeshi Hirakawa
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Haruhide Kimura
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
33
|
Amro Z, Yool AJ, Collins-Praino LE. The potential role of glial cells in driving the prion-like transcellular propagation of tau in tauopathies. Brain Behav Immun Health 2021; 14:100242. [PMID: 34589757 PMCID: PMC8474563 DOI: 10.1016/j.bbih.2021.100242] [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: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Dementia is one of the leading causes of death worldwide, with tauopathies, a class of diseases defined by pathology associated with the microtubule-enriched protein, tau, as the major contributor. Although tauopathies, such as Alzheimer's disease and Frontotemporal dementia, are common amongst the ageing population, current effective treatment options are scarce, primarily due to the incomplete understanding of disease pathogenesis. The mechanisms via which aggregated forms of tau are able to propagate from one anatomical area to another to cause disease spread and progression is yet unknown. The prion-like hypothesis of tau propagation proposes that tau can propagate along neighbouring anatomical areas in a similar manner to prion proteins in prion diseases, such as Creutzfeldt-Jacob disease. This hypothesis has been supported by a plethora of studies that note the ability of tau to be actively secreted by neurons, propagated and internalised by neighbouring neuronal cells, causing disease spread. Surfacing research suggests a role of reactive astrocytes and microglia in early pre-clinical stages of tauopathy through their inflammatory actions. Furthermore, both glial types are able to internalise and secrete tau from the extracellular space, suggesting a potential role in tau propagation; although understanding the physiological mechanisms by which this can occur remains poorly understood. This review will discuss the current literature around the prion-like propagation of tau, with particular emphasis on glial-mediated neuroinflammation and the contribution it may play in this propagation process.
Collapse
Affiliation(s)
- Zein Amro
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | | |
Collapse
|
34
|
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
|
35
|
Bao W, Xie F, Zuo C, Guan Y, Huang YH. PET Neuroimaging of Alzheimer's Disease: Radiotracers and Their Utility in Clinical Research. Front Aging Neurosci 2021; 13:624330. [PMID: 34025386 PMCID: PMC8134674 DOI: 10.3389/fnagi.2021.624330] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's Disease (AD), the leading cause of senile dementia, is a progressive neurodegenerative disorder affecting millions of people worldwide and exerting tremendous socioeconomic burden on all societies. Although definitive diagnosis of AD is often made in the presence of clinical manifestations in late stages, it is now universally believed that AD is a continuum of disease commencing from the preclinical stage with typical neuropathological alterations appearing decades prior to its first symptom, to the prodromal stage with slight symptoms of amnesia (amnestic mild cognitive impairment, aMCI), and then to the terminal stage with extensive loss of basic cognitive functions, i.e., AD-dementia. Positron emission tomography (PET) radiotracers have been developed in a search to meet the increasing clinical need of early detection and treatment monitoring for AD, with reference to the pathophysiological targets in Alzheimer's brain. These include the pathological aggregations of misfolded proteins such as β-amyloid (Aβ) plagues and neurofibrillary tangles (NFTs), impaired neurotransmitter system, neuroinflammation, as well as deficient synaptic vesicles and glucose utilization. In this article we survey the various PET radiotracers available for AD imaging and discuss their clinical applications especially in terms of early detection and cognitive relevance.
Collapse
Affiliation(s)
- Weiqi Bao
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huanshan Hospital, Fudan University, Shanghai, China
| | - Yiyun Henry Huang
- Department of Radiology and Biomedical Imaging, PET Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
36
|
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
|
37
|
Protective Effect of Processed Polygoni multiflori Radix and Its Major Substance during Scopolamine-Induced Cognitive Dysfunction. Processes (Basel) 2021. [DOI: 10.3390/pr9020342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cognitive disorder in the elderly population. However, effective pharmacological agents targeting AD have not been developed. The processed Polygoni multiflori Radix (PPM) and its main active substance, 2,3,5,4′-tetrahydroxystilbene-2-O-β-glucoside (TSG), has received considerable attention, majorly due to its neuroprotective activities against multiple biological activities within the human body. In this study, we provide new evidence on the therapeutic effect of PPM and TSG during cognitive impairment by evaluating the ameliorative potential of PPM and TSG in scopolamine-induced amnesia in ICR mice. PPM (100 or 200 mg/kg) was orally administered during the experimental period (days 1–15), and scopolamine was intraperitoneally injected to induce cognitive deficits during the behavioural test periods (days 8–15). The administration of PPM and TSG significantly improved memory loss and cognitive dysfunction in behavioural tests and regulated the cholinergic function, brain-derived neurotrophic factor, and neural apoptosis. The present study suggests that PPM and TSG improved scopolamine-induced cognitive dysfunction, but further study has to be supported for the clinical application of PPM and TSG for AD prevention and treatment.
Collapse
|
38
|
van der Westhuizen ET, Choy KHC, Valant C, McKenzie-Nickson S, Bradley SJ, Tobin AB, Sexton PM, Christopoulos A. Fine Tuning Muscarinic Acetylcholine Receptor Signaling Through Allostery and Bias. Front Pharmacol 2021; 11:606656. [PMID: 33584282 PMCID: PMC7878563 DOI: 10.3389/fphar.2020.606656] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
The M1 and M4 muscarinic acetylcholine receptors (mAChRs) are highly pursued drug targets for neurological diseases, in particular for Alzheimer's disease and schizophrenia. Due to high sequence homology, selective targeting of any of the M1-M5 mAChRs through the endogenous ligand binding site has been notoriously difficult to achieve. With the discovery of highly subtype selective mAChR positive allosteric modulators in the new millennium, selectivity through targeting an allosteric binding site has opened new avenues for drug discovery programs. However, some hurdles remain to be overcome for these promising new drug candidates to progress into the clinic. One challenge is the potential for on-target side effects, such as for the M1 mAChR where over-activation of the receptor by orthosteric or allosteric ligands can be detrimental. Therefore, in addition to receptor subtype selectivity, a drug candidate may need to exhibit a biased signaling profile to avoid such on-target adverse effects. Indeed, recent studies in mice suggest that allosteric modulators for the M1 mAChR that bias signaling toward specific pathways may be therapeutically important. This review brings together details on the signaling pathways activated by the M1 and M4 mAChRs, evidence of biased agonism at these receptors, and highlights pathways that may be important for developing new subtype selective allosteric ligands to achieve therapeutic benefit.
Collapse
Affiliation(s)
- Emma T. van der Westhuizen
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - K. H. Christopher Choy
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Simon McKenzie-Nickson
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Sophie J. Bradley
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Andrew B. Tobin
- Centre for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Patrick M. Sexton
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute for Pharmaceutical Research, Monash University, Parkville, VIC, Australia
| |
Collapse
|
39
|
Hay YA, Jarzebowski P, Zhang Y, Digby R, Brendel V, Paulsen O, Magloire V. Cholinergic modulation of Up-Down states in the mouse medial entorhinal cortex in vitro. Eur J Neurosci 2020; 53:1378-1393. [PMID: 33131134 DOI: 10.1111/ejn.15032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/07/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022]
Abstract
Cholinergic tone is high during wake and rapid eye movement sleep and lower during slow wave sleep (SWS). Nevertheless, the low tone of acetylcholine during SWS modulates sharp wave ripple incidence in the hippocampus and slow wave activity in the neocortex. Linking the hippocampus and neocortex, the medial entorhinal cortex (mEC) regulates the coupling between these structures during SWS, alternating between silent Down states and active Up states, which outlast neocortical ones. Here, we investigated how low physiological concentrations of acetylcholine (ACh; 100-500 nM) modulate Up and Down states in a mEC slice preparation. We find that ACh has a dual effect on mEC activity: it prolongs apparent Up state duration as recorded in individual cells and decreases the total synaptic charge transfer, without affecting the duration of detectable synaptic activity. The overall outcome of ACh application is excitatory and we show that ACh increases Up state incidence via muscarinic receptor activation. The mean firing rate of principal neurons increased in around half of the cells while the other half showed a decrease in firing rate. Using two-photon calcium imaging of population activity, we found that population-wide network events are more frequent and rhythmic during ACh and confirmed that ACh modulates cell participation in these network events, consistent with a role for cholinergic modulation in regulating information flow between the hippocampus and neocortex during SWS.
Collapse
Affiliation(s)
- Y Audrey Hay
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Przemyslaw Jarzebowski
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Yu Zhang
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Richard Digby
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Viktoria Brendel
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Vincent Magloire
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of Cambridge, Cambridge, UK.,UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
40
|
Stolero N, Frenkel D. The dialog between neurons and microglia in Alzheimer's disease: The neurotransmitters view. J Neurochem 2020; 158:1412-1424. [PMID: 33314073 DOI: 10.1111/jnc.15262] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/12/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Microglia play a vital role in maintaining brain homeostasis. Their continuous sensing of surrounding micro-environments is crucial for their activity. Cross talk between specific neurons and microglia might occur through specific neurotransmitter receptors on microglia. Impairment with this interaction might result in pathological activity of microglia against potential insults. The reason for this activity in many neurodegenerative diseases such as Alzheimer's disease (AD) is not known. However, several papers report of the effects of different neurotransmitter agonists on microglial cells function that relate to their activity in AD. This review aims to summarize those works and to raise potential fundamental questions for future research.
Collapse
Affiliation(s)
- Nofar Stolero
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics School, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
41
|
Effects of muscarinic M 1 receptor stimulation on reinforcing and neurochemical effects of cocaine in rats. Neuropsychopharmacology 2020; 45:1994-2002. [PMID: 32344426 PMCID: PMC7547714 DOI: 10.1038/s41386-020-0684-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
Abstract
Cocaine addiction is a chronic illness characterized by maladaptive drug-induced neuroplastic changes that confer lasting vulnerability to relapse. Over several weeks we observed the effects of the M1 receptor-selective agonist VU0364572 in adult male rats that self-administer cocaine in a cocaine vs. food choice procedure. The drug showed unusual long-lasting effects, as rats gradually stopped self-administering cocaine, reallocating behavior towards the food reinforcer. The effect lasted as long as tested and at least 4 weeks. To begin to elucidate how VU0364572 modulates cocaine self-administration, we then examined its long-term effects using dual-probe in vivo dopamine and glutamate microdialysis in nucleus accumbens and medial prefrontal cortex, and ex vivo striatal dopamine reuptake. Microdialysis revealed marked decreases in cocaine-induced dopamine and glutamate outflow 4 weeks after VU0364572 treatment, without significant changes in dopamine uptake function. These lasting and marked effects of M1 receptor stimulation reinforce our interest in this target as potential treatment of cocaine addiction. M1 receptors are known to modulate medium spiny neuron responses to corticostriatal glutamatergic signaling acutely, and we hypothesize that VU0364572 may oppose the addiction-related effects of cocaine by causing lasting changes in this system.
Collapse
|
42
|
Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, Gyengesi E. Cholinergic Modulation of Glial Function During Aging and Chronic Neuroinflammation. Front Cell Neurosci 2020; 14:577912. [PMID: 33192323 PMCID: PMC7594524 DOI: 10.3389/fncel.2020.577912] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex biological process that increases the risk of age-related cognitive degenerative diseases such as dementia, including Alzheimer’s disease (AD), Lewy Body Dementia (LBD), and mild cognitive impairment (MCI). Even non-pathological aging of the brain can involve chronic oxidative and inflammatory stress, which disrupts the communication and balance between the brain and the immune system. There has been an increasingly strong connection found between chronic neuroinflammation and impaired memory, especially in AD. While microglia and astrocytes, the resident immune cells of the central nervous system (CNS), exerting beneficial effects during the acute inflammatory phase, during chronic neuroinflammation they can become more detrimental. Central cholinergic circuits are involved in maintaining normal cognitive function and regulating signaling within the entire cerebral cortex. While neuronal-glial cholinergic signaling is anti-inflammatory and anti-oxidative, central cholinergic neuronal degeneration is implicated in impaired learning, memory sleep regulation, and attention. Although there is evidence of cholinergic involvement in memory, fewer studies have linked the cholinergic anti-inflammatory and anti-oxidant pathways to memory processes during development, normal aging, and disease states. This review will summarize the current knowledge of cholinergic effects on microglia and astroglia, and their role in both anti-inflammatory and anti-oxidant mechanisms, concerning normal aging and chronic neuroinflammation. We provided details on how stimulation of α7 nicotinic acetylcholine (α7nACh) receptors can be neuroprotective by increasing amyloid-β phagocytosis, decreasing inflammation and reducing oxidative stress by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and decreasing the release of pro-inflammatory cytokines. There is also evidence for astroglial α7nACh receptor stimulation mediating anti-inflammatory and antioxidant effects by inhibiting the nuclear factor-κB (NF-κB) pathway and activating the Nrf2 pathway respectively. We conclude that targeting cholinergic glial interactions between neurons and glial cells via α7nACh receptors could regulate neuroinflammation and oxidative stress, relevant to the treatment of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rashmi Gamage
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ingrid Wagnon
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ilaria Rossetti
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ryan Childs
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
43
|
De La-Rocque S, Moretto E, Butnaru I, Schiavo G. Knockin' on heaven's door: Molecular mechanisms of neuronal tau uptake. J Neurochem 2020; 156:563-588. [PMID: 32770783 PMCID: PMC8432157 DOI: 10.1111/jnc.15144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/18/2022]
Abstract
Since aggregates of the microtubule‐binding protein tau were found to be the main component of neurofibrillary tangles more than 30 years ago, their contribution to neurodegeneration in Alzheimer's disease (AD) and tauopathies has become well established. Recent work shows that both tau load and its distribution in the brain of AD patients correlate with cognitive decline more closely compared to amyloid plaque deposition. In addition, the amyloid cascade hypothesis has been recently challenged because of disappointing results of clinical trials designed to treat AD by reducing beta‐amyloid levels, thus fuelling a renewed interest in tau. There is now robust evidence to indicate that tau pathology can spread within the central nervous system via a prion‐like mechanism following a stereotypical pattern, which can be explained by the trans‐synaptic inter‐neuronal transfer of pathological tau. In the receiving neuron, tau has been shown to take multiple routes of internalisation, which are partially dependent on its conformation and aggregation status. Here, we review the emerging mechanisms proposed for the uptake of extracellular tau in neurons and the requirements for the propagation of its pathological conformers, addressing how they gain access to physiological tau monomers in the cytosol. Furthermore, we highlight some of the key mechanistic gaps of the field, which urgently need to be addressed to expand our understanding of tau propagation and lead to the identification of new therapeutic strategies for tauopathies.
Collapse
Affiliation(s)
- Samantha De La-Rocque
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edoardo Moretto
- UK Dementia Research Institute, University College London, London, UK
| | - Ioana Butnaru
- UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, UK.,Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
44
|
Blockade of the M1 muscarinic acetylcholine receptors impairs eyeblink serial feature-positive discrimination learning in mice. PLoS One 2020; 15:e0237451. [PMID: 32790748 PMCID: PMC7425847 DOI: 10.1371/journal.pone.0237451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/27/2020] [Indexed: 11/19/2022] Open
Abstract
The serial feature-positive discrimination task requires the subjects to respond differentially to the identical stimulus depending on the temporal context given by a preceding cue stimulus. In the present study, we examined the involvement of the M1 muscarinic acetylcholine receptors using a selective M1 antagonist VU0255035 in the serial feature-positive discrimination task of eyeblink conditioning in mice. In this task, mice received a 2-s light stimulus as the conditional cue 5 or 6 s before the presentation of a 350-ms tone conditioned stimulus (CS) paired with a 100-ms peri-orbital electrical shock (cued trials), while they did not receive the cue before the presentation of the CS alone (non-cued trials). Each day mice randomly received 30 cued and 30 non-cued trials. We found that VU0255035 impaired acquisition of the conditional discrimination as well as the overall acquisition of the conditioned response (CR) and diminished the difference in onset latency of the CR between the cued and non-cued trials. VU0255035 administration to the control mice after sufficient learning did not impair the pre-acquired conditional discrimination or the CR expression itself. These effects of VU0255035 were almost similar to those with the scopolamine in our previous study, suggesting that among the several types of muscarinic acetylcholine receptors, the M1 receptors may play an important role in the acquisition of the conditional discrimination memory but not in mediating the discrimination itself after the memory had formed in the eyeblink serial feature-positive discrimination learning.
Collapse
|
45
|
Tong L, Li W, Lo MMC, Gao X, Wai JMC, Rudd M, Tellers D, Joshi A, Zeng Z, Miller P, Salinas C, Riffel K, Haley H, Purcell M, Holahan M, Gantert L, Schubert JW, Jones K, Mulhearn J, Egbertson M, Meng Z, Hanney B, Gomez R, Harrison ST, McQuade P, Bueters T, Uslaner J, Morrow J, Thomson F, Kong J, Liao J, Selyutin O, Bao J, Hastings NB, Agrawal S, Magliaro BC, Monsma FJ, Smith MD, Risso S, Hesk D, Hostetler E, Mazzola R. Discovery of [ 11C]MK-6884: A Positron Emission Tomography (PET) Imaging Agent for the Study of M4Muscarinic Receptor Positive Allosteric Modulators (PAMs) in Neurodegenerative Diseases. J Med Chem 2020; 63:2411-2425. [PMID: 32101422 DOI: 10.1021/acs.jmedchem.9b01406] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The measurement of receptor occupancy (RO) using positron emission tomography (PET) has been instrumental in guiding discovery and development of CNS directed therapeutics. We and others have investigated muscarinic acetylcholine receptor 4 (M4) positive allosteric modulators (PAMs) for the treatment of symptoms associated with neuropsychiatric disorders. In this article, we describe the synthesis, in vitro, and in vivo characterization of a series of central pyridine-related M4 PAMs that can be conveniently radiolabeled with carbon-11 as PET tracers for the in vivo imaging of an allosteric binding site of the M4 receptor. We first demonstrated its feasibility by mapping the receptor distribution in mouse brain and confirming that a lead molecule 1 binds selectively to the receptor only in the presence of the orthosteric agonist carbachol. Through a competitive binding affinity assay and a number of physiochemical properties filters, several related compounds were identified as candidates for in vivo evaluation. These candidates were then radiolabeled with 11C and studied in vivo in rhesus monkeys. This research eventually led to the discovery of the clinical radiotracer candidate [11C]MK-6884.
Collapse
Affiliation(s)
- Ling Tong
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Wenping Li
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michael Man-Chu Lo
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaolei Gao
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jenny Miu-Chen Wai
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michael Rudd
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - David Tellers
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Aniket Joshi
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Zhizhen Zeng
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Patricia Miller
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Cristian Salinas
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Kerry Riffel
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Hyking Haley
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Mona Purcell
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Marie Holahan
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Liza Gantert
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jeffrey W Schubert
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Kristen Jones
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - James Mulhearn
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Melissa Egbertson
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Zhaoyang Meng
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Barbara Hanney
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Robert Gomez
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Scott T Harrison
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Paul McQuade
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Tjerk Bueters
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jason Uslaner
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - John Morrow
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Fiona Thomson
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jongrock Kong
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Jing Liao
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Oleg Selyutin
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jianming Bao
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Nicholas B Hastings
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Sony Agrawal
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Brian C Magliaro
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Frederick J Monsma
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michelle D Smith
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Stefania Risso
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - David Hesk
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Eric Hostetler
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Robert Mazzola
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
46
|
Scarpa M, Hesse S, Bradley SJ. M1 muscarinic acetylcholine receptors: A therapeutic strategy for symptomatic and disease-modifying effects in Alzheimer's disease? ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 88:277-310. [PMID: 32416870 DOI: 10.1016/bs.apha.2019.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The M1 muscarinic acetylcholine receptor (mAChR) plays a crucial role in learning and memory processes and has long been identified as a promising therapeutic target for the improvement of cognitive decline in Alzheimer's disease (AD). As such, clinical trials with xanomeline, a mAChR orthosteric agonist, showed an improvement in cognitive and behavioral symptoms associated with AD. Despite this, the clinical utility of xanomeline was hampered by a lack of M1 receptor selectivity and adverse cholinergic responses attributed to activation of peripheral M2 and M3 mAChRs. More recently, efforts have focused on developing more selective M1 compounds via targeting the less-conserved allosteric binding pockets. As such, positive allosteric modulators (PAMs) have emerged as an exciting strategy to achieve exquisite selectivity for the M1 mAChR in order to deliver improvements in cognitive function in AD. Furthermore, over recent years it has become increasingly apparent that M1 therapeutics may also offer disease-modifying effects in AD, due to the modulation of pathogenic amyloid processing. This article will review the progress made in the development of M1 selective ligands for the treatment of cognitive decline in AD, and will discuss the current evidence that selective targeting of the M1 mAChR could also have the potential to modify AD progression.
Collapse
Affiliation(s)
- Miriam Scarpa
- 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
| | - Sarah Hesse
- 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
| | - 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.
| |
Collapse
|
47
|
Bertrand D, Wallace TL. A Review of the Cholinergic System and Therapeutic Approaches to Treat Brain Disorders. Curr Top Behav Neurosci 2020; 45:1-28. [PMID: 32451956 DOI: 10.1007/7854_2020_141] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Since its identification over a hundred years ago, the neurotransmitter acetylcholine (ACh) has proven to play an essential role in supporting many diverse functions. Some well-characterized functions include: chemical transmission at the neuromuscular junction; autonomic function in the peripheral nervous system; and, sustained attention, sleep/wake regulation, and learning and memory within the central nervous system. Within the brain, major cholinergic projection pathways from the basal forebrain and the brainstem support these centrally mediated processes, and dysregulation of the cholinergic system is implicated in cognitive decline associated with aging and dementias including Alzheimer's disease. ACh exerts its effects by binding to two different membrane-bound receptor classes: (1) G‑protein coupled muscarinic acetylcholine receptors (mAChRs), and (2) ligand-gated nicotinic acetylcholine receptors (nAChRs). These receptor systems are described in detail within this chapter along with discussion on the successes and failures of synthetic ligands designed to selectively target receptor subtypes for treating brain disorders. New molecular approaches and advances in our understanding of the target biology combined with opportunities to re-purpose existing cholinergic drugs for new indications continue to highlight the exciting opportunities for modulating this system for therapeutic purposes.
Collapse
|
48
|
Zenko D, Thompson D, Hislop JN. Endocytic sorting and downregulation of the M2 acetylcholine receptor is regulated by ubiquitin and the ESCRT complex. Neuropharmacology 2020; 162:107828. [PMID: 31654703 DOI: 10.1016/j.neuropharm.2019.107828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 01/14/2023]
Abstract
Cholinergic dysfunction plays a critical role in a number of disease states, and the loss of functional muscarinic acetylcholine receptors plays a key role in disease pathogenesis. Therefore, preventing receptor downregulation would maintain functional receptor number, and be predicted to alleviate symptoms. However, the molecular mechanism(s) underlying muscarinic receptor downregulation are currently unknown. Here we demonstrate that the M2 muscarinic receptor undergoes rapid lysosomal proteolysis, and this lysosomal trafficking is facilitated by ubiquitination of the receptor. Importantly, we show that this trafficking is driven specifically by ESCRT mediated involution. Critically, we provide evidence that disruption of this process leads to a re-routing of the trafficking of the M2 receptor away from the lysosome and into recycling pathway, and eventually back to the plasma membrane. This study is the first to identify the process by which the M2 muscarinic acetylcholine receptor undergoes endocytic sorting, and critically reveals a regulatory checkpoint that represents a target to pharmacologically increase the number of functional muscarinic receptors within the central nervous system.
Collapse
Affiliation(s)
- Dmitry Zenko
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK
| | - Dawn Thompson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK
| | - James N Hislop
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
49
|
Russo M, Carrarini C, Dono F, Rispoli MG, Di Pietro M, Di Stefano V, Ferri L, Bonanni L, Sensi SL, Onofrj M. The Pharmacology of Visual Hallucinations in Synucleinopathies. Front Pharmacol 2019; 10:1379. [PMID: 31920635 PMCID: PMC6913661 DOI: 10.3389/fphar.2019.01379] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Visual hallucinations (VH) are commonly found in the course of synucleinopathies like Parkinson's disease and dementia with Lewy bodies. The incidence of VH in these conditions is so high that the absence of VH in the course of the disease should raise questions about the diagnosis. VH may take the form of early and simple phenomena or appear with late and complex presentations that include hallucinatory production and delusions. VH are an unmet treatment need. The review analyzes the past and recent hypotheses that are related to the underlying mechanisms of VH and then discusses their pharmacological modulation. Recent models for VH have been centered on the role played by the decoupling of the default mode network (DMN) when is released from the control of the fronto-parietal and salience networks. According to the proposed model, the process results in the perception of priors that are stored in the unconscious memory and the uncontrolled emergence of intrinsic narrative produced by the DMN. This DMN activity is triggered by the altered functioning of the thalamus and involves the dysregulated activity of the brain neurotransmitters. Historically, dopamine has been indicated as a major driver for the production of VH in synucleinopathies. In that context, nigrostriatal dysfunctions have been associated with the VH onset. The efficacy of antipsychotic compounds in VH treatment has further supported the notion of major involvement of dopamine in the production of the hallucinatory phenomena. However, more recent studies and growing evidence are also pointing toward an important role played by serotonergic and cholinergic dysfunctions. In that respect, in vivo and post-mortem studies have now proved that serotonergic impairment is often an early event in synucleinopathies. The prominent cholinergic impairment in DLB is also well established. Finally, glutamatergic and gamma aminobutyric acid (GABA)ergic modulations and changes in the overall balance between excitatory and inhibitory signaling are also contributing factors. The review provides an extensive overview of the pharmacology of VH and offers an up to date analysis of treatment options.
Collapse
Affiliation(s)
- Mirella Russo
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Claudia Carrarini
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Fedele Dono
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Marianna Gabriella Rispoli
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Martina Di Pietro
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Vincenzo Di Stefano
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Ferri
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Bonanni
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Stefano Luca Sensi
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
- Behavioral Neurology and Molecular Neurology Units, Center of Excellence on Aging and Translational Medicine—CeSI-MeT, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
- Departments of Neurology and Pharmacology, Institute for Mind Impairments and Neurological Disorders—iMIND, University of California, Irvine, Irvine, CA, United States
| | - Marco Onofrj
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
50
|
Alrefaie Z, Moustafa I. Vitamin D3 favorable outcome on recognition memory and prefrontal cortex expression of choline acetyltransferase and acetylcholinesterase in experimental model of chronic high-fat feeding. Int J Neurosci 2019; 130:262-269. [PMID: 31544572 DOI: 10.1080/00207454.2019.1671839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aim of the study: High-fat diet (HFD) consumption and insufficient vitamin D levels are globally increasing phenomena. The present study assessed the effect of chronic HFD feeding with and without vitamin D supplementation on recognition memory and prefrontal cortex expression of choline acetyltransferase (CAT) and acetylcholinesterase (Achase).Materials and methods: Forty male Wistar rats were subjected to four dietary regimens (n = 10); control diet (10% fat), control + vitamin D3, high-fat diet (HFD 45% fat) and HFD + vitamin D3 for 6 months. Rats were tested for the novel object recognition test, and their prefrontal cortices were assessed for expression of CAT and Achase.Results: Recognition memory was impaired in HFD-fed rats compared to control rats as evidenced by significantly decreased discrimination index in the novel object recognition test. Moreover, CAT expression was significantly decreased while Achase expression was significantly increased in the prefrontal cortex of HFD-fed rats. Vitamin D3 supplementation with HFD significantly increased the exploration of the novel object and the discrimination index and attenuated the alterations in the prefrontal cortex CAT and Achase expression.Conclusions: The present findings support the potential effect of vitamin D on recognition memory and cholinergic transmission in the prefrontal cortex and add to the pathophysiology of HFD consumption.
Collapse
Affiliation(s)
- Zienab Alrefaie
- Faculty of Medicine, Physiology Department, Cairo University, Cairo, Egypt
| | | |
Collapse
|