1
|
Bruszt N, Bali ZK, Nagy LV, Bodó K, Engelmann P, Hernádi I. Synergistic effects of memantine and alpha7 nicotinic acetylcholine receptor agonist PHA-543613 to improve memory of aged rats. Int J Neuropsychopharmacol 2025; 28:pyaf014. [PMID: 39985181 PMCID: PMC11929953 DOI: 10.1093/ijnp/pyaf014] [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: 08/02/2024] [Accepted: 02/20/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Combination treatments based on pharmacological interactions at α7 nicotinic acetylcholine receptors (nAChRs) are promising therapeutic approaches for neurocognitive disorders. METHODS Here, we tested the cognitive efficacy of combinations of memantine with an α7 nAChR-selective agonist (PHA-543613) in naturally aged rats. Age-related changes in the expression of some key genes and proteins were also measured using quantitative polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA). RESULTS Aged rats showed marked cognitive decline in the novel object recognition test, and they also exhibited cholinergic changes such as mRNA upregulation of α7 nAChRs. Upregulation of interleukin-1β, macrophage inflammatory protein 1α, CX3CL1, intercellular adhesion molecule 1, and ciliary neurotrophic factor mRNA was also detected in aged rats. Combination treatment of memantine and PHA-543613 successfully alleviated the age-related decline of recognition memory of rats by exceeding the effects of the corresponding monotreatments. CONCLUSIONS Results indicate a positive interaction between memantine and PHA-543613, which also reflects a putative role of α7 nAChRs in the cognitive enhancer effects of memantine. These findings may facilitate the development of combination therapies for age-related neurocognitive disorders.
Collapse
Affiliation(s)
- Nóra Bruszt
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Grastyán Endre Translational Research Centre, University of Pécs, 4 Pécs, Hungary
| | - Zsolt Kristóf Bali
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Grastyán Endre Translational Research Centre, University of Pécs, 4 Pécs, Hungary
| | - Lili Veronika Nagy
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Department of Neurobiology, Institute of Biology, University of Pécs, Pécs, Hungary
| | - Kornélia Bodó
- Department of Immunology and Biotechnology, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Engelmann
- Department of Immunology and Biotechnology, Medical School, University of Pécs, Pécs, Hungary
| | - István Hernádi
- Translational Neuroscience Research Group, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
- Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
- Grastyán Endre Translational Research Centre, University of Pécs, 4 Pécs, Hungary
- Department of Neurobiology, Institute of Biology, University of Pécs, Pécs, Hungary
| |
Collapse
|
2
|
Lee R, Kim G, Black ER, Kim S. Co-activation of selective nicotinic acetylcholine receptor subtypes is required to reverse hippocampal network dysfunction, fear memory loss, and amyloid pathology in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.08.602576. [PMID: 39026693 PMCID: PMC11257460 DOI: 10.1101/2024.07.08.602576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with no known cause and cure. Research suggests that a reduction of GABAergic inhibitory interneurons' activity in the hippocampus by beta-amyloid peptide (Aβ) is a crucial trigger for amyloid pathology and cognitive impairment in AD via hyperexcitability. Therefore, enhancing hippocampal inhibition is thought to be protective against AD. However, hippocampal inhibitory cells are highly diverse, and these distinct interneuron subtypes differentially regulate hippocampal inhibitory circuits and cognitive processes. Moreover, Aβ unlikely affects all subtypes of inhibitory interneurons in the hippocampus equally. Hence, identifying the affected interneuron subtypes in AD to enhance hippocampal inhibition optimally is conceptually and practically challenging. We have previously found that Aβ selectively binds to two of the three major hippocampal nicotinic acetylcholine receptor (nAChR) subtypes, α7- and α4β2-nAChRs, but not α3β4-nAChRs, and inhibits these two receptors in cultured hippocampal inhibitory interneurons to decrease their activity, leading to hyperexcitation in excitatory neurons. We have also revealed that co-activation of α7- and α4β2-nAChRs is required to reverse the Aβ-induced adverse effects in hippocampal excitatory neurons. Here, we discover that α7- and α4β2-nAChRs predominantly control the nicotinic cholinergic signaling and neuronal activity in hippocampal parvalbumin-positive (PV+) and somatostatin-positive (SST+) inhibitory interneurons, respectively. Furthermore, we reveal that co-activation of these receptors is necessary to reverse hippocampal network dysfunction, amyloid pathology, and fear memory loss in the amyloid pathology model mice. This suggests that co-activation of PV+ and SST+ cells via stimulating α7- and α4β2-nAChRs together is a novel strategy for neuroprotection against AD.
Collapse
|
3
|
Guan Z. Alterations in Neuronal Nicotinic Acetylcholine Receptors in the Pathogenesis of Various Cognitive Impairments. CNS Neurosci Ther 2024; 30:e70069. [PMID: 39370620 PMCID: PMC11456617 DOI: 10.1111/cns.70069] [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: 03/16/2024] [Revised: 09/02/2024] [Accepted: 09/15/2024] [Indexed: 10/08/2024] Open
Abstract
Cognitive impairment is a typical symptom of both neurodegenerative and certain other diseases. In connection with these different pathologies, the etiology and neurological and metabolic changes associated with cognitive impairment must differ. Until these characteristics and differences are understood in greater detail, pharmacological treatment of the different forms of cognitive impairment remains suboptimal. Neurotransmitter receptors, including neuronal nicotinic acetylcholine receptors (nAChRs), dopamine receptors, and glutamine receptors, play key roles in the functions and metabolisms of the brain. Among these, the role of nAChRs in the development of cognitive impairment has attracted more and more attention. The present review summarizes what is presently known concerning the structure, distribution, metabolism, and function of nAChRs, as well as their involvement in major cognitive disorders such as Alzheimer's disease, Parkinson's disease, vascular dementia, schizophrenia, and diabetes mellitus. As will be discussed, the relevant scientific literature reveals clearly that the α4β2 and α7 nAChR subtypes and/or subunits of the receptors play major roles in maintaining cognitive function and in neuroprotection of the brain. Accordingly, focusing on these as targets of drug therapy can be expected to lead to breakthroughs in the treatment of cognitive disorders such as AD and schizophrenia.
Collapse
Affiliation(s)
- Zhi‐Zhong Guan
- Department of PathologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangP.R. China
- Key Laboratory of Endemic and Ethnic DiseasesGuizhou Medical University, Ministry of Education and Provincial Key Laboratory of Medical Molecular BiologyGuiyangP.R. China
| |
Collapse
|
4
|
Chen F, Zhang Z, Zhang H, Guo P, Feng J, Shen H, Liu X. Activation of α7 Nicotinic Acetylcholine Receptor Improves Muscle Endurance by Upregulating Orosomucoid Expression and Glycogen Content in Mice. J Cell Biochem 2024; 125:e30630. [PMID: 39014907 DOI: 10.1002/jcb.30630] [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: 01/24/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
There are presently no acknowledged therapeutic targets or official drugs for the treatment of muscle fatigue. The alpha7 nicotinic acetylcholine receptor (α7nAChR) is expressed in skeletal muscle, with an unknown role in muscle endurance. Here, we try to explore whether α7nAChR could act as a potential therapeutic target for the treatment of muscle fatigue. Results showed that nicotine and PNU-282987 (PNU), as nonspecific and specific agonists of α7nAChR, respectively, could both significantly increase C57BL6/J mice treadmill-running time in a time- and dose-dependent manner. The improvement effect of PNU on running time and ex vivo muscle fatigue index disappeared when α7nAChR deletion. RNA sequencing revealed that the differential mRNAs affected by PNU were enriched in glycolysis/gluconeogenesis signaling pathways. Further studies found that PNU treatment significantly elevates glycogen content and ATP level in the muscle tissues of α7nAChR+/+ mice but not α7nAChR-/- mice. α7nAChR activation specifically increased endogenous glycogen-targeting protein orosomucoid (ORM) expression both in vivo skeletal muscle tissues and in vitro C2C12 skeletal muscle cells. In ORM1 deficient mice, the positive effects of PNU on running time, glycogen and ATP content, as well as muscle fatigue index, were abolished. Therefore, the activation of α7nAChR could enhance muscle endurance via elevating endogenous anti-fatigue protein ORM and might act as a promising therapeutic strategy for the treatment of muscle fatigue.
Collapse
Affiliation(s)
- Fei Chen
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Zhen Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
- Department of Nutrition and Food Hygiene, Second Military Medical University, Shanghai, China
| | - Huimin Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Pengyue Guo
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Jiayi Feng
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Hui Shen
- Department of Nutrition and Food Hygiene, Second Military Medical University, Shanghai, China
| | - Xia Liu
- Department of Clinical Pharmacy, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
5
|
Lee B, Yu MS, Song JG, Lee HM, Kim HW, Na D. Corydalis ternata Nakai Alleviates Cognitive Decline in Alzheimer's Disease by Reducing β-Amyloid and Neuroinflammation. Rejuvenation Res 2024; 27:87-101. [PMID: 38545769 DOI: 10.1089/rej.2023.0069] [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] [Indexed: 04/25/2024] Open
Abstract
Recently, natural herbs have gained increasing attention owing to their comparatively low toxicity levels and the abundance of historical medical documentation regarding their use. Nevertheless, owing to a lack of knowledge regarding these herbs and their compounds, attempts to find those that could be beneficial for treating diseases have often been ad hoc; thus, there is now a growing demand for an in silico method to identify beneficial herbs. In this study, we present a computational approach for identifying natural herbs specifically effective in treating cognitive decline in Alzheimer's disease (AD) sufferers, which analyzes the similarities between herbal compounds and known drugs targeting AD-related proteins. Our in silico method suggests that Corydalis ternata can improve cognitive decline in AD sufferers. Behavioral tests with an AD mouse model for the confirmation of the in silico prediction reveals that C. ternata significantly alleviated the cognitive decline (memory and motor functions) caused by neurodegeneration. Further pathology analyses reveal that C. ternata decreases the level of Aβ plaques, reduces neuroinflammation, and promotes autophagy flux, and thus C. ternata can be clinically effective for preventing mild cognitive impairment during the early stages of AD. These findings highlight the potential utility of our in silico method and the potential clinical application of the identified natural herb in treating and preventing AD.
Collapse
Affiliation(s)
- Bomi Lee
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jae Gwang Song
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung Wook Kim
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Wang H, Yan X, Zhang Y, Wang P, Li J, Zhang X. Mitophagy in Alzheimer's Disease: A Bibliometric Analysis from 2007 to 2022. J Alzheimers Dis Rep 2024; 8:101-128. [PMID: 38312534 PMCID: PMC10836605 DOI: 10.3233/adr-230139] [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: 10/02/2023] [Accepted: 12/15/2023] [Indexed: 02/06/2024] Open
Abstract
Background The investigation of mitophagy in Alzheimer's disease (AD) remains relatively underexplored in bibliometric analysis. Objective To delve into the progress of mitophagy, offering a comprehensive overview of research trends and frontiers for researchers. Methods Basic bibliometric information, targets, and target-drug-clinical trial-disease extracted from publications identified in the Web of Science Core Collection from 2007 to 2022 were assessed using bibliometric software. Results The study encompassed 5,146 publications, displaying a consistent 16-year upward trajectory. The United States emerged as the foremost contributor in publications, with the Journal of Alzheimer's Disease being the most prolific journal. P. Hemachandra Reddy, George Perry, and Xiongwei Zhu are the top 3 most prolific authors. PINK1 and Parkin exhibited an upward trend in the last 6 years. Keywords (e.g., insulin, aging, epilepsy, tauopathy, and mitochondrial quality control) have recently emerged as focal points of interest within the past 3 years. "Mitochondrial dysfunction" is among the top terms in disease clustering. The top 10 drugs/molecules (e.g., curcumin, insulin, and melatonin) were summarized, accompanied by their clinical trials and related targets. Conclusions This study presents a comprehensive overview of the mitophagy research landscape in AD over the past 16 years, underscoring mitophagy as an emerging molecular mechanism and a crucial focal point for potential drug in AD. This study pioneers the inclusion of targets and their correlations with drugs, clinical trials, and diseases in bibliometric analysis, providing valuable insights and inspiration for scholars and readers of JADR interested in understanding the potential mechanisms and clinical trials in AD.
Collapse
Affiliation(s)
- Hongqi Wang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaodong Yan
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yiming Zhang
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Peifu Wang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
| | - Jilai Li
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
| | - Xia Zhang
- Department of Neurology, Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, China
- Department of Anatomy, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Singh S, Agrawal N, Goyal A. Role of Alpha-7-Nicotinic Acetylcholine Receptor in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:384-394. [PMID: 37366362 DOI: 10.2174/1871527322666230627123426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder affecting millions worldwide. One of the leading hypotheses for the underlying cause of AD is a reduction in nicotinic receptor levels in the brain. Among the nicotinic receptors, the alpha-7-nicotinic acetylcholine receptor (α7nAChR) has received particular attention due to its involvement in cognitive function.α7nAChR is a ligand-gated ion channel that is primarily found in the hippocampus and prefrontal cortex, areas of the brain responsible for learning, memory, and attention. Studies have shown that α7nAChR dysfunction is a key contributor to the pathogenesis of AD. The receptor is involved in regulating amyloidbeta (Aβ) production, a hallmark of AD pathology. Many drugs have been investigated as α7nAChR agonists or allosteric modulators to improve cognitive deficits in AD. Clinical studies have shown promising results with α7nAChR agonists, including improved memory and cognitive function. Although several studies have shown the significance of the α7 nAChR in AD, little is known about its function in AD pathogenesis. As a result, in this review, we have outlined the basic information of the α7 nAChR's structure, functions, cellular responses to its activation, and its role in AD's pathogenesis.
Collapse
Affiliation(s)
- Sushma Singh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
- Pharmacy College, Azamgarh- 276128, UP, India
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| |
Collapse
|
8
|
García AP, Gaydou L, Pérez E, Barrantes FJ. Insulin resistance induced by long-term hyperinsulinemia abolishes the effects of acute insulin exposure on cell-surface nicotinic acetylcholine receptor levels and actin cytoskeleton morphology. Biochem Biophys Res Commun 2023; 685:149165. [PMID: 37922786 DOI: 10.1016/j.bbrc.2023.149165] [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: 10/04/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
Using CHO-K1/A5 cells, a clonal cell line that robustly expresses adult muscle-type nicotinic acetylcholine receptor (nAChR), we explored whether insulin resistance in these mammalian cells affects cell-surface expression of the nAChR, its endocytic internalization, and actin cytoskeleton integrity. Acute nanomolar insulin stimulation resulted in a slow increase in nAChR cell-surface levels, reaching maximum levels at ∼1 h. Long periods of insulin incubation caused CHO-K1/A5 cells to become insulin resistant, as previously observed with several other cell types. Furthermore, long-term insulin treatment abolished the effects of short-term insulin exposure on cell-surface nAChR levels, suggestive of a desensitization phenomenon. It also affected the kinetics of ligand-induced nAChR internalization. Since the integrity of the cortical actin cytoskeleton affects nAChR endocytosis, we also studied the effects of long-term insulin treatment on this meshwork. We found that it significantly affected the cortical actin morphology of CHO-K1/A5 cells and the response of the actin cytoskeleton to a subsequent short-term insulin stimulus. Overall, the present results show for the first time the effects of insulin signaling on cell-surface nAChR expression and actin cytoskeleton-associated internalization.
Collapse
Affiliation(s)
- Ana Paula García
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina; Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina
| | - Luisa Gaydou
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral-CONICET, Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Eugenia Pérez
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina
| | - Francisco J Barrantes
- Laboratorio de Neurobiología Molecular, Instituto de Investigaciones Biomédicas (BIOMED) UCA-CONICET, Facultad de Ciencias Médicas, Universidad Católica de Argentina, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Esaki H, Deyama S, Izumi S, Katsura A, Nishikawa K, Nishitani N, Kaneda K. Varenicline enhances recognition memory via α7 nicotinic acetylcholine receptors in the medial prefrontal cortex in male mice. Neuropharmacology 2023; 239:109672. [PMID: 37506875 DOI: 10.1016/j.neuropharm.2023.109672] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Previous studies postulated that chronic administration of varenicline, a partial and full agonist at α4β2 and α7 nicotinic acetylcholine receptors (nAChRs), respectively, enhances recognition memory. However, whether its acute administration is effective, on which brain region(s) it acts, and in what signaling it is involved, remain unknown. To address these issues, we conducted a novel object recognition test using male C57BL/6J mice, focusing on the medial prefrontal cortex (mPFC), a brain region associated with nicotine-induced enhancement of recognition memory. Systemic administration of varenicline before the training dose-dependently enhanced recognition memory. Intra-mPFC varenicline infusion also enhanced recognition memory, and this enhancement was blocked by intra-mPFC co-infusion of a selective α7, but not α4β2, nAChR antagonist. Consistent with this, intra-mPFC infusion of a selective α7 nAChR agonist augmented object recognition memory. Furthermore, intra-mPFC co-infusion of U-73122, a phospholipase C (PLC) inhibitor, or 2-aminoethoxydiphenylborane (2-APB), an inositol trisphosphate (IP3) receptor inhibitor, suppressed the varenicline-induced memory enhancement, suggesting that α7 nAChRs may also act as Gq-coupled metabotropic receptors. Additionally, whole-cell recordings from mPFC layer V pyramidal neurons in vitro revealed that varenicline significantly increased the summation of evoked excitatory postsynaptic potentials, and this effect was suppressed by U-73122 or 2-APB. These findings suggest that varenicline might acutely enhance recognition memory via mPFC α7 nAChR stimulation, followed by mPFC neuronal excitation, which is mediated by the activation of PLC and IP3 receptor signaling. Our study provides evidence supporting the potential repositioning of varenicline as a treatment for cognitive impairment.
Collapse
Affiliation(s)
- Hirohito Esaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Shoma Izumi
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ayano Katsura
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Keisuke Nishikawa
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
10
|
Ma W, Si T, Wang Z, Wen P, Zhu Z, Liu Q, Wang J, Xu F, Li Q. Astrocytic α4-containing nAChR signaling in the hippocampus governs the formation of temporal association memory. Cell Rep 2023; 42:112674. [PMID: 37352098 DOI: 10.1016/j.celrep.2023.112674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/24/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023] Open
Abstract
Everyday episodic memories involve linking together related events that are temporally separated. However, the mechanisms of forming this temporal association have remained unclear. Here, using astrocyte-specific manipulations, we show that potentiating astrocyte Ca2+ signaling in the hippocampal cornu ammonis 1 (CA1) enhances the strength of such temporal association, in parallel with long-term potentiation (LTP) enhancement of temporoammonic pathway to CA1, whereas attenuation of astrocyte Ca2+ signaling has the opposite effect. Moreover, we identify that these effects are mediated by astrocytic α4 subunit-containing nicotinic acetylcholine receptors (α4-nAChRs) via mechanisms involving NMDAR co-agonist supply. Finally, astrocytic α4-nAChRs underlie the cognitive enhancer nicotine's physiological effects. Together, these findings highlight the importance of astrocyte Ca2+ signaling in cognitive behavior and reveal a mechanism in governing the temporal association of episodic memory formation that operates through α4-nAChRs on hippocampal astrocytes.
Collapse
Affiliation(s)
- Wenyu Ma
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tengxiao Si
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zan Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengjie Wen
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhenxiang Zhu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Liu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; Shenzhen Key Laboratory of Viral Vectors for Biomedicine, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qin Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
11
|
Younger DS. Autonomic failure: Clinicopathologic, physiologic, and genetic aspects. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:55-102. [PMID: 37562886 DOI: 10.1016/b978-0-323-98818-6.00020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Over the past century, generations of neuroscientists, pathologists, and clinicians have elucidated the underlying causes of autonomic failure found in neurodegenerative, inherited, and antibody-mediated autoimmune disorders, each with pathognomonic clinicopathologic features. Autonomic failure affects central autonomic nervous system components in the α-synucleinopathy, multiple system atrophy, characterized clinically by levodopa-unresponsive parkinsonism or cerebellar ataxia, and pathologically by argyrophilic glial cytoplasmic inclusions (GCIs). Two other central neurodegenerative disorders, pure autonomic failure characterized clinically by deficits in norepinephrine synthesis and release from peripheral sympathetic nerve terminals; and Parkinson's disease, with early and widespread autonomic deficits independent of the loss of striatal dopamine terminals, both express Lewy pathology. The rare congenital disorder, hereditary sensory, and autonomic neuropathy type III (or Riley-Day, familial dysautonomia) causes life-threatening autonomic failure due to a genetic mutation that results in loss of functioning baroreceptors, effectively separating afferent mechanosensing neurons from the brain. Autoimmune autonomic ganglionopathy caused by autoantibodies targeting ganglionic α3-acetylcholine receptors instead presents with subacute isolated autonomic failure affecting sympathetic, parasympathetic, and enteric nervous system function in various combinations. This chapter is an overview of these major autonomic disorders with an emphasis on their historical background, neuropathological features, etiopathogenesis, diagnosis, and treatment.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
12
|
Xia Y, Wu Q, Mak S, Liu EYL, Zheng BZY, Dong TTX, Pi R, Tsim KWK. Regulation of acetylcholinesterase during the lipopolysaccharide-induced inflammatory responses in microglial cells. FASEB J 2022; 36:e22189. [PMID: 35129858 DOI: 10.1096/fj.202101302rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 01/06/2022] [Accepted: 01/18/2022] [Indexed: 01/04/2023]
Abstract
The non-classical function of acetylcholine (ACh) has been reported in neuroinflammation that represents the modulating factor in immune responses via activation of α7 nicotinic acetylcholine receptor (α7 nAChR), i.e., a cholinergic anti-inflammatory pathway (CAP). Acetylcholinesterase (AChE), an enzyme for ACh hydrolysis, has been proposed to have a non-classical function in immune cells. However, the involvement of AChE in neuroinflammation is unclear. Here, cultured BV2 cell, a microglial cell line, and primary microglia from rats were treated with lipopolysaccharide (LPS) to induce inflammation and to explore the regulation of AChE during this process. The expression profiles of AChE, α7 nAChR, and choline acetyltransferase (ChAT) were revealed in BV2 cells. The expression of AChE (G4 form) was induced significantly in LPS-treated BV2 cells: the induction was triggered by NF-κB and cAMP signaling. Moreover, ACh or α7 nAChR agonist suppressed the LPS-induced production of pro-inflammatory cytokines, as well as the phagocytosis of microglia, by activating α7 nAChR and followed by the regulation of NF-κB and CREB signaling. The ACh-induced suppression of inflammation was abolished in AChE overexpressed cells, but did not show a significant change in AChE mutant (enzymatic activity knockout) transfected cells. These results indicate that the neuroinflammation-regulated function of AChE may be mediated by controlling the ACh level in the brain system.
Collapse
Affiliation(s)
- Yingjie Xia
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qiyun Wu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Shinghung Mak
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Etta Y L Liu
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Brody Z Y Zheng
- Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Tina T X Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Rongbiao Pi
- School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Karl W K Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, SRI, The Hong Kong University of Science and Technology, Shenzhen, China.,Division of Life Science, Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
13
|
Richter N, David LS, Grothe MJ, Teipel S, Dietlein M, Tittgemeyer M, Neumaier B, Fink GR, Onur OA, Kukolja J. Age and Anterior Basal Forebrain Volume Predict the Cholinergic Deficit in Patients with Mild Cognitive Impairment due to Alzheimer’s Disease. J Alzheimers Dis 2022; 86:425-440. [DOI: 10.3233/jad-210261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background: Early and severe neuronal loss in the cholinergic basal forebrain is observed in Alzheimer’s disease (AD). To date, cholinomimetics play a central role in the symptomatic treatment of AD dementia. Although basic research indicates that a cholinergic deficit is present in AD before dementia, the efficacy of cholinomimetics in mild cognitive impairment (MCI) remains controversial. Predictors of cholinergic impairment could guide individualized therapy. Objective: To investigate if the extent of the cholinergic deficit, measured using positron emission tomography (PET) and the tracer 11C-N-methyl-4-piperidyl acetate (MP4A), could be predicted from the volume of cholinergic basal forebrain nuclei in non-demented AD patients. Methods: Seventeen patients with a high likelihood of MCI due to AD and 18 age-matched cognitively healthy adults underwent MRI-scanning. Basal forebrain volume was assessed using voxel-based morphometry and a cytoarchitectonic atlas of cholinergic nuclei. Cortical acetylcholinesterase (AChE) activity was measured using MP4A-PET. Results: Cortical AChE activity and nucleus basalis of Meynert (Ch4 area) volume were significantly decreased in MCI. The extent of the cholinergic deficit varied considerably across patients. Greater volumes of anterior basal forebrain nuclei (Ch1/2 area) and younger age (Spearman’s rho (17) = –0.596, 95% -CI [–0.905, –0.119] and 0.593, 95% -CI [0.092, 0.863])) were associated with a greater cholinergic deficit. Conclusion: Data suggest that less atrophy of the Ch1/2 area and younger age are associated with a more significant cholinergic deficit in MCI due to AD. Further investigations are warranted to determine if the individual response to cholinomimetics can be inferred from these measures.
Collapse
Affiliation(s)
- Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Lara-Sophia David
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Movement Disorders Group, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Marc Tittgemeyer
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
| | - Bernd Neumaier
- Max-Planck-Institute for Metabolism Research, Cologne, Cologne, Germany
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, Jülich, Germany
- Institute for Radiochemistry and Experimental Molecular Imaging, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
| | - Juraj Kukolja
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, Jülich, Germany
- Department of Neurology, Medical Faculty and University Hospital of Cologne, Cologne, Germany
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, Wuppertal, Germany
- Faculty of Health, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
14
|
Mukherjee I, Singh UK. Exploring a variance decomposition approach integrated with the Monte Carlo method to evaluate groundwater fluoride exposure on the residents of a typical fluorosis endemic semi-arid tract of India. ENVIRONMENTAL RESEARCH 2022; 203:111697. [PMID: 34358509 DOI: 10.1016/j.envres.2021.111697] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/07/2021] [Accepted: 07/11/2021] [Indexed: 06/13/2023]
Abstract
This study appraised the groundwater fluoride (F-) endemicity and the exposure levels under the Central Tendency Exposure (CTE) condition and the Reasonable Maximum Exposure (RME) condition on the residents of the semi-arid parts of the Birbhum district of Peninsular India using a Variance Decomposition (Sobol Sensitivity Indices) approach combined with Monte Carlo Simulations. The study finds the national scale drinking water standard limit for F- (1.5 mg L-1) is inappropriate for the present survey area where F- concentration in groundwater varied between 0.26 and 11.82 mg L-1 and ~54.5% of the samples (N = 400) exceeded this limit. Therefore, estimated the optimum F- concentration of 0.733 mg L-1 for the region using the method recommended by the World Health Organization (WHO) to calculate the optimum F- limit at a regional scale. The average value of F- concentrations for this region (1.71 mg L-1) is considerably higher than the estimated optimum concentration or even the maximum permissible limits recommended for the subtropical regions (0.5-0.7 mg L-1). The exposure analysis revealed the infants and children as potentially vulnerable populations compared to adolescents and adults of the study area for CTE and RME scenarios. The multi-exposure pathways indicated oral intake as the main exposure pathway whereas exposure through dermal contact was insignificant for the residents of all age groups of this region. Based on the first, second and total order Sobol Sensitivity Indices, F- concentration (C) in groundwater, the groundwater ingestion rate and their combined interaction are the greatest significant parameters for the oral exposure model whereas C and its interaction effects with the proportion of the skin surface area in contact with groundwater as the utmost sensitive variables for the dermal health risks assessment model. The present study insists the inhabitants to intake defluoridated groundwater.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Integrated Science Education and Research Centre (ISERC), Institute of Science, Visva- Bharati, Santiniketan-731235, Birbhum, West Bengal, India
| | - Umesh Kumar Singh
- Department of Environmental Science, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, 824236, Bihar, India.
| |
Collapse
|
15
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
16
|
Martinelli I, Tomassoni D, Roy P, Amenta F, Tayebati SK. Altered Brain Cholinergic and Synaptic Markers in Obese Zucker Rats. Cells 2021; 10:cells10102528. [PMID: 34685507 PMCID: PMC8534069 DOI: 10.3390/cells10102528] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/27/2022] Open
Abstract
The association between obesity and loss of cognitive performance has been recognized. Although there are data regarding the metabolic alterations in obese conditions and the development of neuroinflammation, no clear evidence concerning obesity-related cholinergic and synaptic impairments in the frontal cortex and hippocampus has been reported yet. Here, we investigate different cholinergic and synaptic markers in 12-, 16-, and 20-week-old obese Zucker rats (OZRs) compared with lean littermate rats (LZRs), using immunochemical and immunohistochemical analysis. Consequently, OZRs showed body weight gain, hypertension, and dysmetabolism. In 20-week-old OZRs, the reduction of vesicular acetylcholine transporter (VAChT) and alpha7 nicotinic acetylcholine receptors (α7nAChR) occurred both in the frontal cortex and in the hippocampus, suggesting a cognitive dysfunction due to obesity and aging. Among the muscarinic receptors analyzed, the level of expression of type 1 (mAChR1) was lower in the hippocampus of the older OZRs. Finally, we showed synaptic dysfunctions in OZRs, with a reduction of synaptophysin (SYP) and synaptic vesicle glycoprotein 2B (SV2B) in 20-week-old OZRs, both in the frontal cortex and in the hippocampus. Taken together, our data suggest specific alterations of cholinergic and synaptic markers that can be targeted to prevent cognitive deficits related to obesity and aging.
Collapse
Affiliation(s)
- Ilenia Martinelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (I.M.); (F.A.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Francesco Amenta
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (I.M.); (F.A.)
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (I.M.); (F.A.)
- Correspondence:
| |
Collapse
|
17
|
Eadaim A, Hahm ET, Justice ED, Tsunoda S. Cholinergic Synaptic Homeostasis Is Tuned by an NFAT-Mediated α7 nAChR-K v4/Shal Coupled Regulatory System. Cell Rep 2021; 32:108119. [PMID: 32905767 PMCID: PMC7521586 DOI: 10.1016/j.celrep.2020.108119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/23/2020] [Accepted: 08/17/2020] [Indexed: 11/26/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) involves compensatory mechanisms employed by neurons and circuits to preserve signaling when confronted with global changes in activity that may occur during physiological and pathological conditions. Cholinergic neurons, which are especially affected in some pathologies, have recently been shown to exhibit HSP mediated by nicotinic acetylcholine receptors (nAChRs). In Drosophila central neurons, pharmacological blockade of activity induces a homeostatic response mediated by the Drosophila α7 (Dα7) nAChR, which is tuned by a subsequent increase in expression of the voltage-dependent Kv4/Shal channel. Here, we show that an in vivo reduction of cholinergic signaling induces HSP mediated by Dα7 nAChRs, and this upregulation of Dα7 itself is sufficient to trigger transcriptional activation, mediated by nuclear factor of activated T cells (NFAT), of the Kv4/Shal gene, revealing a receptor-ion channel system coupled for homeostatic tuning in cholinergic neurons. Eadaim et al. show that in vivo reduction of cholinergic signaling in Drosophila neurons induces synaptic homeostasis mediated by Dα7 nAChRs. This upregulation of Dα7 induces Kv4/Shal gene expression mediated by nuclear factor of activated T cells (NFAT), revealing a receptor-ion channel system coupled for homeostatic tuning in cholinergic neurons.
Collapse
Affiliation(s)
- Abdunaser Eadaim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Eu-Teum Hahm
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Elizabeth D Justice
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
18
|
Cheng YJ, Lin CH, Lane HY. Involvement of Cholinergic, Adrenergic, and Glutamatergic Network Modulation with Cognitive Dysfunction in Alzheimer's Disease. Int J Mol Sci 2021; 22:2283. [PMID: 33668976 PMCID: PMC7956475 DOI: 10.3390/ijms22052283] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a progressive neurodegenerative disease. The number of AD cases has been rapidly growing worldwide. Several the related etiological hypotheses include atypical amyloid β (Aβ) deposition, neurofibrillary tangles of tau proteins inside neurons, disturbed neurotransmission, inflammation, and oxidative stress. During AD progression, aberrations in neurotransmission cause cognitive decline-the main symptom of AD. Here, we review the aberrant neurotransmission systems, including cholinergic, adrenergic, and glutamatergic network, and the interactions among these systems as they pertain to AD. We also discuss the key role of N-methyl-d-aspartate receptor (NMDAR) dysfunction in AD-associated cognitive impairment. Furthermore, we summarize the results of recent studies indicating that increasing glutamatergic neurotransmission through the alteration of NMDARs shows potential for treating cognitive decline in mild cognitive impairment or early stage AD. Future studies on the long-term efficiency of NMDA-enhancing strategies in the treatment of AD are warranted.
Collapse
Affiliation(s)
- Yu-Jung Cheng
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung 40402, Taiwan;
- Department of Rehabilitation, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chieh-Hsin Lin
- Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsien-Yuan Lane
- Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung 40402, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
19
|
Fahanik-Babaei J, Baluchnejadmojarad T, Roghani M. Differential Effect of Amyloid Beta1-40 on Short-term and Long-term Plasticity in Dentate Gyrus of a Rat Model of Alzheimer Disease. Basic Clin Neurosci 2021; 11:517-524. [PMID: 33613890 PMCID: PMC7878034 DOI: 10.32598/bcn.9.10.190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/10/2018] [Accepted: 07/24/2018] [Indexed: 11/24/2022] Open
Abstract
Introduction: Synaptic plasticity is inappropriately affected by neurodegenerative diseases, including Alzheimer Disease (AD). In this study, we examined the effect of intrahippocampal amyloid-beta (Aβ1-40) on dentate gyrus Long-term Potentiation (LTP) and presynaptic short-term plasticity in a rat model of AD. Methods: The experimental groups in this research included the control with no treatment, sham-operated receiving the vehicle (normal saline), and Aβ-lesioned groups. For modeling AD, aggregated Aβ1-40 (10 μg/2 μl on each side) was injected into the hippocampal CA1. Three weeks later, Population Spike (PS) amplitude and slope ratios were determined at different Inter-pulse Intervals (IPI) of 10, 20, 30, and 50 ms as a valid indicator of the short-term presynaptic facilitation and/or depression. In addition, PS amplitude and slope were taken as an index of long-term synaptic plasticity after application of High-frequency Stimulation (HFS) to induce LTP in the medial perforant-dentate gyrus pathway. Results: No significant differences were noted amongst the experimental groups regarding fEPSP slope and paired-pulse indices as indicators of short-term plasticity. In contrast, fEPSP slope and PS amplitude significantly decreased following the application of HFS in Aβ-injected group. In addition, there was no significant difference between the control and sham-operated groups regarding the mentioned parameters. Conclusion: Findings of this study clearly demonstrated that microinjection of Aβ1-40 into the CA1 could impair LTP in dentate gyrus but could not modify short-term plasticity.
Collapse
Affiliation(s)
| | - Tourandokht Baluchnejadmojarad
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
20
|
Zhou Y, Kuang G, Li J, Halldin C, Nordberg A, Långström B, Tu Y, Ågren H. In silico studies of ASEM analogues targeting α7-nAChR and experimental verification. RSC Adv 2021; 11:3942-3951. [PMID: 35747361 PMCID: PMC9134020 DOI: 10.1039/d0ra10435c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/11/2021] [Indexed: 01/05/2023] Open
Abstract
The α7 nicotinic acetylcholine receptor (α7-nAChR) is implicated in a variety of neurodegenerative and neuropsychiatric disorders, such as Alzheimer's disease (AD) and schizophrenia. The progress of these disorders can be studied using positron emission tomography (PET) with radiotracers for α7-nAChR. [18F]ASEM and [18F] para-ASEM (also referred to as [18F]DBT-10) are novel and potent α7-nAChR PET radiotracers which have successfully been used in human subjects and nonhuman primates, though further improvement of them is still a pressing task in the community of neurodegeneration research. In this work, we demonstrate the use of modern in silico techniques to predict the binding modes, binding strengths, and residence times for molecular PET tracers binding to proteins, using ASEM and DBT-10 as a showcase of the predictive and interpretational power of such techniques, in particular free energy perturbation theory. The corresponding compounds were synthesized and further tested by in vitro binding experiment for validation. Encouragingly, our in silico modeling can correctly predict the binding affinities of the ASEM analogues. The structure-activity relationships for the ortho- and para-substitutions are well explained at the atomistic level and provide structure-based guiding for the future development of PET tracers for α7-nAChR. A discussion is presented on the complementary use of in silico rational methods based on atomic and electronic principles for in vitro characterization of PET tracers.
Collapse
Affiliation(s)
- Yang Zhou
- School of Pharmacy, Jinan University Guangzhou 510632 China
- Department of Physics and Astronomy, Uppsala University Box 516 SE-751 20 Uppsala Sweden
| | - Guanglin Kuang
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center S-106 91 Stockholm Sweden
| | - Junhao Li
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center S-106 91 Stockholm Sweden
| | - Christer Halldin
- Karolinska Institutet, Department of Clinical Neuroscience, Centre for Psychiatric Research 171 76 Stockholm Sweden
| | - Agneta Nordberg
- Nordberg Translational Molecular Imaging Lab, Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet 141 84 Stockholm Sweden
- Theme Aging Karolinska University Hospital S-141 86 Stockholm Sweden
| | - Bengt Långström
- Department of Chemistry, Uppsala University SE- 751 23 Uppsala Sweden
| | - Yaoquan Tu
- Division of Theoretical Chemistry and Biology, School of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center S-106 91 Stockholm Sweden
| | - Hans Ågren
- Department of Physics and Astronomy, Uppsala University Box 516 SE-751 20 Uppsala Sweden
- College of Chemistry and Chemical Engineering, Henan University Kaifeng Henan 475004 P.R. China
| |
Collapse
|
21
|
Roberts JP, Stokoe SA, Sathler MF, Nichols RA, Kim S. Selective coactivation of α7- and α4β2-nicotinic acetylcholine receptors reverses beta-amyloid-induced synaptic dysfunction. J Biol Chem 2021; 296:100402. [PMID: 33571523 PMCID: PMC7961090 DOI: 10.1016/j.jbc.2021.100402] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 01/04/2023] Open
Abstract
Beta-amyloid (Aβ) has been recognized as an early trigger in the pathogenesis of Alzheimer's disease (AD) leading to synaptic and cognitive impairments. Aβ can alter neuronal signaling through interactions with nicotinic acetylcholine receptors (nAChRs), contributing to synaptic dysfunction in AD. The three major nAChR subtypes in the hippocampus are composed of α7-, α4β2-, and α3β4-nAChRs. Aβ selectively affects α7- and α4β2-nAChRs, but not α3β4-nAChRs in hippocampal neurons, resulting in neuronal hyperexcitation. However, how nAChR subtype selectivity for Aβ affects synaptic function in AD is not completely understood. Here, we showed that Aβ associated with α7- and α4β2-nAChRs but not α3β4-nAChRs. Computational modeling suggested that two amino acids in α7-nAChRs, arginine 208 and glutamate 211, were important for the interaction between Aβ and α7-containing nAChRs. These residues are conserved only in the α7 and α4 subunits. We therefore mutated these amino acids in α7-containing nAChRs to mimic the α3 subunit and found that mutant α7-containing receptors were unable to interact with Aβ. In addition, mutant α3-containing nAChRs mimicking the α7 subunit interact with Aβ. This provides direct molecular evidence for how Aβ selectively interacted with α7- and α4β2-nAChRs, but not α3β4-nAChRs. Selective coactivation of α7- and α4β2-nAChRs also sufficiently reversed Aβ-induced AMPA receptor dysfunction, including Aβ-induced reduction of AMPA receptor phosphorylation and surface expression in hippocampal neurons. Moreover, costimulation of α7- and α4β2-nAChRs reversed the Aβ-induced disruption of long-term potentiation. These findings support a novel mechanism for Aβ's impact on synaptic function in AD, namely, the differential regulation of nAChR subtypes.
Collapse
Affiliation(s)
- Jessica P Roberts
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, Colorado, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Sarah A Stokoe
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, Colorado, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Matheus F Sathler
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Robert A Nichols
- Department of Cell and Molecular Biology, University of Hawai'i at Manoa, Honolulu, Hawaii, USA
| | - Seonil Kim
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, Colorado, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.
| |
Collapse
|
22
|
He B, Perez SE, Lee SH, Ginsberg SD, Malek-Ahmadi M, Mufson EJ. Expression profiling of precuneus layer III cathepsin D-immunopositive pyramidal neurons in mild cognitive impairment and Alzheimer's disease: Evidence for neuronal signaling vulnerability. J Comp Neurol 2020; 528:2748-2766. [PMID: 32323319 PMCID: PMC7492791 DOI: 10.1002/cne.24929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
The precuneus (PreC; Brodmann area 7), a key hub within the default mode network (DMN) displays amyloid and tau-containing neurofibrillary tangle (NFT) pathology during the onset of Alzheimer's disease (AD). PreC layer III projection neurons contain lysosomal hydrolase cathepsin D (CatD), a marker of neurons vulnerable to NFT pathology. Here we applied single population laser capture microdissection coupled with custom-designed microarray profiling to determine the genetic signature of PreC CatD-positive-layer III neurons accrued from postmortem tissue obtained from the Rush Religious Orders Study (RROS) cases with a premortem clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI) and AD. Expression profiling revealed significant differential expression of key transcripts in MCI and AD compared to NCI that underlie signaling defects, including dysregulation of genes within the endosomal-lysosomal and autophagy pathways, cytoskeletal elements, AD-related genes, ionotropic and metabotropic glutamate receptors, cholinergic enzymes and receptors, markers of monoamine neurotransmission as well as steroid-related transcripts. Pervasive defects in both MCI and AD were found in select transcripts within these key gene ontology categories, underscoring the vulnerability of these corticocortical projection neurons during the onset and progression of dementia. Select PreC dysregulated genes detected via custom-designed microarray analysis were validated using qPCR. In summary, expression profiling of PreC CatD -positive layer III neurons revealed significant dysregulation of a mosaic of genes in MCI and AD that were not previously appreciated in terms of their indication of systems-wide signaling defects in a key hub of the DMN.
Collapse
Affiliation(s)
- Bin He
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| | - Sylvia E. Perez
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| | - Sang Han Lee
- Center for Biomedical Imaging and Neuromodulation, Nathan
Kline Institute, Orangeburg, New York
- Child and Adolescent Psychiatry, New York University School
of Medicine, New York, New York
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute,
Orangeburg, New York
- Department of Psychiatry, New York University School of
Medicine, New York, New York
- Neuroscience & Physiology, New York University School
of Medicine, New York, New York
- NYU Neuroscience Institute, New York University School of
Medicine, New York, New York
| | | | - Elliott J. Mufson
- Department of Neurobiology and Neurology, Barrow
Neurological Institute, Phoenix, Arizona
| |
Collapse
|
23
|
Martínez-Cué C, Rueda N. Signalling Pathways Implicated in Alzheimer's Disease Neurodegeneration in Individuals with and without Down Syndrome. Int J Mol Sci 2020; 21:E6906. [PMID: 32962300 PMCID: PMC7555886 DOI: 10.3390/ijms21186906] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Down syndrome (DS), the most common cause of intellectual disability of genetic origin, is characterized by alterations in central nervous system morphology and function that appear from early prenatal stages. However, by the fourth decade of life, all individuals with DS develop neuropathology identical to that found in sporadic Alzheimer's disease (AD), including the development of amyloid plaques and neurofibrillary tangles due to hyperphosphorylation of tau protein, loss of neurons and synapses, reduced neurogenesis, enhanced oxidative stress, and mitochondrial dysfunction and neuroinflammation. It has been proposed that DS could be a useful model for studying the etiopathology of AD and to search for therapeutic targets. There is increasing evidence that the neuropathological events associated with AD are interrelated and that many of them not only are implicated in the onset of this pathology but are also a consequence of other alterations. Thus, a feedback mechanism exists between them. In this review, we summarize the signalling pathways implicated in each of the main neuropathological aspects of AD in individuals with and without DS as well as the interrelation of these pathways.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, 39011 Santander, Spain;
| | | |
Collapse
|
24
|
Koulousakis P, Andrade P, Visser-Vandewalle V, Sesia T. The Nucleus Basalis of Meynert and Its Role in Deep Brain Stimulation for Cognitive Disorders: A Historical Perspective. J Alzheimers Dis 2020; 69:905-919. [PMID: 31104014 DOI: 10.3233/jad-180133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The nucleus basalis of Meynert (nbM) was first described at the end of the 19th century and named after its discoverer, Theodor Meynert. The nbM contains a large population of cholinergic neurons that project their axons to the entire cortical mantle, the olfactory tubercle, and the amygdala. It has been functionally associated with the control of attention and maintenance of arousal, both key functions for appropriate learning and memory formation. This structure is well-conserved across vertebrates, although its degree of organization varies between species. Since early in the investigation of its functional and pathological significance, its degeneration has been linked to various major neuropsychiatric disorders. For instance, Lewy bodies, a hallmark in the diagnosis of Parkinson's disease, were originally described in the nbM. Since then, its involvement in other Lewy body and dementia-related disorders has been recognized. In the context of recent positive outcomes following nbM deep brain stimulation in subjects with dementia-associated disorders, we review the literature from an historical perspective focusing on how the nbM came into focus as a promising therapeutic option for patients with Alzheimer's disease. Moreover, we will discuss what is needed to further develop and widely implement this approach as well as examine novel medical indications for which nbM deep brain stimulation may prove beneficial.
Collapse
Affiliation(s)
- Philippos Koulousakis
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Cologne, Germany.,European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Pablo Andrade
- Department of Neurosurgery, University Hospital of Cologne, Germany.,European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Veerle Visser-Vandewalle
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Cologne, Germany.,European Graduate School of Neuroscience, Maastricht, The Netherlands
| | - Thibaut Sesia
- Department of Stereotactic and Functional Neurosurgery, University Hospital of Cologne, Germany.,European Graduate School of Neuroscience, Maastricht, The Netherlands
| |
Collapse
|
25
|
Dhara M, Matta JA, Lei M, Knowland D, Yu H, Gu S, Bredt DS. Polyamine regulation of ion channel assembly and implications for nicotinic acetylcholine receptor pharmacology. Nat Commun 2020; 11:2799. [PMID: 32493979 PMCID: PMC7271128 DOI: 10.1038/s41467-020-16629-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/15/2020] [Indexed: 01/31/2023] Open
Abstract
Small molecule polyamines are abundant in all life forms and participate in diverse aspects of cell growth and differentiation. Spermidine/spermine acetyltransferase (SAT1) is the rate-limiting enzyme in polyamine catabolism and a primary genetic risk factor for suicidality. Here, using genome-wide screening, we find that SAT1 selectively controls nicotinic acetylcholine receptor (nAChR) biogenesis. SAT1 specifically augments assembly of nAChRs containing α7 or α4β2, but not α6 subunits. Polyamines are classically studied as regulators of ion channel gating that engage the nAChR channel pore. In contrast, we find polyamine effects on assembly involve the nAChR cytosolic loop. Neurological studies link brain polyamines with neurodegenerative conditions. Our pharmacological and transgenic animal studies find that reducing polyamines enhances cortical neuron nAChR expression and augments nicotine-mediated neuroprotection. Taken together, we describe a most unexpected role for polyamines in regulating ion channel assembly, which provides a new avenue for nAChR neuropharmacology. Small molecule polyamines participate in diverse aspects of cell growth and differentiation and are known to regulate ion channel gating. Here authors reveal that cellular polyamines control nicotinic acetylcholine receptor (nAChR) biogenesis, and either catabolic degradation or inhibition of polyamine production augments nAChR assembly.
Collapse
Affiliation(s)
- Madhurima Dhara
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Jose A Matta
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Min Lei
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Daniel Knowland
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Hong Yu
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Shenyan Gu
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - David S Bredt
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson and Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| |
Collapse
|
26
|
Majdi A, Sadigh-Eteghad S, Rahigh Aghsan S, Farajdokht F, Vatandoust SM, Namvaran A, Mahmoudi J. Amyloid-β, tau, and the cholinergic system in Alzheimer's disease: seeking direction in a tangle of clues. Rev Neurosci 2020; 31:391-413. [PMID: 32017704 DOI: 10.1515/revneuro-2019-0089] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/22/2019] [Indexed: 12/14/2022]
Abstract
The link between histopathological hallmarks of Alzheimer's disease (AD), i.e. amyloid plaques, and neurofibrillary tangles, and AD-associated cognitive impairment, has long been established. However, the introduction of interactions between amyloid-beta (Aβ) as well as hyperphosphorylated tau, and the cholinergic system to the territory of descriptive neuropathology has drastically changed this field by adding the theory of synaptic neurotransmission to the toxic pas de deux in AD. Accumulating data show that a multitarget approach involving all amyloid, tau, and cholinergic hypotheses could better explain the evolution of events happening in AD. Various species of both Aβ and tau could be traced in cholinergic neurons of the basal forebrain system early in the course of the disease. These molecules induce degeneration in the neurons of this system. Reciprocally, aberrant cholinergic system modulation promotes changes in amyloid precursor protein (APP) metabolism and tau phosphorylation, resulting in neurotoxicity, neuroinflammation, and neuronal death. Altogether, these changes may better correlate with the clinical findings and cognitive impairment detected in AD patients. Failure of several of Aβ- and tau-related therapies further highlights the need for special attention to molecules that target all of these mentioned pathologic changes. Another noteworthy fact here is that none of the popular hypotheses of AD such as amyloidopathy or tauopathy seem to be responsible for the changes observed in AD alone. Thus, the main culprit should be sought higher in the stream somewhere in APP metabolism or Wnt signaling in the cholinergic system of the basal forebrain. Future studies should target these pathological events.
Collapse
Affiliation(s)
- Alireza Majdi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Sepideh Rahigh Aghsan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Seyed Mehdi Vatandoust
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Ali Namvaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| |
Collapse
|
27
|
He W, Tu M, Du Y, Li J, Pang Y, Dong Z. Nicotine Promotes AβPP Nonamyloidogenic Processing via RACK1-Dependent Activation of PKC in SH-SY5Y-AβPP695 Cells. J Alzheimers Dis 2020; 75:451-460. [PMID: 32250310 DOI: 10.3233/jad-200003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Accumulation of amyloid-β (Aβ) peptides, generated from amyloid-β precursor protein (AβPP) amyloidogenic processing, is one of the most salient disease hallmarks of Alzheimer's disease (AD). Nicotine is able to promote α-secretase-mediated AβPP nonamyloidogenic processing and increase the release of sAβPPα and C-terminal fragment of 83 amino acids (C83). However, the potential molecular mechanism remains elusive. OBJECTIVE The aim of the present study was to investigate the effect of nicotine on AβPP processing in SH-SY5Y cells that stably express human Swedish mutant AβPP695 (SH-SY5Y-AβPP695). METHODS The expression of AβPP and its C-terminal fragments including C99, C89, and C83, was measured in SH-SY5Y-AβPP695 cells treated with nicotine for 6 h. Protein kinase C (PKC) antagonist Ro30-8220 or agonist PMA was used to determine the role of PKC in AβPP processing. Lentivirus-mediated shRNA targeting receptor for activated C-kinase 1 (RACK1) gene was added into the media to knockdown RACK1 expression, and then AβPP processing was examined. RESULTS The results showed that 6 h of nicotine exposure increased the expression of α-secretase (ADAM10) and C83 in a dose dependent manner. While the β-secretase (BACE1), AβPP amyloidogenic processing products C89 and C99 as well as Aβ peptides (including Aβ40 and Aβ42) remained unchanged. We also found that nicotine elevated the expression of phosphorylated PKC (P-PKC) and RACK1 on the cytomembrane. PKC antagonist Ro30-8220 treatment prevented the increase of ADAM10 and C83 by nicotine. Genetic knockdown RACK1 significantly inhibited P-PKC, and consequently abolished the increase of ADAM10 and C83 by nicotine. CONCLUSION Taken together, these results indicate that nicotine effectively promotes AβPP nonamyloidogenic processing via RACK1-dependent activation of PKC in SH-SY5Y-AβPP695 cells and could be a potential molecule for AD treatment.
Collapse
Affiliation(s)
- Wenting He
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Man Tu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yehong Du
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Junjie Li
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yayan Pang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
28
|
Gao H, Wang S, Qiang B, Wang S, Zhang H. Radioiodinated 9-fluorenone derivatives for imaging α7-nicotinic acetylcholine receptors. MEDCHEMCOMM 2019; 10:2102-2110. [PMID: 32904124 DOI: 10.1039/c9md00415g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/14/2019] [Indexed: 11/21/2022]
Abstract
A series of 9H-fluoren-9-one substituents were synthesized and evaluated for imaging cerebral α7-nAChRs. Meta-iodine substituted 9-fluorenone 5 with high binding affinity (K i = 9.3 nM) and selectivity was radiolabeled with 125I. Fully in vitro and in vivo studies of [125I]5 have been performed. [125I]5 exhibited well brain uptake with a peak concentration of 7.5 ± 0.9% ID/g in mice brains. Moreover, ex vivo autoradiography studies and micro single-photon emission computed tomography (micro-SPECT/CT) dynamic imaging in mice confirmed its in vivo imaging properties. Besides, molecular docking and MD studies were also performed to interpret the binding mechanisms of the two series of ligands towards α7-nAChRs. To conclude, the meta-iodine substituted 9-fluorenone [125I]5 could be a promising tracer for imaging α7-nAChRs.
Collapse
Affiliation(s)
- Hang Gao
- Key Laboratory of Radiopharmaceuticals of Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , China .
| | - Shuxia Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , China .
| | - Bingchao Qiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , China .
| | - Sixuan Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , China .
| | - Huabei Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education , College of Chemistry , Beijing Normal University , Beijing 100875 , China .
| |
Collapse
|
29
|
Hahm ET, Nagaraja RY, Waro G, Tsunoda S. Cholinergic Homeostatic Synaptic Plasticity Drives the Progression of Aβ-Induced Changes in Neural Activity. Cell Rep 2019; 24:342-354. [PMID: 29996096 DOI: 10.1016/j.celrep.2018.06.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/03/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) is the ability of neurons to exert compensatory changes in response to altered neural activity. How pathologically induced activity changes are intertwined with HSP mechanisms is unclear. We show that, in cholinergic neurons from Drosophila, beta-amyloid (Aβ) peptides Aβ40 and Aβ42 both induce an increase in spontaneous activity. In a transgenic line expressing Aβ42, we observe that this early increase in spontaneous activity is followed by a dramatic reduction in spontaneous events, a progression that has been suggested to occur in cholinergic brain regions of mammalian models of Alzheimer's disease. We present evidence that the early enhancement in synaptic activity is mediated by the Drosophila α7 nicotinic acetylcholine receptor (nAChR) and that, later, Aβ42-induced inhibition of synaptic events is a consequence of Dα7-dependent HSP mechanisms induced by earlier hyperactivity. Thus, while HSP may initially be an adaptive response, it may also drive maladaptive changes and downstream pathologies.
Collapse
Affiliation(s)
- Eu-Teum Hahm
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Raghavendra Y Nagaraja
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Girma Waro
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA.
| |
Collapse
|
30
|
Sun JL, Stokoe SA, Roberts JP, Sathler MF, Nip KA, Shou J, Ko K, Tsunoda S, Kim S. Co-activation of selective nicotinic acetylcholine receptors is required to reverse beta amyloid-induced Ca 2+ hyperexcitation. Neurobiol Aging 2019; 84:166-177. [PMID: 31629115 DOI: 10.1016/j.neurobiolaging.2019.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022]
Abstract
Beta-amyloid (Aβ) peptide accumulation has long been implicated in the pathogenesis of Alzheimer's disease (AD). Hippocampal network hyperexcitability in the early stages of the disease leads to increased epileptiform activity and eventually cognitive decline. We found that acute application of 250 nM soluble Aβ42 oligomers increased Ca2+ activity in hippocampal neurons in parallel with a significant decrease in activity in Aβ42-treated interneurons. A potential target of Aβ42 is the nicotinic acetylcholine receptor (nAChR). Three major subtypes of nAChRs (α7, α4β2, and α3β4) have been reported in the human hippocampus. Simultaneous inhibition of both α7 and α4β2 nAChRs mimicked the Aβ42 effects on both excitatory and inhibitory neurons. However, inhibition of all 3 subtypes showed the opposite effect. Importantly, simultaneous activation of α7 and α4β2 nAChRs was required to reverse Aβ42-induced neuronal hyperexcitation. We suggest co-activation of α7 and α4β2 nAChRs is required to reverse Aβ42-induced Ca2+ hyperexcitation.
Collapse
Affiliation(s)
- Julianna L Sun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Sarah A Stokoe
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Jessica P Roberts
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Matheus F Sathler
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kaila A Nip
- Cellular and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| | - Jiayi Shou
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kaitlyn Ko
- Poudre High School, Fort Collins, CO, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Seonil Kim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA; Cellular and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
31
|
Isomerization of Asp7 in Beta-Amyloid Enhances Inhibition of the α7 Nicotinic Receptor and Promotes Neurotoxicity. Cells 2019; 8:cells8080771. [PMID: 31349637 PMCID: PMC6721525 DOI: 10.3390/cells8080771] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 01/02/2023] Open
Abstract
Cholinergic dysfunction in Alzheimer’s disease (AD) can be mediated by the neuronal α7 nicotinic acetylcholine receptor (α7nAChR). Beta-amyloid peptide (Aβ) binds to the α7nAChR, disrupting the receptor’s function and causing neurotoxicity. In vivo not only Aβ but also its modified forms can drive AD pathogenesis. One of these forms, iso-Aβ (containing an isomerized Asp7 residue), shows an increased neurotoxicity in vitro and stimulates amyloidogenesis in vivo. We suggested that such effects of iso-Aβ are α7nAChR-dependent. Here, using calcium imaging and electrophysiology, we found that iso-Aβ is a more potent inhibitor of the α7nAChR-mediated calcium current than unmodified Aβ. However, Asp7 isomerization eliminated the ability of Aβ to decrease the α7nAChR levels. These data indicate differences in the interaction of the peptides with the α7nAChR, which we demonstrated using computer modeling. Neither Aβ nor iso-Aβ competed with 125I-α-bungarotoxin for binding to the orthosteric site of the receptor, suggesting the allosteric binging mode of the peptides. Further we found that increased neurotoxicity of iso-Aβ was mediated by the α7nAChR. Thus, the isomerization of Asp7 enhances the inhibitory effect of Aβ on the functional activity of the α7nAChR, which may be an important factor in the disruption of the cholinergic system in AD.
Collapse
|
32
|
Richter N, Beckers N, Onur OA, Dietlein M, Tittgemeyer M, Kracht L, Neumaier B, Fink GR, Kukolja J. Effect of cholinergic treatment depends on cholinergic integrity in early Alzheimer's disease. Brain 2019; 141:903-915. [PMID: 29309600 DOI: 10.1093/brain/awx356] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/08/2017] [Indexed: 02/04/2023] Open
Abstract
In early Alzheimer's disease, which initially presents with progressive loss of short-term memory, neurodegeneration especially affects cholinergic neurons of the basal forebrain. Pharmacotherapy of Alzheimer's disease therefore often targets the cholinergic system. In contrast, cholinergic pharmacotherapy of mild cognitive impairment is debated since its efficacy to date remains controversial. We here investigated the relationship between cholinergic treatment effects and the integrity of the cholinergic system in mild cognitive impairment due to Alzheimer's disease. Fourteen patients with high likelihood of mild cognitive impairment due to Alzheimer's disease and 16 age-matched cognitively normal adults performed an episodic memory task during functional magnetic resonance imaging under three conditions: (i) without pharmacotherapy; (ii) with placebo; and (iii) with a single dose of rivastigmine (3 mg). Cortical acetylcholinesterase activity was measured using PET with the tracer 11C-N-methyl-4-piperidyl acetate (MP4A). Cortical acetylcholinesterase activity was significantly decreased in patients relative to controls, especially in the lateral temporal lobes. Without pharmacotherapy, mild cognitive impairment was associated with less memory-related neural activation in the fusiform gyrus and impaired deactivation in the posterior cingulate cortex, relative to controls. These differences were attenuated under cholinergic stimulation with rivastigmine: patients showed increased neural activation in the right fusiform gyrus but enhanced deactivation of the posterior cingulate cortex under rivastigmine, compared to placebo. Conversely, controls showed reduced activation of the fusiform gyrus and reduced deactivation of the posterior cingulate under rivastigmine, compared to placebo. In both groups, the change in neural activation in response to rivastigmine was negatively associated with local acetylcholinesterase activity. At the behavioural level, an analysis of covariance revealed a significant group × treatment interaction in episodic memory performance when accounting for hippocampal grey matter atrophy and function. Our results indicate that rivastigmine differentially affects memory-related neural activity in patients with mild cognitive impairment and cognitively normal, age-matched adults, depending on acetylcholinesterase activity as a marker for the integrity of the cortical cholinergic system. Furthermore, hippocampal integrity showed an independent association with the response of memory performance to acetylcholinesterase inhibition.
Collapse
Affiliation(s)
- Nils Richter
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany.,Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany
| | - Nora Beckers
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany
| | - Oezguer A Onur
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Marc Tittgemeyer
- Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany
| | - Lutz Kracht
- Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany.,Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Bernd Neumaier
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, 52425 Jülich, Germany.,Institute for Radiochemistry and Experimental Molecular Imaging, University Hospital Cologne, 50937 Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Juraj Kukolja
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany.,Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| |
Collapse
|
33
|
Chang YT, Kazui H, Ikeda M, Huang CW, Huang SH, Hsu SW, Chang WN, Chang CC. Genetic Interaction of APOE and FGF1 is Associated with Memory Impairment and Hippocampal Atrophy in Alzheimer's Disease. Aging Dis 2019; 10:510-519. [PMID: 31164996 PMCID: PMC6538224 DOI: 10.14336/ad.2018.0606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/06/2018] [Indexed: 11/30/2022] Open
Abstract
The APOE and fibroblast growth factor 1 (FGF1) have both been associated with amyloid β accumulation and neurodegeneration. Investigation the effect of APOE-FGF1 interactions on episodic memory (EM) deficits and hippocampus atrophy (HA) might elucidate the complex clinical-pathological relationship in Alzheimer’s disease (AD). EM performance and hippocampal volume (HV) were characterized in patients with mild AD based on APOE-ε4 carrier status (APOE-ε4 carriers versus non-carriers) and FGF1 single nucleotide polymorphism (FGF1-rs34011-GG versus FGF1-rs34011-A-allele carriers). The clinical-pathological relationships within each genotypic group (ε4+/GG-carrier, ε4+/A-allele-carrier, ε4-/GG-carrier and ε4-/A-allele-carrier) were analyzed. There were no significant differences between the FGF1-rs34011-GG and FGF1-rs34011-A-allele carriers for the level of EM performance or HV (p> 0.05). The bilateral HV was significantly smaller and EM impairment was significantly worse in ε4+/GG-carrier than in ε4-/A-allele-carrier, and an interaction effect of APOE (APOE-ε4 carriers versus non-carriers) with FGF1 (FGF1-rs34011-GG versus FGF1-rs34011-A-allele carriers) predicted EM impairment (F4,92= 3.516, p= 0.018) and structural changes in voxel-based morphometry. Our data shows that concurrent consideration of APOE and FGF1 polymorphisms might be required to understand the clinical-pathological relationship in AD.
Collapse
Affiliation(s)
- Ya-Ting Chang
- 1Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Hiroaki Kazui
- 2Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Manabu Ikeda
- 2Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Chi-Wei Huang
- 1Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Shu-Hua Huang
- 3Department of Nuclear Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Shih-Wei Hsu
- 4Department of Radiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Wen-Neng Chang
- 1Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chiung-Chih Chang
- 1Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| |
Collapse
|
34
|
Dey S, Ray K. Cholinergic activity is essential for maintaining the anterograde transport of Choline Acetyltransferase in Drosophila. Sci Rep 2018; 8:8028. [PMID: 29795337 PMCID: PMC5966444 DOI: 10.1038/s41598-018-26176-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/30/2018] [Indexed: 12/14/2022] Open
Abstract
Cholinergic activity is essential for cognitive functions and neuronal homeostasis. Choline Acetyltransferase (ChAT), a soluble protein that synthesizes acetylcholine at the presynaptic compartment, is transported in bulk in the axons by the heterotrimeric Kinesin-2 motor. Axonal transport of soluble proteins is described as a constitutive process assisted by occasional, non-specific interactions with moving vesicles and motor proteins. Here, we report that an increase in the influx of Kinesin-2 motor and association between ChAT and the motor during a specific developmental period enhances the axonal entry, as well as the anterograde flow of the protein, in the sensory neurons of intact Drosophila nervous system. Loss of cholinergic activity due to Hemicholinium and Bungarotoxin treatments, respectively, disrupts the interaction between ChAT and Kinesin-2 in the axon, and the episodic enhancement of axonal influx of the protein. Altogether, these observations highlight a phenomenon of synaptic activity-dependent, feedback regulation of a soluble protein transport in vivo, which could potentially define the quantum of its pre-synaptic influx.
Collapse
Affiliation(s)
- Swagata Dey
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| | - Krishanu Ray
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
35
|
Xu R, Xiao G, Li Y, Liu H, Song Q, Zhang X, Yang Z, Zheng Y, Tan Z, Deng Y. Multifunctional 5,6-dimethoxybenzo[d]isothiazol-3(2H)-one-N-alkylbenzylamine derivatives with acetylcholinesterase, monoamine oxidases and β-amyloid aggregation inhibitory activities as potential agents against Alzheimer’s disease. Bioorg Med Chem 2018; 26:1885-1895. [DOI: 10.1016/j.bmc.2018.02.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/07/2018] [Accepted: 02/20/2018] [Indexed: 12/17/2022]
|
36
|
Zhao L, Xiao Y, Xiu J, Tan LC, Guan ZZ. Protection against the Neurotoxic Effects of β-Amyloid Peptide on Cultured Neuronal Cells by Lovastatin Involves Elevated Expression of α7 Nicotinic Acetylcholine Receptors and Activating Phosphorylation of Protein Kinases. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1081-1093. [DOI: 10.1016/j.ajpath.2017.11.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/27/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
|
37
|
Sacramento AS, Moreira FT, Guerreiro JL, Tavares AP, Sales MGF. Novel biomimetic composite material for potentiometric screening of acetylcholine, a neurotransmitter in Alzheimer's disease. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017. [DOI: 10.1016/j.msec.2017.05.098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
38
|
Castillo-Ordóñez WO, Tamarozzi ER, da Silva GM, Aristizabal-Pachón AF, Sakamoto-Hojo ET, Takahashi CS, Giuliatti S. Exploration of the Acetylcholinesterase Inhibitory Activity of Some Alkaloids from Amaryllidaceae Family by Molecular Docking In Silico. Neurochem Res 2017; 42:2826-2830. [DOI: 10.1007/s11064-017-2295-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/27/2017] [Accepted: 05/06/2017] [Indexed: 01/24/2023]
|
39
|
Fernandes CEM, Serafim KR, Gianlorenço ACL, Mattioli R. Cholinergic agonist reverses H1-induced memory deficit in mice. Prog Neuropsychopharmacol Biol Psychiatry 2017; 72:16-22. [PMID: 27528108 DOI: 10.1016/j.pnpbp.2016.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 07/28/2016] [Accepted: 08/11/2016] [Indexed: 01/15/2023]
Abstract
This study investigated the effects of bilateral intraamygdalar microinjections of PNU-282987, a nicotinic cholinergic agonist, on anxiety and the reversal of amnesia induced by chlorpheniramine (CPA), an H1 histaminergic antagonist, in mice subjected to the elevated plusmaze (EPM). Two experiments were performed with seventy-nine adult male Swiss mice. The isolated microinjections of PNU-282987 did not produce effects on emotional memory; however, the combined microinjections of PNU-282987 and CPA were able to reverse the deficit in memory induced by CPA (ANOVA, p<0.05). Taken together, these results suggest that intraamygdalar injections of PNU-282987 did not induce effects on anxiety and emotional memory per se; however, concurrent microinjections of PNU-282987 and CPA-reverse amnesia induced-CPA which is suggestive of an interaction between the histaminergic and cholinergic systems in the modulation of emotion memory acquisition in mice.
Collapse
Affiliation(s)
- C E M Fernandes
- Laboratory of Neuroscience, Physiotherapy Department, Center of Biological Sciences and Health, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, 13565-905 Sao Carlos, Brazil.
| | - K R Serafim
- Laboratory of Neuroscience, Physiotherapy Department, Center of Biological Sciences and Health, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, 13565-905 Sao Carlos, Brazil.
| | - A C L Gianlorenço
- Laboratory of Neuroscience, Physiotherapy Department, Center of Biological Sciences and Health, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, 13565-905 Sao Carlos, Brazil.
| | - R Mattioli
- Laboratory of Neuroscience, Physiotherapy Department, Center of Biological Sciences and Health, Federal University of Sao Carlos, Rod. Washington Luis, Km 235, 13565-905 Sao Carlos, Brazil.
| |
Collapse
|
40
|
Vicens P, Heredia L, Torrente M, Domingo JL. Behavioural effects of PNU-282987 and stress in an animal model of Alzheimer's disease. Psychogeriatrics 2017; 17:33-42. [PMID: 26817787 DOI: 10.1111/psyg.12189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cholinergic deficits play an important role in both cognitive and behavioural alterations in Alzheimer's disease. This study was aimed at evaluating the possible therapeutic role of PNU-282987 (PNU), an α7 nicotinic cholinergic receptor agonist, and the possible effects of stress in precipitating the onset of behavioural deficits in animals with susceptibility to Alzheimer's disease. METHODS B6C3-Tg mice with susceptibility to Alzheimer's disease and wild-type mice either with or without restraint stress received 0- or 1-mg/kg PNU. At 12 months old, mice were evaluated for activity levels, anxiety-like levels, and spatial learning and memory. RESULTS Data did not show the effects of PNU on activity and anxiety-like behaviour. No effect of PNU on acquisition of a spatial learning task was detected, but a reversal of stress effects on retention in the Morris water maze was observed in transgenic mice. CONCLUSIONS Further studies are needed in order to better understand the role of α7 nicotinic cholinergic receptor agonists in motor activity, anxiety, and spatial learning and memory and to develop more accurate pharmacological treatment of psychopathological diseases.
Collapse
Affiliation(s)
- Paloma Vicens
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - Luis Heredia
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - Margarita Torrente
- Department of Psychology, Universitat Rovira i Virgili, Tarragona, Spain.,Research Center in Behavioral Assessment (CRAMC), Universitat Rovira i Virgili, Tarragona, Spain.,Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| | - José L Domingo
- Laboratory of Toxicology and Environmental Health, School of Medicine, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Tarragona, Spain
| |
Collapse
|
41
|
Castillo WO, Aristizabal-Pachon AF, de Lima Montaldi AP, Sakamoto-Hojo ET, Takahashi CS. Galanthamine decreases genotoxicity and cell death induced by β-amyloid peptide in SH-SY5Y cell line. Neurotoxicology 2016; 57:291-297. [DOI: 10.1016/j.neuro.2016.10.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/11/2016] [Accepted: 10/23/2016] [Indexed: 12/11/2022]
|
42
|
Pardo M, Beurel E, Jope RS. Cotinine administration improves impaired cognition in the mouse model of Fragile X syndrome. Eur J Neurosci 2016; 45:490-498. [PMID: 27775852 DOI: 10.1111/ejn.13446] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 01/15/2023]
Abstract
Cotinine is the major metabolite of nicotine and has displayed some capacity for improving cognition in mouse models following chronic administration. We tested if acute cotinine treatment is capable of improving cognition in the mouse model of Fragile X syndrome, Fmr1-/- knockout mice, and if this is related to inhibition by cotinine treatment of glycogen synthase kinase-3β (GSK3β), which is abnormally active in Fmr1-/- mice. Acute cotinine treatment increased the inhibitory serine-phosphorylation of GSK3β and the activating phosphorylation of AKT, which can mediate serine-phosphorylation of GSK3β, in both wild-type and Fmr1-/- mouse hippocampus. Acute cotinine treatment improved cognitive functions of Fmr1-/- mice in coordinate and categorical spatial processing, novel object recognition, and temporal ordering. However, cotinine failed to restore impaired cognition in GSK3β knockin mice, in which a serine9-to-alanine9 mutation blocks the inhibitory serine phosphorylation of GSK3β, causing GSK3β to be hyperactive. These results indicate that acute cotinine treatment effectively repairs impairments of these four cognitive tasks in Fmr1-/- mice, and suggest that this cognition-enhancing effect of cotinine is linked to its induction of inhibitory serine-phosphorylation of GSK3. Taken together, these results show that nicotinic receptor agonists can act as cognitive enhancers in a mouse model of Fragile X syndrome and highlight the potential role of inhibiting GSK3β in mediating the beneficial effects of cotinine on memory.
Collapse
Affiliation(s)
- Marta Pardo
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Eleonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Richard S Jope
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.,Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
43
|
Lombardo S, Catteau J, Besson M, Maskos U. A role for β2* nicotinic receptors in a model of local amyloid pathology induced in dentate gyrus. Neurobiol Aging 2016; 46:221-34. [DOI: 10.1016/j.neurobiolaging.2016.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 05/22/2016] [Accepted: 06/09/2016] [Indexed: 12/22/2022]
|
44
|
Garg PK, Lokitz SJ, Nazih R, Garg S. Biodistribution and Radiation Dosimetry of 11C-Nicotine from Whole-Body PET Imaging in Humans. J Nucl Med 2016; 58:473-478. [PMID: 27660145 DOI: 10.2967/jnumed.116.180059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/28/2016] [Indexed: 11/16/2022] Open
Abstract
This study assessed the in vivo distribution of 11C-nicotine and the absorbed radiation dose from whole-body 11C-nicotine PET imaging of 11 healthy (5 male and 6 female) subjects. Methods: After an initial CT attenuation scan, 11C-nicotine was administered via intravenous injection. A dynamic PET scan was acquired for 90 s with the brain in the field of view, followed by a series of 13 whole-body PET scans acquired over a 90-min period. Regions of interest were drawn over organs visible in the reconstructed PET images. Time-activity curves were generated, and the residence times were calculated. The absorbed radiation dose for the whole body was calculated by entering the residence time in OLINDA/EXM 1.0 software to model the equivalent organ dose and the effective dose for a 70-kg man. Results: The mean residence times for 11C-nicotine in the liver, red marrow, brain, and lungs were 0.048 ± 0.010, 0.031 ± 0.005, 0.021 ± 0.004, and 0.020 ± 0.005 h, respectively. The mean effective dose for 11C-nicotine was 5.44 ± 0.67 μSv/MBq. The organs receiving the highest absorbed dose from the 11C-nicotine injection were the urinary bladder wall (14.68 ± 8.70 μSv/MBq), kidneys (9.56 ± 2.46 μSv/MBq), liver (8.94 ± 1.67 μSv/MBq), and spleen (9.49 ± 3.89 μSv/MBq). The renal and hepatobiliary systems were the major clearance and excretion routes for radioactivity. Conclusion: The estimated radiation dose from 11C-nicotine administration is relatively modest and would allow for multiple PET examinations on the same subject.
Collapse
Affiliation(s)
- Pradeep K Garg
- Department of Radiology, Wake Forest University Health Sciences, Winston Salem, North Carolina; and Biomedical Research Foundation, Shreveport, Louisiana
| | - Stephen J Lokitz
- Department of Radiology, Wake Forest University Health Sciences, Winston Salem, North Carolina; and Biomedical Research Foundation, Shreveport, Louisiana
| | - Rachid Nazih
- Department of Radiology, Wake Forest University Health Sciences, Winston Salem, North Carolina; and Biomedical Research Foundation, Shreveport, Louisiana
| | - Sudha Garg
- Department of Radiology, Wake Forest University Health Sciences, Winston Salem, North Carolina; and Biomedical Research Foundation, Shreveport, Louisiana
| |
Collapse
|
45
|
LC-MS Supported Studies on the in Vitro Metabolism of both Enantiomers of Flubatine and the in Vivo Metabolism of (+)-[(18)F]Flubatine-A Positron Emission Tomography Radioligand for Imaging α4β2 Nicotinic Acetylcholine Receptors. Molecules 2016; 21:molecules21091200. [PMID: 27617996 PMCID: PMC6273452 DOI: 10.3390/molecules21091200] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 12/27/2022] Open
Abstract
Both enantiomers of [18F]flubatine are promising radioligands for neuroimaging of α4β2 nicotinic acetylcholine receptors (nAChRs) by positron emission tomography (PET). To support clinical studies in patients with early Alzheimer’s disease, a detailed examination of the metabolism in vitro and in vivo has been performed. (+)- and (−)-flubatine, respectively, were incubated with liver microsomes from mouse and human in the presence of NADPH (β-nicotinamide adenine dinucleotide 2′-phosphate reduced tetrasodium salt). Phase I in vitro metabolites were detected and their structures elucidated by LC-MS/MS (liquid chromatography-tandem mass spectrometry). Selected metabolite candidates were synthesized and investigated for structural confirmation. Besides a high level of in vitro stability, the microsomal incubations revealed some species differences as well as enantiomer discrimination with regard to the formation of monohydroxylated products, which was identified as the main metabolic pathway in this assay. Furthermore, after injection of 250 MBq (+)-[18F]flubatine (specific activity > 350 GBq/μmol) into mouse, samples were prepared from brain, liver, plasma, and urine after 30 min and investigated by radio-HPLC (high performance liquid chromatography with radioactivity detection). For structure elucidation of the radiometabolites of (+)-[18F]flubatine formed in vivo, identical chromatographic conditions were applied to LC-MS/MS and radio-HPLC to compare samples obtained in vitro and in vivo. By this correlation approach, we assigned three of four main in vivo radiometabolites to products that are exclusively C- or N-hydroxylated at the azabicyclic ring system of the parent molecule.
Collapse
|
46
|
Woodruff-Pak DS, Gould TJ. Neuronal Nicotinic Acetylcholine Receptors: Involvement in Alzheimer’s Disease and Schizophrenia. ACTA ACUST UNITED AC 2016; 1:5-20. [PMID: 17715584 DOI: 10.1177/1534582302001001002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) play a role in a variety of diseases of the central nervous system including Alzheimer's disease (AD) and schizophrenia. There is great interest in evaluating disease-related nAChR changes, and pharmacological treatment of nAChR deficits is a promising therapy. In AD, 7 nAChRs remain relatively stable, contrasting to 4 2 nAChRs that are lost in substantial numbers. -amyloid, a major neuropathology in AD, blocks 4 2 and 7 nAChRs. Agonists selective to 7 nAChRs are neuroprotective against amyloid. Paradoxically, 7 nAChRs may function as receptors for -amyloid. These results indicate 7 nAChR antagonists may be appropriate therapy in AD. In schizophrenia, 7 nAChRs are significantly reduced in hippocampus and neocortex. The exceptionally high rate of smoking in schizophrenics is likely a form of self-medication. Therapy with 7 nAChR agonists relieves some schizophrenic symptoms. Despite disparities in etiology and symptomatology, AD and schizophrenia share a target for therapeutic intervention— 7 nAChRs.
Collapse
|
47
|
Zhao L, Xiao Y, Wang XL, Pei J, Guan ZZ. Original Research: Influence of okadaic acid on hyperphosphorylation of tau and nicotinic acetylcholine receptors in primary neurons. Exp Biol Med (Maywood) 2016; 241:1825-33. [PMID: 27190248 DOI: 10.1177/1535370216650759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 04/25/2016] [Indexed: 11/15/2022] Open
Abstract
The aim of the study was to investigate the influence of hyperphosphorylation of tau induced by okadaic acid on the expression of nicotinic acetylcholine receptors and the neurotoxicity of β-amyloid peptide. Primary cultures of neurons isolated from the hippocampus of the brains of neonatal rats were exposed to okadaic acid or/and Aβ1-42 Tau phosphorylated at Ser404 and Ser202, and the protein expressions of α7, α4 and α3 nAChR subunits were quantified by Western blotting, and their corresponding mRNAs by real-time PCR. Superoxide dismutase activity was assayed biochemically and malondialdehyde by thiobarbituric acid-reactive substance. As compared to controls, phosphorylations of tau at Ser404 and Ser202 in the neurons were elevated by exposure to 20 nM okadaic acid for 48 h but not by 1 or 2 µM Aβ1-42 Treatment with 20 nM okadaic acid or 1 µM Aβ1-42 for 48 h resulted in the reduced α7, α4 and α3 proteins, and α4 and α3 mRNAs, as well as the decreased activity of superoxide dismutase and the increased malondialdehyde. Okadaic acid and Aβ1-42 together caused more pronounced changes in the expressions of α7 and α4, superoxide dismutase activity and lipid peroxidation than either alone. When pre-treatment with vitamin E or lovastatin, the neurotoxicity induced by okadaic acid was significantly attenuated. These findings indicate that hyperphosphorylation of tau induced by okadaic acid inhibits the expression of nicotinic acetylcholine receptors at both the protein and mRNA levels, as well as enhances the neurotoxicity of β-amyloid peptide.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Pathology at the Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Yan Xiao
- The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Xiao-Liang Wang
- The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| | - Jinjing Pei
- KI-Alzheimer Disease Research Center, Karolinska Institutet, Novum, SE14186 Huddinge, Sweden
| | - Zhi-Zhong Guan
- Department of Pathology at the Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou, PR China The Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang 550004, Guizhou, PR China
| |
Collapse
|
48
|
Chalon S, Vercouillie J, Guilloteau D, Suzenet F, Routier S. PET tracers for imaging brain α7 nicotinic receptors: an update. Chem Commun (Camb) 2016; 51:14826-31. [PMID: 26359819 DOI: 10.1039/c5cc04536c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Positron emission tomography (PET) molecular imaging of brain targets is a powerful tool to diagnose, follow up, and develop treatments and personalized medicine for a number of acute and chronic brain disorders. The availability of β+ emitter tracers labelled with [(11)C] or [(18)F] having optimal characteristics of affinity and selectivity for alpha-7 nicotinic receptors (α7R) has received considerable attention, due to the major implication of these receptors in brain functions. The aim of this review is to identify the interest and need for the in vivo exploration of α7R by PET molecular imaging, which tools are currently available for this and how to progress.
Collapse
Affiliation(s)
- S Chalon
- UMR Inserm U930, Université François-Rabelais de Tours, F-37000 Tours, France.
| | | | | | | | | |
Collapse
|
49
|
Impaired Cholinergic Excitation of Prefrontal Attention Circuitry in the TgCRND8 Model of Alzheimer's Disease. J Neurosci 2016; 35:12779-91. [PMID: 26377466 DOI: 10.1523/jneurosci.4501-14.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Attention deficits in Alzheimer's disease can exacerbate its other cognitive symptoms, yet relevant disruptions of key prefrontal circuitry are not well understood. Here, in the TgCRND8 mouse model of this neurological disorder, we demonstrate and characterize a disruption of cholinergic excitation in the major corticothalamic layer of the prefrontal cortex, in which modulation by acetylcholine is essential for optimal attentional function. Using electrophysiology with concurrent multiphoton imaging, we show that layer 6 pyramidal cells are unable to sustain cholinergic excitation to the same extent as their nontransgenic littermate controls, as a result of the excessive activation of calcium-activated hyperpolarizing conductances. We report that cholinergic excitation can be improved in TgCRND8 cortex by pharmacological blockade of SK channels, suggesting a novel target for the treatment of cognitive dysfunction in Alzheimer's disease. SIGNIFICANCE STATEMENT Alzheimer's disease is accompanied by attention deficits that exacerbate its other cognitive symptoms. In brain slices of a mouse model of this neurological disorder, we demonstrate, characterize, and rescue impaired cholinergic excitation of neurons essential for optimal attentional performance. In particular, we show that the excessive activation of a calcium-activated potassium conductance disrupts the acetylcholine excitation of prefrontal layer 6 pyramidal neurons and that its blockade normalizes responses. These findings point to a novel potential target for the treatment of cognitive dysfunction in Alzheimer's disease.
Collapse
|
50
|
Abstract
The review considers treatment aspects of dementia in Alzheimer's disease (DAD), vascular dementia (VD) and some other forms of dementia. Pathogenetic pharmacotherapy of DAD aims to maintain the activity of cholinergic neurons with anticholinesterase drugs including donepezil, which is used most often. The main objective of VD treatment is prevention of new cerebral infarctions using cardiotropic medications, antiaggregants, anticoagulants, hypotensive drugs and statins. In Russia, patients with DAD and VD receive polymodal drugs as well. These drugs have been included in the list of antidementia medications due to their wide use in treatment of discirculatory encephalopathy and chronic brain ischemia.
Collapse
Affiliation(s)
- M Yu Drobizhev
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - A V Fedotova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - S V Kikta
- Out-patient Clinic #3 of Medical Center of General Management Department of Presidental Administration of the Russian Federation, Moscow, Russia
| | | |
Collapse
|