1
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [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: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanjun Liu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chuhui Yin
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
2
|
Lee R, Kim G, Kim S. Co-activation of selective nicotinic acetylcholine receptor subtypes is required to reverse hippocampal network dysfunction and prevent fear memory loss in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602576. [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 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 and synaptic dysfunction 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 and fear memory loss in the amyloid pathology model mice. We thus suggest that co-activation of PV+ and SST+ cells is a novel strategy to reverse hippocampal dysfunction and cognitive decline in 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
|
Sabec MH, Savage QR, Wood JL, Maskos U. Targeting high-affinity nicotinic receptors protects against the functional consequences of β-amyloid in mouse hippocampus. Mol Psychiatry 2024:10.1038/s41380-024-02666-7. [PMID: 39164528 DOI: 10.1038/s41380-024-02666-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 08/22/2024]
Abstract
The accumulation of β-amyloid oligomers is a hallmark of Alzheimer's disease, inducing neural and network dysfunction in the early stages of pathology. The hippocampus is affected early in the pathogenesis of AD, however the impact of soluble β-amyloid on the dentate gyrus (DG) subregion of the hippocampus and its interaction with nicotinic acetylcholine receptors (nAChRs) within this region are not known. Using a localized model of over-expression, we show that β-amyloid induces early-onset neuronal hyperactivity and hippocampal-dependent memory deficits in mice. Further, we find the DG region to be under potent and sub-type specific nicotinic control in both healthy and pathophysiological conditions, with targeted receptor inhibition leading to a mnemonic rescue against localized amyloidosis. We show that while neurogenesis and synaptic functions are not severely affected in our model, reducing β2-containing nAChR function is associated with the promotion of young adult-born neurons within the pathological network, suggesting a possible protective mechanism. Our data thus reveal the DG network level changes which occur in the early-stages of β-amyloid accumulation and highlight the downstream consequences of targeted nicotinic neuromodulation.
Collapse
Affiliation(s)
- Marie H Sabec
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, 75015, Paris, France.
- Physiology, Pharmacology, and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK.
| | - Quentin R Savage
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76798, USA
| | - John L Wood
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, 76798, USA
| | - Uwe Maskos
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems, 75015, Paris, France.
| |
Collapse
|
5
|
Roh SH, Mendez-Vazquez H, Sathler MF, Doolittle MJ, Zaytseva A, Brown H, Sainsbury M, Kim S. Prenatal exposure to valproic acid reduces synaptic δ-catenin levels and disrupts ultrasonic vocalization in neonates. Neuropharmacology 2024; 253:109963. [PMID: 38657945 PMCID: PMC11127754 DOI: 10.1016/j.neuropharm.2024.109963] [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: 12/14/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Valproic acid (VPA) is an effective and commonly prescribed drug for epilepsy and bipolar disorder. However, children born from mothers treated with VPA during pregnancy exhibit an increased incidence of autism spectrum disorder (ASD). Although VPA may impair brain development at the cellular level, the mechanism of VPA-induced ASD has not been completely addressed. A previous study has found that VPA treatment strongly reduces δ-catenin mRNA levels in cultured human neurons. δ-catenin is important for the control of glutamatergic synapses and is strongly associated with ASD. VPA inhibits dendritic morphogenesis in developing neurons, an effect that is also found in neurons lacking δ-catenin expression. We thus hypothesize that prenatal exposure to VPA significantly reduces δ-catenin levels in the brain, which impairs glutamatergic synapses to cause ASD. Here, we found that prenatal exposure to VPA markedly reduced δ-catenin levels in the brain of mouse pups. VPA treatment also impaired dendritic branching in developing mouse cortical neurons, which was partially reversed by elevating δ-catenin expression. Prenatal VPA exposure significantly reduced synaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor levels and postsynaptic density 95 (PSD95) in the brain of mouse pups, indicating dysfunctions in glutamatergic synaptic transmission. VPA exposure also significantly altered ultrasonic vocalization (USV) in newly born pups when they were isolated from their nest. Moreover, VPA-exposed pups show impaired hypothalamic response to isolation, which is required to produce animals' USVs following isolation from the nest. Therefore, these results suggest that VPA-induced ASD pathology can be mediated by the loss of δ-catenin functions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Morgan Sainsbury
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Seonil Kim
- Department of Biomedical Sciences, USA; Molecular, Cellular and Integrative Neurosciences Program, USA.
| |
Collapse
|
6
|
Liu L, Tang L, Wang Y, Liu S, Zhang Y. Expression of ITPR2 regulated by lncRNA-NONMMUT020270.2 in LPS-stimulated HT22 cells. Heliyon 2024; 10:e33491. [PMID: 39040287 PMCID: PMC11260991 DOI: 10.1016/j.heliyon.2024.e33491] [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: 03/04/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/24/2024] Open
Abstract
Background Long non-coding RNA (lncRNA)-NONMMUT020270.2 is downregulated and co-expressed with inositol 1,4,5-trisphosphate receptor type 2 (ITPR2) in the hippocampus of Alzheimer's disease (AD) mice. However, whether the expression of ITPR2 was regulated by lncRNA-NONMMUT020270.2 remains unclear. we aimed to investigate regulating relationship of lncRNA-NONMMUT020270.2 and ITPR2. Methods HT22 cells were firstly transfected with the pcDNA3.1-lncRNA-NONMMUT020270.2 overexpression plasmid or with the lncRNA-NONMMUT020270.2 smart silencer, and then were stimulated with lipopolysaccharide (LPS) for 24h. The mRNA expression levels of lncRNA-NONMMUT020270.2 and ITPR2 were measured by reverse transcription-quantitative PCR. Cell viability was assessed using a Cell Counting Kit 8 assay. The expression of Aβ1-42 was detected by ELISA. The expression levels of p-tau, caspase-1, and inositol trisphosphate receptor (IP3R) proteins were detected by western-blotting. Nuclear morphological changes were detected by Hoechst staining. Flow cytometry and Fluo-3/AM were carried out to determine cell apoptosis and the intracellular Ca2+. Results LPS significantly decreased cell viability, and ITPR2 mRNA and IP3R protein expression levels. While it markedly enhanced the expression levels of p-tau and Aβ1-42, cell apoptosis rate, as well as intracellular Ca2+ concentration (P < 0.05). In addition, lncRNA-NONMMUT020270.2 overexpression significantly increased the expressions levels of ITPR2 mRNA and IP3R protein (P < 0.05), and inhibited expression of p-tau and Aβ1-42, cell apoptosis rate, and reduced intracellular Ca2+ concentration (P < 0.05). By contrast, lncRNA-NONMMUT020270.2 silencing notably downregulated expressions levels of ITPR2 mRNA and IP3R protein (P < 0.05), and elevated expression levels of p-tau and Aβ1-42, cell apoptosis rate, and intracellular Ca2+ concentration (P < 0.05). Conclusion lncRNA-NONMMUT020270.2 was positively correlated with ITPR2 expression in LPS-induced cell. Downregulating the lncRNA-NONMMUT020270.2 and ITPR2 may promote cell apoptosis and increase intracellular Ca2+ concentration.
Collapse
Affiliation(s)
- Lan Liu
- Medical College, Tibet University, Lhasa, Tibet, 850000, People's Republic of China
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Liang Tang
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Yan Wang
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha, 410219, People's Republic of China
| | - Shanling Liu
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yongcang Zhang
- Medical College, Tibet University, Lhasa, Tibet, 850000, People's Republic of China
| |
Collapse
|
7
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
8
|
Roh SH, Mendez-Vazquez H, Sathler MF, Doolittle MJ, Zaytseva A, Brown H, Sainsbury M, Kim S. Prenatal exposure to valproic acid reduces synaptic δ-catenin levels and disrupts ultrasonic vocalization in neonates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571709. [PMID: 38168404 PMCID: PMC10760095 DOI: 10.1101/2023.12.14.571709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Valproic acid (VPA) is an effective and commonly prescribed drug for epilepsy and bipolar disorder. However, children born from mothers treated with VPA during pregnancy exhibit an increased incidence of autism spectrum disorder (ASD). Although VPA may impair brain development at the cellular level, the mechanism of VPA-induced ASD has not been completely addressed. A previous study has found that VPA treatment strongly reduces δ-catenin mRNA levels in cultured human neurons. δ-catenin is important for the control of glutamatergic synapses and is strongly associated with ASD. VPA inhibits dendritic morphogenesis in developing neurons, an effect that is also found in neurons lacking δ-catenin expression. We thus hypothesize that prenatal exposure to VPA significantly reduces δ-catenin levels in the brain, which impairs glutamatergic synapses to cause ASD. Here, we found that prenatal exposure to VPA markedly reduced δ-catenin levels in the brain of mouse pups. VPA treatment also impaired dendritic branching in developing mouse cortical neurons, which was reversed by elevating δ-catenin expression. Prenatal VPA exposure significantly reduced synaptic AMPA receptor levels and postsynaptic density 95 (PSD95) in the brain of mouse pups, indicating dysfunctions in glutamatergic synaptic transmission. VPA exposure also significantly altered ultrasonic vocalization (USV) in newly born pups when they were isolated from their nest. Moreover, VPA-exposed pups show impaired hypothalamic response to isolation, which is required to produce animals' USVs following isolation from the nest. Therefore, these results suggest that VPA-induced ASD pathology can be mediated by the loss of δ-catenin functions. Highlights Prenatal exposure of valproic acid (VPA) in mice significantly reduces synaptic δ-catenin protein and AMPA receptor levels in the pups' brains.VPA treatment significantly impairs dendritic branching in cultured cortical neurons, which is reversed by increased δ-catenin expression.VPA exposed pups exhibit impaired communication such as ultrasonic vocalization.Neuronal activation linked to ultrasonic vocalization is absent in VPA-exposed pups.The loss of δ-catenin functions underlies VPA-induced autism spectrum disorder (ASD) in early childhood.
Collapse
|
9
|
Perversi F, Costa C, Labate A, Lattanzi S, Liguori C, Maschio M, Meletti S, Nobili L, Operto FF, Romigi A, Russo E, Di Bonaventura C. The broad-spectrum activity of perampanel: state of the art and future perspective of AMPA antagonism beyond epilepsy. Front Neurol 2023; 14:1182304. [PMID: 37483446 PMCID: PMC10359664 DOI: 10.3389/fneur.2023.1182304] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/07/2023] [Indexed: 07/25/2023] Open
Abstract
Glutamate is the brain's main excitatory neurotransmitter. Glutamatergic neurons primarily compose basic neuronal networks, especially in the cortex. An imbalance of excitatory and inhibitory activities may result in epilepsy or other neurological and psychiatric conditions. Among glutamate receptors, AMPA receptors are the predominant mediator of glutamate-induced excitatory neurotransmission and dictate synaptic efficiency and plasticity by their numbers and/or properties. Therefore, they appear to be a major drug target for modulating several brain functions. Perampanel (PER) is a highly selective, noncompetitive AMPA antagonist approved in several countries worldwide for treating different types of seizures in various epileptic conditions. However, recent data show that PER can potentially address many other conditions within epilepsy and beyond. From this perspective, this review aims to examine the new preclinical and clinical studies-especially those produced from 2017 onwards-on AMPA antagonism and PER in conditions such as mesial temporal lobe epilepsy, idiopathic and genetic generalized epilepsy, brain tumor-related epilepsy, status epilepticus, rare epileptic syndromes, stroke, sleep, epilepsy-related migraine, cognitive impairment, autism, dementia, and other neurodegenerative diseases, as well as provide suggestions on future research agenda aimed at probing the possibility of treating these conditions with PER and/or other AMPA receptor antagonists.
Collapse
Affiliation(s)
| | - Cinzia Costa
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Neurological Clinic, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Angelo Labate
- Neurophysiopatology and Movement Disorders Clinic, University of Messina, Messina, Italy
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome ‘Tor Vergata”, Rome, Italy
- Epilepsy Center, Neurology Unit, University Hospital “Tor Vergata”, Rome, Italy
| | - Marta Maschio
- Center for Tumor-Related Epilepsy, UOSD Neuro-Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Meletti
- Neurology Department, University Hospital of Modena, Modena, Italy
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio-Emilia, Modena, Italy
| | - Lino Nobili
- Child Neuropsychiatry Unit, IRCCS Istituto G. Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health (DINOGMI), University of Genova, Genova, Italy
| | - Francesca Felicia Operto
- Child and Adolescent Neuropsychiatry Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
- Department of Science of Health, School of Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Andrea Romigi
- Sleep Medicine Center, Neurological Mediterranean Institute IRCCS Neuromed, Pozzilli, Italy
- Psychology Faculty, International Telematic University Uninettuno, Rome, Italy
| | - Emilio Russo
- Department of Science of Health, School of Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Carlo Di Bonaventura
- Epilepsy Unit, Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Zaytseva A, Bouckova E, Wiles MJ, Wustrau MH, Schmidt IG, Mendez-Vazquez H, Khatri L, Kim S. Ketamine's rapid antidepressant effects are mediated by Ca 2+-permeable AMPA receptors. eLife 2023; 12:e86022. [PMID: 37358072 PMCID: PMC10319435 DOI: 10.7554/elife.86022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023] Open
Abstract
Ketamine is shown to enhance excitatory synaptic drive in multiple brain areas, which is presumed to underlie its rapid antidepressant effects. Moreover, ketamine's therapeutic actions are likely mediated by enhancing neuronal Ca2+ signaling. However, ketamine is a noncompetitive NMDA receptor (NMDAR) antagonist that reduces excitatory synaptic transmission and postsynaptic Ca2+ signaling. Thus, it is a puzzling question how ketamine enhances glutamatergic and Ca2+ activity in neurons to induce rapid antidepressant effects while blocking NMDARs in the hippocampus. Here, we find that ketamine treatment in cultured mouse hippocampal neurons significantly reduces Ca2+ and calcineurin activity to elevate AMPA receptor (AMPAR) subunit GluA1 phosphorylation. This phosphorylation ultimately leads to the expression of Ca2+-Permeable, GluA2-lacking, and GluA1-containing AMPARs (CP-AMPARs). The ketamine-induced expression of CP-AMPARs enhances glutamatergic activity and glutamate receptor plasticity in cultured hippocampal neurons. Moreover, when a sub-anesthetic dose of ketamine is given to mice, it increases synaptic GluA1 levels, but not GluA2, and GluA1 phosphorylation in the hippocampus within 1 hr after treatment. These changes are likely mediated by ketamine-induced reduction of calcineurin activity in the hippocampus. Using the open field and tail suspension tests, we demonstrate that a low dose of ketamine rapidly reduces anxiety-like and depression-like behaviors in both male and female mice. However, when in vivo treatment of a CP-AMPAR antagonist abolishes the ketamine's effects on animals' behaviors. We thus discover that ketamine at the low dose promotes the expression of CP-AMPARs via reduction of calcineurin activity, which in turn enhances synaptic strength to induce rapid antidepressant actions.
Collapse
Affiliation(s)
- Anastasiya Zaytseva
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - Evelina Bouckova
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - McKennon J Wiles
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | - Madison H Wustrau
- Department of Biomedical Sciences, Colorado State University,Fort CollinsUnited States
| | - Isabella G Schmidt
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
| | | | - Latika Khatri
- Department of Cell Biology, New York University Grossman School of MedicineNew YorkUnited States
| | - Seonil Kim
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State UniversityFort CollinsUnited States
- Department of Biomedical Sciences, Colorado State University,Fort CollinsUnited States
| |
Collapse
|
11
|
Kumro J, Tripathi A, Lei Y, Sword J, Callahan P, Terry A, Lu XY, Kirov SA, Pillai A, Blake DT. Chronic basal forebrain activation improves spatial memory, boosts neurotrophin receptor expression, and lowers BACE1 and Aβ42 levels in the cerebral cortex in mice. Cereb Cortex 2023; 33:7627-7641. [PMID: 36939283 PMCID: PMC10267632 DOI: 10.1093/cercor/bhad066] [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: 12/19/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 03/21/2023] Open
Abstract
The etiology of Alzheimer's dementia has been hypothesized in terms of basal forebrain cholinergic decline, and in terms of reflecting beta-amyloid neuropathology. To study these different biological elements, we activated the basal forebrain in 5xFAD Alzheimer's model mice and littermates. Mice received 5 months of 1 h per day intermittent stimulation of the basal forebrain, which includes cholinergic projections to the cortical mantle. Then, mice were behaviorally tested followed by tissue analysis. The 5xFAD mice performed worse in water-maze testing than littermates. Stimulated groups learned the water maze better than unstimulated groups. Stimulated groups had 2-3-fold increases in frontal cortex immunoblot measures of the neurotrophin receptors for nerve growth factor and brain-derived neurotrophic factor, and a more than 50% decrease in the expression of amyloid cleavage enzyme BACE1. Stimulation also led to lower Aβ42 in 5xFAD mice. These data support a causal relationship between basal forebrain activation and both neurotrophin activation and reduced Aβ42 generation and accumulation. The observation that basal forebrain activation suppresses Aβ42 accumulation, combined with the known high-affinity antagonism of nicotinic receptors by Aβ42, documents bidirectional antagonism between acetylcholine and Aβ42.
Collapse
Affiliation(s)
- Jacob Kumro
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Ashutosh Tripathi
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, United States
| | - Yun Lei
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Jeremy Sword
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Patrick Callahan
- Department of Pharmacology/Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Alvin Terry
- Department of Pharmacology/Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Xin-yun Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Sergei A Kirov
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Anilkumar Pillai
- Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77054, United States
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA 30904, United States
| | - David T Blake
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
12
|
Liu H, Zhang X, Shi P, Yuan J, Jia Q, Pi C, Chen T, Xiong L, Chen J, Tang J, Yue R, Liu Z, Shen H, Zuo Y, Wei Y, Zhao L. α7 Nicotinic acetylcholine receptor: a key receptor in the cholinergic anti-inflammatory pathway exerting an antidepressant effect. J Neuroinflammation 2023; 20:84. [PMID: 36973813 PMCID: PMC10041767 DOI: 10.1186/s12974-023-02768-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/17/2023] [Indexed: 03/28/2023] Open
Abstract
Depression is a common mental illness, which is related to monoamine neurotransmitters and the dysfunction of the cholinergic, immune, glutamatergic, and neuroendocrine systems. The hypothesis of monoamine neurotransmitters is one of the commonly recognized pathogenic mechanisms of depression; however, the drugs designed based on this hypothesis have not achieved good clinical results. A recent study demonstrated that depression and inflammation were strongly correlated, and the activation of alpha7 nicotinic acetylcholine receptor (α7 nAChR)-mediated cholinergic anti-inflammatory pathway (CAP) in the cholinergic system exhibited good therapeutic effects against depression. Therefore, anti-inflammation might be a potential direction for the treatment of depression. Moreover, it is also necessary to further reveal the key role of inflammation and α7 nAChR in the pathogenesis of depression. This review focused on the correlations between inflammation and depression as well-discussed the crucial role of α7 nAChR in the CAP.
Collapse
Affiliation(s)
- Huiyang Liu
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Xiaomei Zhang
- grid.469520.c0000 0004 1757 8917Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, Institute of Medicinal Chemistry of Chinese Medicine, Chongqing Academy of Chinese Materia Medica, Chongqing, 400065 People’s Republic of China
| | - Peng Shi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jiyuan Yuan
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Qiang Jia
- grid.488387.8Ethics Committee Office, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Chao Pi
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
| | - Tao Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Linjin Xiong
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jinglin Chen
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Jia Tang
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ruxu Yue
- grid.410578.f0000 0001 1114 4286Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000 People’s Republic of China
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Zerong Liu
- Central Nervous System Drug Key Laboratory of Sichuan Province, Sichuan Credit Pharmaceutical CO., Ltd., Luzhou, 646000 Sichuan China
- grid.190737.b0000 0001 0154 0904Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030 China
| | - Hongping Shen
- grid.488387.8Clinical Trial Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ying Zuo
- grid.488387.8Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan China
| | - Yumeng Wei
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| | - Ling Zhao
- grid.488387.8Key Laboratory of Medical Electrophysiology, Ministry of Education, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, No. 182, Chunhui Road, Longmatan District, Luzhou, 646000 Sichuan People’s Republic of China
- grid.410578.f0000 0001 1114 4286Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
- grid.488387.8Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000 Sichuan People’s Republic of China
| |
Collapse
|
13
|
Mendez-Vazquez H, Roach RL, Nip K, Sathler MF, Garver T, Danzman RA, Moseley MC, Roberts JP, Koch ON, Steger AA, Lee R, Arikkath J, Kim S. The autism-associated loss of δ-catenin functions disrupts social behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523372. [PMID: 36711484 PMCID: PMC9882145 DOI: 10.1101/2023.01.12.523372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
δ-catenin is expressed in excitatory synapses and functions as an anchor for the glutamatergic AMPA receptor (AMPAR) GluA2 subunit in the postsynaptic density. The glycine 34 to serine (G34S) mutation in the δ-catenin gene is found in autism spectrum disorder (ASD) patients and induces loss of δ-catenin functions at excitatory synapses, which is presumed to underlie ASD pathogenesis in humans. However, how the G34S mutation causes loss of δ-catenin functions to induce ASD remains unclear. Here, using neuroblastoma cells, we discover that the G34S mutation generates an additional phosphorylation site for glycogen synthase kinase 3β (GSK3β). This promotes δ-catenin degradation and causes the reduction of δ-catenin levels, which likely contributes to the loss of δ-catenin functions. Synaptic δ-catenin and GluA2 levels in the cortex are significantly decreased in mice harboring the δ-catenin G34S mutation. The G34S mutation increases glutamatergic activity in cortical excitatory neurons while it is decreased in inhibitory interneurons, indicating changes in cellular excitation and inhibition. δ-catenin G34S mutant mice also exhibit social dysfunction, a common feature of ASD. Most importantly, inhibition of GSK3β activity reverses the G34S-induced loss of δ-catenin function effects in cells and mice. Finally, using δ-catenin knockout mice, we confirm that δ-catenin is required for GSK3β inhibition-induced restoration of normal social behaviors in δ-catenin G34S mutant animals. Taken together, we reveal that the loss of δ-catenin functions arising from the ASD-associated G34S mutation induces social dysfunction via alterations in glutamatergic activity and that GSK3β inhibition can reverse δ-catenin G34S-induced synaptic and behavioral deficits. Significance Statement δ-catenin is important for the localization and function of glutamatergic AMPA receptors at synapses in many brain regions. The glycine 34 to serine (G34S) mutation in the δ-catenin gene is found in autism patients and results in the loss of δ-catenin functions. δ-catenin expression is also closely linked to other autism-risk genes involved in synaptic structure and function, further implying that it is important for the autism pathophysiology. Importantly, social dysfunction is a key characteristic of autism. Nonetheless, the links between δ-catenin functions and social behaviors are largely unknown. The significance of the current research is thus predicated on filling this gap by discovering the molecular, cellular, and synaptic underpinnings of the role of δ-catenin in social behaviors.
Collapse
|
14
|
Xie D, Deng T, Zhai Z, Sun T, Xu Y. The cellular model for Alzheimer's disease research: PC12 cells. Front Mol Neurosci 2023; 15:1016559. [PMID: 36683856 PMCID: PMC9846650 DOI: 10.3389/fnmol.2022.1016559] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is a common age-related neurodegenerative disease characterized by progressive cognitive decline and irreversible memory impairment. Currently, several studies have failed to fully elucidate AD's cellular and molecular mechanisms. For this purpose, research on related cellular models may propose potential predictive models for the drug development of AD. Therefore, many cells characterized by neuronal properties are widely used to mimic the pathological process of AD, such as PC12, SH-SY5Y, and N2a, especially the PC12 pheochromocytoma cell line. Thus, this review covers the most systematic essay that used PC12 cells to study AD. We depict the cellular source, culture condition, differentiation methods, transfection methods, drugs inducing AD, general approaches (evaluation methods and metrics), and in vitro cellular models used in parallel with PC12 cells.
Collapse
Affiliation(s)
- Danni Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Xiong Y, Ye C, Sun R, Chen Y, Zhong X, Zhang J, Zhong Z, Chen H, Huang M. Disrupted Balance of Gray Matter Volume and Directed Functional Connectivity in Mild Cognitive Impairment and Alzheimer's Disease. Curr Alzheimer Res 2023; 20:161-174. [PMID: 37278043 PMCID: PMC10514512 DOI: 10.2174/1567205020666230602144659] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/11/2023] [Accepted: 04/04/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Alterations in functional connectivity have been demonstrated in Alzheimer's disease (AD), an age-progressive neurodegenerative disorder that affects cognitive function; however, directional information flow has never been analyzed. OBJECTIVE This study aimed to determine changes in resting-state directional functional connectivity measured using a novel approach, granger causality density (GCD), in patients with AD, and mild cognitive impairment (MCI) and explore novel neuroimaging biomarkers for cognitive decline detection. METHODS In this study, structural MRI, resting-state functional magnetic resonance imaging, and neuropsychological data of 48 Alzheimer's Disease Neuroimaging Initiative participants were analyzed, comprising 16 patients with AD, 16 with MCI, and 16 normal controls. Volume-based morphometry (VBM) and GCD were used to calculate the voxel-based gray matter (GM) volumes and directed functional connectivity of the brain. We made full use of voxel-based between-group comparisons of VBM and GCD values to identify specific regions with significant alterations. In addition, Pearson's correlation analysis was conducted between directed functional connectivity and several clinical variables. Furthermore, receiver operating characteristic (ROC) analysis related to classification was performed in combination with VBM and GCD. RESULTS In patients with cognitive decline, abnormal VBM and GCD (involving inflow and outflow of GCD) were noted in default mode network (DMN)-related areas and the cerebellum. GCD in the DMN midline core system, hippocampus, and cerebellum was closely correlated with the Mini- Mental State Examination and Functional Activities Questionnaire scores. In the ROC analysis combining VBM with GCD, the neuroimaging biomarker in the cerebellum was optimal for the early detection of MCI, whereas the precuneus was the best in predicting cognitive decline progression and AD diagnosis. CONCLUSION Changes in GM volume and directed functional connectivity may reflect the mechanism of cognitive decline. This discovery could improve our understanding of the pathology of AD and MCI and provide available neuroimaging markers for the early detection, progression, and diagnosis of AD and MCI.
Collapse
Affiliation(s)
- Yu Xiong
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Chenghui Ye
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Ruxin Sun
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Ying Chen
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Xiaochun Zhong
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jiaqi Zhang
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Zhanhua Zhong
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Hongda Chen
- Department of Traditional Chinese Medicine, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Min Huang
- Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| |
Collapse
|
16
|
Alan E, Kerry Z, Sevin G. Molecular mechanisms of Alzheimer's disease: From therapeutic targets to promising drugs. Fundam Clin Pharmacol 2022; 37:397-427. [PMID: 36576325 DOI: 10.1111/fcp.12861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by cognitive impairment so widespread that it interferes with a person's ability to complete daily activities. AD is becoming increasingly common, and it is estimated that the number of patients will reach 152 million by 2050. Current treatment options for AD are symptomatic and have modest benefits. Therefore, considering the human, social, and economic burden of the disease, the development of drugs with the potential to alter disease progression has become a global priority. In this review, the molecular mechanisms involved in the pathology of AD were evaluated as therapeutic targets. The main aim of the review is to focus on new knowledge about mitochondrial dysfunction, oxidative stress, and neuronal transmission in AD, as well as a range of cellular signaling mechanisms and associated treatments. Important molecular interactions leading to AD were described in amyloid cascade and in tau protein function, oxidative stress, mitochondrial dysfunction, cholinergic and glutamatergic neurotransmission, cAMP-regulatory element-binding protein (CREB), the silent mating type information regulation 2 homolog 1 (SIRT-1), neuroinflammation (glial cells), and synaptic alterations. This review summarizes recent experimental and clinical research in AD pathology and analyzes the potential of therapeutic applications based on molecular disease mechanisms.
Collapse
Affiliation(s)
- Elif Alan
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Zeliha Kerry
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Gulnur Sevin
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
17
|
Tian X, Wang Y, Zhou Y, Wu B, Lu Y, Du J, Wang W, Cai W, Xiao Y. Beta-amyloid Deposition in Biliary Atresia Reduces Liver Regeneration by Inhibiting Energy Metabolism and Mammalian Target of Rapamycin Signaling. Clin Transl Gastroenterol 2022; 13:e00536. [PMID: 36137184 PMCID: PMC10476755 DOI: 10.14309/ctg.0000000000000536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/16/2022] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Biliary atresia (BA) is a devastating obstructive bile duct disease found in newborns. This study aims to investigate the roles and involved mechanisms of beta-amyloid (Aβ) in the pathogenesis of BA. METHODS We examined the distribution of Aβ protein and its precursor in the livers of patients with BA. A murine liver organoid and a zebrafish model were established to investigate the exact roles of Aβ in liver regeneration for BA. RESULTS Both Aβ mRNA and protein significantly increased in livers of infants with BA and deposited around the central vein. In the plasma, Aβ elevated significantly in patients with BA and positively correlated with liver injury progression. In vitro , Aβ treatment induced abnormal morphology and caused impaired growth in liver organoids. Energy metabolism analysis demonstrated Aβ increased aerobic glycolysis and reduced ATP synthase in organoids, in which the mammalian target of rapamycin signaling was suppressed. In vivo , Aβ42 exposure caused liver degeneration in zebrafish larvae. DISCUSSION Aβ depositing in livers of infants with BA reduced the liver regeneration through attenuating mitochondrial respiration and mammalian target of rapamycin signaling.
Collapse
Affiliation(s)
- Xinbei Tian
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute for Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Zhou
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Wu
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Lu
- Shanghai Institute for Pediatric Research, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Du
- Shanghai Institute for Pediatric Research, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weipeng Wang
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute for Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongtao Xiao
- Division of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Institute for Pediatric Research, Shanghai, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Sathler MF, Doolittle MJ, Cockrell JA, Nadalin IR, Hofmann F, VandeWoude S, Kim S. HIV and FIV glycoproteins increase cellular tau pathology via cGMP-dependent kinase II activation. J Cell Sci 2022; 135:jcs259764. [PMID: 35638570 PMCID: PMC9270957 DOI: 10.1242/jcs.259764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
As the development of combination antiretroviral therapy (cART) against human immunodeficiency virus (HIV) drastically improves the lifespan of individuals with HIV, many are now entering the prime age when Alzheimer's disease (AD)-like symptoms begin to manifest. It has been shown that hyperphosphorylated tau, a known AD pathological characteristic, is prematurely increased in the brains of HIV-infected individuals as early as in their 30s and that its levels increase with age. This suggests that HIV infection might lead to accelerated AD phenotypes. However, whether HIV infection causes AD to develop more quickly in the brain is not yet fully determined. Interestingly, we have previously revealed that the viral glycoproteins HIV gp120 and feline immunodeficiency virus (FIV) gp95 induce neuronal hyperexcitation via cGMP-dependent kinase II (cGKII; also known as PRKG2) activation in cultured hippocampal neurons. Here, we use cultured mouse cortical neurons to demonstrate that the presence of HIV gp120 and FIV gp95 are sufficient to increase cellular tau pathology, including intracellular tau hyperphosphorylation and tau release to the extracellular space. We further reveal that viral glycoprotein-induced cellular tau pathology requires cGKII activation. Taken together, HIV infection likely accelerates AD-related tau pathology via cGKII activation.
Collapse
Affiliation(s)
- Matheus F. Sathler
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO 80523, USA
| | - Michael J. Doolittle
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO 80523, USA
| | - James A. Cockrell
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, CO 80523, USA
| | - India R. Nadalin
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO 80523, USA
| | - Franz Hofmann
- Technical University of Munich, Arcisstraße 21, D-80333 Munich, Germany
| | - Sue VandeWoude
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Seonil Kim
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO 80523, USA
- Molecular, Cellular and Integrative Neurosciences Program, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
19
|
B. Szabo A, Cretin B, Gérard F, Curot J, J. Barbeau E, Pariente J, Dahan L, Valton L. Sleep: The Tip of the Iceberg in the Bidirectional Link Between Alzheimer's Disease and Epilepsy. Front Neurol 2022; 13:836292. [PMID: 35481265 PMCID: PMC9035794 DOI: 10.3389/fneur.2022.836292] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
The observation that a pathophysiological link might exist between Alzheimer's disease (AD) and epilepsy dates back to the identification of the first cases of the pathology itself and is now strongly supported by an ever-increasing mountain of literature. An overwhelming majority of data suggests not only a higher prevalence of epilepsy in Alzheimer's disease compared to healthy aging, but also that AD patients with a comorbid epileptic syndrome, even subclinical, have a steeper cognitive decline. Moreover, clinical and preclinical investigations have revealed a marked sleep-related increase in the frequency of epileptic activities. This characteristic might provide clues to the pathophysiological pathways underlying this comorbidity. Furthermore, the preferential sleep-related occurrence of epileptic events opens up the possibility that they might hasten cognitive decline by interfering with the delicately orchestrated synchrony of oscillatory activities implicated in sleep-related memory consolidation. Therefore, we scrutinized the literature for mechanisms that might promote sleep-related epileptic activity in AD and, possibly dementia onset in epilepsy, and we also aimed to determine to what degree and through which processes such events might alter the progression of AD. Finally, we discuss the implications for patient care and try to identify a common basis for methodological considerations for future research and clinical practice.
Collapse
Affiliation(s)
- Anna B. Szabo
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- *Correspondence: Anna B. Szabo
| | - Benjamin Cretin
- Clinical Neuropsychology Unit, Neurology Department, CM2R (Memory Resource and Research Centre), University Hospital of Strasbourg, Strasbourg, France
- CNRS, ICube Laboratory, UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), Team IMIS, University of Strasbourg, Strasbourg, France
- CMRR d'Alsace, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Pôle Tête et Cou, Strasbourg, France
| | - Fleur Gérard
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jonathan Curot
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Emmanuel J. Barbeau
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
| | - Jérémie Pariente
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Toulouse NeuroImaging Center (ToNIC), INSERM-University of Toulouse Paul Sabatier, Toulouse, France
| | - Lionel Dahan
- Centre de Recherches sur la Cognition Animale, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Luc Valton
- Centre de Recherche Cerveau & Cognition (CerCo), UMR 5549, CNRS-UPS, Toulouse, France
- Neurology Department, Hôpital Purpan Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Luc Valton
| |
Collapse
|
20
|
Wang T, Chen Y, Zou Y, Pang Y, He X, Chen Y, Liu Y, Feng W, Zhang Y, Li Q, Shi J, Ding F, Marshall C, Gao J, Xiao M. Locomotor Hyperactivity in the Early-Stage Alzheimer’s Disease-like Pathology of APP/PS1 Mice: Associated with Impaired Polarization of Astrocyte Aquaporin 4. Aging Dis 2022; 13:1504-1522. [PMID: 36186142 PMCID: PMC9466968 DOI: 10.14336/ad.2022.0219] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/19/2022] [Indexed: 12/21/2022] Open
Abstract
Non-cognitive behavioral and psychological symptoms often occur in Alzheimer's disease (AD) patients and mouse models, although the exact neuropathological mechanism remains elusive. Here, we report hyperactivity with significant inter-individual variability in 4-month-old APP/PS1 mice. Pathological analysis revealed that intraneuronal accumulation of amyloid-β (Aβ), c-Fos expression in glutamatergic neurons and activation of astrocytes were more evident in the frontal motor cortex of hyperactive APP/PS1 mice, compared to those with normal activity. Moreover, the hyperactive phenotype was associated with mislocalization of perivascular aquaporin 4 (AQP4) and glymphatic transport impairment. Deletion of the AQP4 gene increased hyperactivity, intraneuronal Aβ load and glutamatergic neuron activation, but did not influence working memory or anxiety-like behaviors of 4-month-old APP/PS1 mice. Together, these results demonstrate that AQP4 mislocalization or deficiency leads to increased intraneuronal Aβ load and neuronal hyperactivity in the motor cortex, which in turn causes locomotor over-activity during the early pathophysiology of APP/PS1 mice. Therefore, improving AQP4 mediated glymphatic clearance may offer a new strategy for early intervention of hyperactivity in the prodromal phase of AD.
Collapse
Affiliation(s)
- Tianqi Wang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yan Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Ying Zou
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Yingting Pang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaoxin He
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yali Chen
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yun Liu
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Weixi Feng
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Yanli Zhang
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Qian Li
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Jingping Shi
- Department of Neurology, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Fengfei Ding
- Department of Pharmacology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Charles Marshall
- College of Health Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY 41701, USA
| | - Junying Gao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Correspondence should be addressed to: Dr. Ming Xiao (E-mail: ) or Dr. Junying Gao (), Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211166, China.
- Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Correspondence should be addressed to: Dr. Ming Xiao (E-mail: ) or Dr. Junying Gao (), Jiangsu Province Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Koike H, Iguchi Y, Sahashi K, Katsuno M. Significance of Oligomeric and Fibrillar Species in Amyloidosis: Insights into Pathophysiology and Treatment. Molecules 2021; 26:molecules26165091. [PMID: 34443678 PMCID: PMC8401015 DOI: 10.3390/molecules26165091] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
Amyloidosis is a term referring to a group of various protein-misfolding diseases wherein normally soluble proteins form aggregates as insoluble amyloid fibrils. How, or whether, amyloid fibrils contribute to tissue damage in amyloidosis has been the topic of debate. In vitro studies have demonstrated the appearance of small globular oligomeric species during the incubation of amyloid beta peptide (Aβ). Nerve biopsy specimens from patients with systemic amyloidosis have suggested that globular structures similar to Aβ oligomers were generated from amorphous electron-dense materials and later developed into mature amyloid fibrils. Schwann cells adjacent to amyloid fibrils become atrophic and degenerative, suggesting that the direct tissue damage induced by amyloid fibrils plays an important role in systemic amyloidosis. In contrast, there is increasing evidence that oligomers, rather than amyloid fibrils, are responsible for cell death in neurodegenerative diseases, particularly Alzheimer’s disease. Disease-modifying therapies based on the pathophysiology of amyloidosis have now become available. Aducanumab, a human monoclonal antibody against the aggregated form of Aβ, was recently approved for Alzheimer’s disease, and other monoclonal antibodies, including gantenerumab, solanezumab, and lecanemab, could also be up for approval. As many other agents for amyloidosis will be developed in the future, studies to develop sensitive clinical scales for identifying improvement and markers that can act as surrogates for clinical scales should be conducted.
Collapse
|
22
|
Sathler MF, Khatri L, Roberts JP, Schmidt IG, Zaytseva A, Kubrusly RCC, Ziff EB, Kim S. Phosphorylation of AMPA receptor subunit GluA1 regulates clathrin-mediated receptor internalization. J Cell Sci 2021; 134:272078. [PMID: 34369573 DOI: 10.1242/jcs.257972] [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: 12/03/2020] [Accepted: 07/29/2021] [Indexed: 11/20/2022] Open
Abstract
Synaptic strength is altered during synaptic plasticity by controlling the number of AMPA receptors (AMPARs) at excitatory synapses. During long-term potentiation and synaptic up-scaling, AMPARs are accumulated at synapses to increase synaptic strength. Neuronal activity leads to phosphorylation of AMPAR subunit GluA1 and subsequent elevation of GluA1 surface expression, either by an increase in receptor forward trafficking to the synaptic membrane or a decrease in receptor internalization. However, the molecular pathways underlying GluA1 phosphorylation-induced elevation of surface AMPAR expression are not completely understood. Here, we employ fluorescence recovery after photobleaching (FRAP) to reveal that phosphorylation of GluA1 Serine 845 (S845) predominantly plays a role in receptor internalization than forward trafficking during synaptic plasticity. Notably, internalization of AMPARs depends upon the clathrin adaptor, AP2, which recruits cargo proteins into endocytic clathrin coated pits. In fact, we further reveal that an increase in GluA1 S845 phosphorylation by two distinct forms of synaptic plasticity diminishes the binding of the AP2 adaptor, reducing internalization, and resulting in elevation of GluA1 surface expression. We thus demonstrate a mechanism of GluA1 phosphorylation-regulated clathrin-mediated internalization of AMPARs.
Collapse
Affiliation(s)
- Matheus F Sathler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA.,Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Latika Khatri
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | | | | | | | - Regina C C Kubrusly
- Neuroscience Program, Department of Physiology and Pharmacology, Rua São João Batista, 187, sala 428, Fluminense Federal University, Niterói, RJ, 24020-005, Brazil
| | - Edward B Ziff
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Seonil Kim
- Department of Biomedical Sciences, 1617 Campus Delivery, Colorado State University, Fort Collins, CO, 80525, USA.,Molecular, Cellular and Integrative Neurosciences Program
| |
Collapse
|
23
|
Zolotarev YA, Mitkevich VA, Shram SI, Adzhubei AA, Tolstova AP, Talibov OB, Dadayan AK, Myasoyedov NF, Makarov AA, Kozin SA. Pharmacokinetics and Molecular Modeling Indicate nAChRα4-Derived Peptide HAEE Goes through the Blood-Brain Barrier. Biomolecules 2021; 11:biom11060909. [PMID: 34207317 PMCID: PMC8234734 DOI: 10.3390/biom11060909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022] Open
Abstract
One of the treatment strategies for Alzheimer's disease (AD) is based on the use of pharmacological agents capable of binding to beta-amyloid (Aβ) and blocking its aggregation in the brain. Previously, we found that intravenous administration of the synthetic tetrapeptide Acetyl-His-Ala-Glu-Glu-Amide (HAEE), which is an analogue of the 35-38 region of the α4 subunit of α4β2 nicotinic acetylcholine receptor and specifically binds to the 11-14 site of Aβ, reduced the development of cerebral amyloidogenesis in a mouse model of AD. In the current study on three types of laboratory animals, we determined the biodistribution and tissue localization patterns of HAEE peptide after single intravenous bolus administration. The pharmacokinetic parameters of HAEE were established using uniformly tritium-labeled HAEE. Pharmacokinetic data provided evidence that HAEE goes through the blood-brain barrier. Based on molecular modeling, a role of LRP1 in receptor-mediated transcytosis of HAEE was proposed. Altogether, the results obtained indicate that the anti-amyloid effect of HAEE, previously found in a mouse model of AD, most likely occurs due to its interaction with Aβ species directly in the brain.
Collapse
Affiliation(s)
- Yurii A. Zolotarev
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Vladimir A. Mitkevich
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Stanislav I. Shram
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Alexei A. Adzhubei
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Anna P. Tolstova
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Oleg B. Talibov
- Department of Clinical Pharmacology, Faculty of Common Medicine, Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia;
| | - Alexander K. Dadayan
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Nikolai F. Myasoyedov
- Department of Physiologically Active Substances Chemistry, Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (S.I.S.); (A.K.D.); (N.F.M.)
| | - Alexander A. Makarov
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
| | - Sergey A. Kozin
- Laboratory of Protein Conformational Polymorphism in Health and Disease, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (Y.A.Z.); (V.A.M.); (A.A.A.); (A.P.T.); (A.A.M.)
- Correspondence: ; Tel.: +7-499-135-98-24
| |
Collapse
|
24
|
Sciaccaluga M, Megaro A, Bellomo G, Ruffolo G, Romoli M, Palma E, Costa C. An Unbalanced Synaptic Transmission: Cause or Consequence of the Amyloid Oligomers Neurotoxicity? Int J Mol Sci 2021; 22:ijms22115991. [PMID: 34206089 PMCID: PMC8199544 DOI: 10.3390/ijms22115991] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/18/2022] Open
Abstract
Amyloid-β (Aβ) 1-40 and 1-42 peptides are key mediators of synaptic and cognitive dysfunction in Alzheimer's disease (AD). Whereas in AD, Aβ is found to act as a pro-epileptogenic factor even before plaque formation, amyloid pathology has been detected among patients with epilepsy with increased risk of developing AD. Among Aβ aggregated species, soluble oligomers are suggested to be responsible for most of Aβ's toxic effects. Aβ oligomers exert extracellular and intracellular toxicity through different mechanisms, including interaction with membrane receptors and the formation of ion-permeable channels in cellular membranes. These damages, linked to an unbalance between excitatory and inhibitory neurotransmission, often result in neuronal hyperexcitability and neural circuit dysfunction, which in turn increase Aβ deposition and facilitate neurodegeneration, resulting in an Aβ-driven vicious loop. In this review, we summarize the most representative literature on the effects that oligomeric Aβ induces on synaptic dysfunction and network disorganization.
Collapse
Affiliation(s)
- Miriam Sciaccaluga
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| | - Alfredo Megaro
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Giovanni Bellomo
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
- IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Michele Romoli
- Neurology Unit, Rimini “Infermi” Hospital—AUSL Romagna, 47923 Rimini, Italy;
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome Sapienza, 00185 Rome, Italy; (G.R.); (E.P.)
| | - Cinzia Costa
- Neurology Clinic, Department of Medicine and Surgery, University of Perugia, Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (A.M.); (G.B.)
- Correspondence: (M.S.); (C.C.); Tel.: +39-0755858180 (M.S.); +39-0755784233 (C.C.)
| |
Collapse
|
25
|
Liu D, de Souza JV, Ahmad A, Bronowska AK. Structure, Dynamics, and Ligand Recognition of Human-Specific CHRFAM7A (Dupα7) Nicotinic Receptor Linked to Neuropsychiatric Disorders. Int J Mol Sci 2021; 22:5466. [PMID: 34067314 PMCID: PMC8196834 DOI: 10.3390/ijms22115466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
Cholinergic α7 nicotinic receptors encoded by the CHRNA7 gene are ligand-gated ion channels directly related to memory and immunomodulation. Exons 5-7 in CHRNA7 can be duplicated and fused to exons A-E of FAR7a, resulting in a hybrid gene known as CHRFAM7A, unique to humans. Its product, denoted herein as Dupα7, is a truncated subunit where the N-terminal 146 residues of the ligand binding domain of the α7 receptor have been replaced by 27 residues from FAM7. Dupα7 negatively affects the functioning of α7 receptors associated with neurological disorders, including Alzheimer's diseases and schizophrenia. However, the stoichiometry for the α7 nicotinic receptor containing dupα7 monomers remains unknown. In this work, we developed computational models of all possible combinations of wild-type α7 and dupα7 pentamers and evaluated their stability via atomistic molecular dynamics and coarse-grain simulations. We assessed the effect of dupα7 subunits on the Ca2+ conductance using free energy calculations. We showed that receptors comprising of four or more dupα7 subunits are not stable enough to constitute a functional ion channel. We also showed that models with dupα7/α7 interfaces are more stable and are less detrimental for the ion conductance in comparison to dupα7/dupα7 interfaces. Based on these models, we used protein-protein docking to evaluate how such interfaces would interact with an antagonist, α-bungarotoxin, and amyloid Aβ42. Our findings show that the optimal stoichiometry of dupα7/α7 functional pentamers should be no more than three dupα7 monomers, in favour of a dupα7/α7 interface in comparison to a homodimer dupα7/dupα7 interface. We also showed that receptors bearing dupα7 subunits are less sensitive to Aβ42 effects, which may shed light on the translational gap reported for strategies focused on nicotinic receptors in 'Alzheimer's disease research.
Collapse
Affiliation(s)
- Danlin Liu
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle NE1 7RU, UK; (D.L.); (J.V.d.S.); (A.A.)
| | - João V. de Souza
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle NE1 7RU, UK; (D.L.); (J.V.d.S.); (A.A.)
| | - Ayaz Ahmad
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle NE1 7RU, UK; (D.L.); (J.V.d.S.); (A.A.)
| | - Agnieszka K. Bronowska
- Chemistry—School of Natural and Environmental Sciences, Newcastle University, Newcastle NE1 7RU, UK; (D.L.); (J.V.d.S.); (A.A.)
- Newcastle University Centre for Cancer, Newcastle University, Newcastle NE1 7RU, UK
| |
Collapse
|
26
|
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: 26] [Impact Index Per Article: 8.7] [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
|
27
|
Huang YR, Liu RT. The Toxicity and Polymorphism of β-Amyloid Oligomers. Int J Mol Sci 2020; 21:E4477. [PMID: 32599696 PMCID: PMC7352971 DOI: 10.3390/ijms21124477] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/26/2022] Open
Abstract
It is widely accepted that β-amyloid oligomers (Aβos) play a key role in the progression of Alzheimer's disease (AD) by inducing neuron damage and cognitive impairment, but Aβos are highly heterogeneous in their size, structure and cytotoxicity, making the corresponding studies tough to carry out. Nevertheless, a number of studies have recently made remarkable progress in the describing the characteristics and pathogenicity of Aβos. We here review the mechanisms by which Aβos exert their neuropathogenesis for AD progression, including receptor binding, cell membrane destruction, mitochondrial damage, Ca2+ homeostasis dysregulation and tau pathological induction. We also summarize the characteristics and pathogenicity such as the size, morphology and cytotoxicity of dimers, trimers, Aβ*56 and spherical oligomers, and suggest that Aβos may play a different role at different phases of AD pathogenesis, resulting in differential consequences on neuronal synaptotoxicity and survival. It is warranted to investigate the temporal sequence of Aβos in AD human brain and examine the relationship between different Aβos and cognitive impairment.
Collapse
Affiliation(s)
- Ya-ru Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China;
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui-tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China;
| |
Collapse
|
28
|
Millan MJ, Dekeyne A, Gobert A, Brocco M, Mannoury la Cour C, Ortuno JC, Watson D, Fone KCF. Dual-acting agents for improving cognition and real-world function in Alzheimer's disease: Focus on 5-HT6 and D3 receptors as hubs. Neuropharmacology 2020; 177:108099. [PMID: 32525060 DOI: 10.1016/j.neuropharm.2020.108099] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023]
Abstract
To date, there are no interventions that impede the inexorable progression of Alzheimer's disease (AD), and currently-available drugs cholinesterase (AChE) inhibitors and the N-Methyl-d-Aspartate receptor antagonist, memantine, offer only modest symptomatic benefit. Moreover, a range of mechanistically-diverse agents (glutamatergic, histaminergic, monoaminergic, cholinergic) have disappointed in clinical trials, alone and/or in association with AChE inhibitors. This includes serotonin (5-HT) receptor-6 antagonists, despite compelling preclinical observations in rodents and primates suggesting a positive influence on cognition. The emphasis has so far been on high selectivity. However, for a multi-factorial disorder like idiopathic AD, 5-HT6 antagonists possessing additional pharmacological actions might be more effective, by analogy to "multi-target" antipsychotics. Based on this notion, drug discovery programmes have coupled 5-HT6 blockade to 5-HT4 agonism and inhibition of AchE. Further, combined 5-HT6/dopamine D3 receptor (D3) antagonists are of especial interest since D3 blockade mirrors 5-HT6 antagonism in exerting broad-based pro-cognitive properties in animals. Moreover, 5-HT6 and dopamine D3 antagonists promote neurocognition and social cognition via both distinctive and convergent actions expressed mainly in frontal cortex, including suppression of mTOR over-activation and reinforcement of cholinergic and glutamatergic transmission. In addition, 5-HT6 blockade affords potential anti-anxiety, anti-depressive and anti-epileptic properties, and antagonising 5-HT6 receptors may be associated with neuroprotective ("disease-modifying") properties. Finally D3 antagonism may counter psychotic episodes and D3 receptors themselves offer a promising hub for multi-target agents. The present article reviews the status of "R and D" into multi-target 5-HT6 and D3 ligands for improved treatment of AD and other neurodegenerative disorders of aging. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France.
| | - Anne Dekeyne
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Alain Gobert
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Mauricette Brocco
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Clotilde Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Jean-Claude Ortuno
- Centre for Excellence in Chemistry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - David Watson
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| | - Kevin C F Fone
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| |
Collapse
|
29
|
Xing H, Andrud KW, Soti F, Rouchaud A, Jahn SC, Lu Z, Cho YH, Habibi S, Corsino P, Slavov S, Rocca JR, Lindstrom JM, Lukas RJ, Kem WR. A Methyl Scan of the Pyrrolidinium Ring of Nicotine Reveals Significant Differences in Its Interactions with α7 and α4 β2 Nicotinic Acetylcholine Receptors. Mol Pharmacol 2020; 98:168-180. [PMID: 32474444 DOI: 10.1124/mol.119.118786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 05/06/2020] [Indexed: 01/15/2023] Open
Abstract
The two major nicotinic acetylcholine receptors (nAChRs) in the brain are the α4β2 and α7 subtypes. A "methyl scan" of the pyrrolidinium ring was used to detect differences in nicotine's interactions with these two receptors. Each methylnicotine was investigated using voltage-clamp and radioligand binding techniques. Methylation at each ring carbon elicited unique changes in nicotine's receptor interactions. Replacing the 1'-N-methyl with an ethyl group or adding a second 1'-N-methyl group significantly reduced interaction with α4β2 but not α7 receptors. The 2'-methylation uniquely enhanced binding and agonist potency at α7 receptors. Although 3'- and 5'-trans-methylations were much better tolerated by α7 receptors than α4β2 receptors, 4'-methylation decreased potency and efficacy at α7 receptors much more than at α4β2 receptors. Whereas cis-5'-methylnicotine lacked agonist activity and displayed a low affinity at both receptors, trans-5'-methylnicotine retained considerable α7 receptor activity. Differences between the two 5'-methylated analogs of the potent pyridyl oxymethylene-bridged nicotine analog A84543 were consistent with what was found for the 5'-methylnicotines. Computer docking of the methylnicotines to the Lymnaea acetylcholine binding protein crystal structure containing two persistent waters predicted most of the changes in receptor affinity that were observed with methylation, particularly the lower affinities of the cis-methylnicotines. The much smaller effects of 1'-, 3'-, and 5'-methylations and the greater effects of 2'- and 4'-methylations on nicotine α7 nAChR interaction might be exploited for the design of new drugs based on the nicotine scaffold. SIGNIFICANCE STATEMENT: Using a comprehensive "methyl scan" approach, we show that the orthosteric binding sites for acetylcholine and nicotine in the two major brain nicotinic acetylcholine receptors interact differently with the pyrrolidinium ring of nicotine, and we suggest reasons for the higher affinity of nicotine for the heteromeric receptor. Potential sites for nicotine structure modification were identified that may be useful in the design of new drugs targeting these receptors.
Collapse
Affiliation(s)
- Hong Xing
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Kristin W Andrud
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Ferenc Soti
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Anne Rouchaud
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Stephan C Jahn
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Ziang Lu
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Yeh-Hyon Cho
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Sophia Habibi
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Patrick Corsino
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Svetoslav Slavov
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - James R Rocca
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Jon M Lindstrom
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - Ron J Lukas
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| | - William R Kem
- Department of Pharmacology and Therapeutics (H.X., K.W.A., F.S., A.R., S.C.J., Z.L., Y.-H.C., S.H., P.C., W.R.K.) and AMRIS, McKnight Brain Institute (J.R.R.), College of Medicine, University of Florida, Gainesville, Florida; National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas (S.S.); Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania (J.M.L.); and Division of Neurobiology, Barrow Neurologic Institute, Phoenix, Arizona (R.J.L.)
| |
Collapse
|
30
|
Houchat JN, Cartereau A, Le Mauff A, Taillebois E, Thany SH. An Overview on the Effect of Neonicotinoid Insecticides on Mammalian Cholinergic Functions through the Activation of Neuronal Nicotinic Acetylcholine Receptors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17093222. [PMID: 32384754 PMCID: PMC7246883 DOI: 10.3390/ijerph17093222] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 01/04/2023]
Abstract
Neonicotinoid insecticides are used worldwide and have been demonstrated as toxic to beneficial insects such as honeybees. Their effectiveness is predominantly attributed to their high affinity for insect neuronal nicotinic acetylcholine receptors (nAChRs). Mammalian neuronal nAChRs are of major importance because cholinergic synaptic transmission plays a key role in rapid neurotransmission, learning and memory processes, and neurodegenerative diseases. Because of the low agonist effects of neonicotinoid insecticides on mammalian neuronal nAChRs, it has been suggested that they are relatively safe for mammals, including humans. However, several lines of evidence have demonstrated that neonicotinoid insecticides can modulate cholinergic functions through neuronal nAChRs. Major studies on the influence of neonicotinoid insecticides on cholinergic functions have been conducted using nicotine low-affinity homomeric α7 and high-affinity heteromeric α4β2 receptors, as they are the most abundant in the nervous system. It has been found that the neonicotinoids thiamethoxam and clothianidin can activate the release of dopamine in rat striatum. In some contexts, such as neurodegenerative diseases, they can disturb the neuronal distribution or induce oxidative stress, leading to neurotoxicity. This review highlights recent studies on the mode of action of neonicotinoid insecticides on mammalian neuronal nAChRs and cholinergic functions.
Collapse
|
31
|
Xing H, Keshwah S, Rouchaud A, Kem WR. A Pharmacological Comparison of Two Isomeric Nicotinic Receptor Agonists: The Marine Toxin Isoanatabine and the Tobacco Alkaloid Anatabine. Mar Drugs 2020; 18:E106. [PMID: 32053997 PMCID: PMC7073524 DOI: 10.3390/md18020106] [Citation(s) in RCA: 8] [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: 12/10/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Many organisms possess "secondary" compounds to avoid consumption or to immobilize prey. While the most abundant or active compounds are initially investigated, more extensive analyses reveal other "minor" compounds with distinctive properties that may also be of biomedical and pharmaceutical significance. Here, we present an initial in vitro investigation of the actions of two isomeric tetrahydropyridyl ring-containing anabasine analogs: isoanatabine, an alkaloid isolated from a marine worm, and anatabine, a relatively abundant minor alkaloid in commercial tobacco plants. Both compounds have a double bond that is distal to the piperidine ring nitrogen of anabasine. Racemic isoanatabine and anatabine were synthesized and their S- and R-enantiomers were isolated by chiral high pressure liquid chromatography (HPLC). Both isoanatabines displayed higher efficacies at α4β2 nicotinic acetylcholine receptors (nAChRs) relative to the anatabines; R-isoanatabine was most potent. Radioligand binding experiments revealed similar α4β2 nAChR binding affinities for the isoanatabines, but R-anatabine affinity was twice that of S-anatabine. While the two anatabines and S-isoanatabine were highly efficacious agonists at α7 nAChRs, R-isoanatabine was only a weak partial agonist. The four compounds share an ability to stimulate both α4β2 and α7 nAChRs, a property that may be useful in developing more efficacious drugs to treat neurodegenerative and other medical disorders.
Collapse
Affiliation(s)
| | | | | | - William R. Kem
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA (S.K.)
| |
Collapse
|