1
|
Jagielska A, Sałaciak K, Pytka K. Beyond the blur: Scopolamine's utility and limits in modeling cognitive disorders across sexes - Narrative review. Ageing Res Rev 2024; 104:102635. [PMID: 39653154 DOI: 10.1016/j.arr.2024.102635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
Scopolamine, widely regarded as the gold standard in preclinical studies of memory impairments, acts as a non-selective antagonist of central and peripheral muscarinic receptors. While its application in modeling dementia primarily involves antagonism at the M1 receptor, its non-selective peripheral actions may introduce adverse effects that influence behavioral test outcomes. This review analyzes preclinical findings to consolidate knowledge on scopolamine's use and elucidate potential mechanisms responsible for its amnestic effects. We focused on recognition, spatial, and emotional memory processes, alongside executive functions such as attention, cognitive flexibility, and working memory. The cognitive effects of scopolamine are highly dose-dependent, influenced by factors such as species, age, and sex of subjects. Notably, scopolamine rapidly induces observable memory impairments across species, from fish to rodents and primates, often with deficits that can persist for days. However, the compound's broad action on muscarinic receptors and its peripheral side effects, including pupil dilation and reduced salivation, complicates result interpretation, particularly in tasks requiring visual discrimination or food intake. The review also highlights scopolamine's translational value in modeling dementia and Alzheimer's disease, emphasizing the importance of considering individual factors and task-specific designs. Despite its widespread use, scopolamine's limited specificity for cholinergic dysfunction and inability to fully mimic the complex pathophysiology of cognitive disorders like Alzheimer's and Parkinson's disease point to the need for complementary models. This review aims to guide researchers in using scopolamine for modeling cognitive impairments, ensuring attention to factors impacting experimental outcomes.
Collapse
Affiliation(s)
- Angelika Jagielska
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland; Jagiellonian University Medical College, Doctoral School of Medical and Health Sciences, Krakow, Poland
| | - Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
2
|
Petkova-Kirova P, Anastassova N, Minchev B, Uzunova D, Grigorova V, Tsvetanova E, Georgieva A, Alexandrova A, Stefanova M, Yancheva D, Kalfin R, Tancheva L. Behavioral and Biochemical Effects of an Arylhydrazone Derivative of 5-Methoxyindole-2-Carboxylic Acid in a Scopolamine-Induced Model of Alzheimer's Type Dementia in Rats. Molecules 2024; 29:5711. [PMID: 39683869 DOI: 10.3390/molecules29235711] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) has long proven to be a complex neurodegenerative disorder, with cholinergic dysfunction, oxidative stress, and neuroinflammation being just a few of its pathological features. The complexity of the disease requires a multitargeted treatment covering its many aspects. In the present investigation, an arylhydrazone derivative of 5-methoxyindole-2-carboxylic acid (5MeO), with in vitro strong antioxidant, neuroprotective and monoamine oxidase B-inhibiting effects, was studied in a scopolamine-induced Alzheimer-type dementia in rats. Using behavioral and biochemical methods, we evaluated the effects of 5MeO on learning and memory, and elucidated the mechanisms of these effects. Our experiments demonstrated that 5MeO had a beneficial effect on different types of memory as assessed by the step-through and the Barnes maze tasks. It efficiently restored the decreased by scopolamine brain-derived neurotrophic factor and acetylcholine levels and normalized the increased by scopolamine acetylcholine esterase activity in hippocampus. Most effective 5MeO was in counteracting the induced by scopolamine oxidative stress by decreasing the increased by scopolamine levels of lipid peroxidation and by increasing the reduced by scopolamine catalase activity. Blood biochemical analyses demonstrated a favorable safety profile of 5MeO, prompting further pharmacological studies suggesting 5MeO as a safe and efficient candidate in a multitargeted treatment of AD.
Collapse
Affiliation(s)
- Polina Petkova-Kirova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Neda Anastassova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Borislav Minchev
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Diamara Uzunova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Valya Grigorova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Elina Tsvetanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Almira Georgieva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Albena Alexandrova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
- Department of Physiology and Biochemistry, National Sports Academy, Acad. S. Mladenov Str. 21, 1700 Sofia, Bulgaria
| | - Miroslava Stefanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Building 9, 1113 Sofia, Bulgaria
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health, Healthcare and Sport, South-West University, Ivan Mihailov 66, 2700 Blagoevgrad, Bulgaria
| | - Lyubka Tancheva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 23, 1113 Sofia, Bulgaria
| |
Collapse
|
3
|
Liu C, Wu J, Li M, Gao R, Zhang X, Ye-Lehmann S, Song J, Zhu T, Chen C. Smad7 in the hippocampus contributes to memory impairment in aged mice after anesthesia and surgery. J Neuroinflammation 2023; 20:175. [PMID: 37507781 PMCID: PMC10375636 DOI: 10.1186/s12974-023-02849-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a common neurological complication following anesthesia and surgery. Increasing evidence has demonstrated that neuroinflammation caused by systemic inflammatory responses during the perioperative period is a key factor in the occurrence of POCD. In addition, SMAD family member 7 (Smad7) has been confirmed to play vital roles in the pathogenesis and treatment of inflammatory diseases, such as inflammatory bowel disease. However, whether Smad7 participates in the regulatory process of neuroinflammation and apoptosis in the development of POCD is still unknown. METHODS In this study, a POCD mouse model was constructed by unilateral nephrectomy under anesthesia, and cognitive function was assessed using the fear conditioning test and open field test. The expression of Smad7 at the mRNA and protein levels in the hippocampus 3 days after surgery was examined by qRT-PCR, western blot and immunofluorescence assays. Furthermore, to identify whether the elevation of Smad7 in the hippocampus after unilateral nephrectomy contributes to cognitive impairment, the expression of Smad7 in the hippocampal CA1 region was downregulated by crossing Smad7fl/fl conditional mutant mice and CaMKIIα-Cre line T29-1 transgenic mice or stereotaxic injection of shRNA-Smad7. Inflammation and apoptosis in the hippocampus were assessed by measuring the mRNA levels of typical inflammatory cytokines, including TNF-α, IL-1β, IL-6, CCL2, CXCL1, and CXCL2, and the protein levels of apoptotic proteins, including Bax and Bcl2. In addition, apoptosis in the hippocampus postoperation was investigated by a terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining assay. Finally, western blotting was used to explore how Smad7 mediates inflammation and apoptosis postoperation. RESULTS The results unequivocally revealed that elevated Smad7 in the hippocampal CA1 region significantly inhibited TGF-β signal transduction by blocking Smad2/3 phosphorylation, which enhanced neuroinflammation and apoptosis in the hippocampus and further led to learning and memory impairment after surgery. CONCLUSIONS Our results revealed that Smad7 contributes to cognitive impairment after surgery by enhancing neuroinflammation and apoptosis in the hippocampus and might serve as a promising therapeutic target for the treatment of memory impairment after anesthesia surgery.
Collapse
Affiliation(s)
- Changliang Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiahui Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Gao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xueying Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Shixin Ye-Lehmann
- Diseases and Hormones of the Nervous System, University of Paris-Scalay Bicêtre Hosptial Bât. Grégory Pincus, 80 Rue du Gal Leclerc, Le Kremlin Bicêtre, 94276, CEDEX, Paris, France
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Monash Data Futures Institute, Monash University, VIC, Melbourne, Australia
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
| | - Chan Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Yang YS, He SL, Chen WC, Wang CM, Huang QM, Shi YC, Lin S, He HF. Recent progress on the role of non-coding RNA in postoperative cognitive dysfunction. Front Cell Neurosci 2022; 16:1024475. [PMID: 36313620 PMCID: PMC9608859 DOI: 10.3389/fncel.2022.1024475] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD), especially in elderly patients, is a serious complication characterized by impairment of cognitive and sensory modalities after surgery. The pathogenesis of POCD mainly includes neuroinflammation, neuronal apoptosis, oxidative stress, accumulation of Aβ, and tau hyperphosphorylation; however, the exact mechanism remains unclear. Non-coding RNA (ncRNA) may play an important role in POCD. Some evidence suggests that microRNA, long ncRNA, and circular RNA can regulate POCD-related processes, making them promising biomarkers in POCD diagnosis, treatment, and prognosis. This article reviews the crosstalk between ncRNAs and POCD, and systematically discusses the role of ncRNAs in the pathogenesis and diagnosis of POCD. Additionally, we explored the possible mechanisms of ncRNA-associated POCD, providing new knowledge for developing ncRNA-based treatments for POCD.
Collapse
Affiliation(s)
- Yu-Shen Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shi-Ling He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiao-Mei Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-Chuan Shi
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Yan-Chuan Shi,
| | - Shu Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Shu Lin,
| | - He-fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- He-fan He,
| |
Collapse
|
5
|
Oyovwi MO, Ben-Azu B, Edesiri TP, Victor E, Rotu RA, Ozegbe QEB, Nwangwa EK, Atuadu V, Adebayo OG. Kolaviron abates busulfan-induced episodic memory deficit and testicular dysfunction in rats: The implications for neuroendopathobiological changes during chemotherapy. Biomed Pharmacother 2021; 142:112022. [PMID: 34411912 DOI: 10.1016/j.biopha.2021.112022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 07/28/2021] [Accepted: 08/07/2021] [Indexed: 12/22/2022] Open
Abstract
Busulfan is a popular antileukemia chemotherapeutic alkylating agent widely known to induce variety of serious adverse effects including chemobrain-related cognitive impairments and dysfunction in male reproductive system. Whether kolaviron, a neuro- and repro-active compound obtained from Garcinia kola, with neuroprotective and reproductive-promoting activities, mitigates busulfan-induced cognitive and male reproductive impairments remain unknown. Hence, we investigated the reversal effects of kolaviron on busulfan-induced episodic memory deficit and testicular dysfunction, and its underlying mechanisms in male rats. In the treatment-protocol, rats in groups 1 and 2 received saline (10 mL/kg/p.o./day) and DMSO (10 mL/kg/p.o./day) respectively, group 3 was given kolaviron (200 mg/kg/p.o./day), group 4 received busulfan (50 mg/kg/p.o./day) and group 5 was pretreated with busulfan (50 mg/kg/p.o./day) consecutively for 56 days prior to kolaviron treatment (200 mg/kg/p.o./day) from days 29-56. Episodic memory deficit was assessed using passive avoidance task (PAT). Following euthanization, blood samples, epididymal sperm, testes and brain were harvested and hormonal and neurochemical contents and their metabolizing enzymes were assayed. Kolaviron reversed busulfan-induced episodic cognitive deficit in the PAT. The reduced serotonin, dopamine, noradrenaline concentrations, elevated glutamate levels, acetylcholinesterase, monoamine oxidase-A and B activities were normalized by kolaviron. Kolaviron also reversed the busulfan-induced decreased testicular/body weights and spermatogenesis. Kolaviron abated busulfan-induced changes in androgenic hormones (testosterone, FSH, LH), dehydrogenase enzymes (3ß-HSD and 17ß-HSD), altered sperm-chromatin, sperm-membrane integrity and sperm-acrosomal reaction and capacitation impairments. Our findings suggest that kolaviron could mitigate busulfan-induced episodic memory deficit and dysfunction in male reproductive system via neurochemical modulations and increase testicular androgenic hormones/enzymes in rats.
Collapse
Affiliation(s)
- Mega O Oyovwi
- Department of Hunan Physiology, Achievers University, Owo, Ondo, Nigeria; Department of Human Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta, Nigeria
| | - Benneth Ben-Azu
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Science, College of Health Sciences, Delta State University, Abraka, Delta, Nigeria.
| | - Tesi P Edesiri
- Department of Science Laboratory Technology, Delta State Polytechnic, Ogwash-Uku, Delta, Nigeria
| | - Emojevwe Victor
- Department of Physiology, University of Medical Sciences, Ondo, Ondo, Nigeria
| | - Rume A Rotu
- Department of Physiology, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Queen E B Ozegbe
- Department of Hunan Physiology, Achievers University, Owo, Ondo, Nigeria
| | - Eze K Nwangwa
- Department of Human Physiology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta, Nigeria
| | - Vivian Atuadu
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Medicine, Enugu State University of Science and Technology (ESUT), Enugu, Enugu, Nigeria
| | - Olusegun G Adebayo
- Neurophysiology Unit, Department of Physiology, Faculty of Basic Medical Sciences PAMO University of Medical Sciences, Port Harcourt, Nigeria
| |
Collapse
|
6
|
Kii N, Sawada A, Yoshikawa Y, Tachibana S, Yamakage M. Dexmedetomidine Ameliorates Perioperative Neurocognitive Disorders by Suppressing Monocyte-Derived Macrophages in Mice With Preexisting Traumatic Brain Injury. Anesth Analg 2021; 134:869-880. [PMID: 34319918 DOI: 10.1213/ane.0000000000005699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI) initiates immune responses involving infiltration of monocyte-derived macrophages (MDMs) in the injured brain tissue. These MDMs play a key role in perioperative neurocognitive disorders (PNDs). We tested the hypothesis that preanesthetic treatment with dexmedetomidine (DEX) could suppress infiltration of MDMs into the hippocampus of TBI model mice, ameliorating PND. METHODS We first performed bone marrow transplantation from green fluorescent protein-transgenic mice to C57BL/6 mice to identify MDMs. We used only male mice for homogeneity. Four weeks after transplantation, a controlled cortical impact model of TBI was created using recipient mice. Four weeks after TBI, mice received pretreatment with DEX before general anesthesia (GA). Mice performed the Barnes maze test (8-12 mice/group) 2 weeks after GA and were euthanized for immunohistochemistry (4-5 mice/group) or immunoblotting (7 mice/group) 4 weeks after GA. RESULTS In Barnes maze tests, TBI model mice showed longer primary latency (mean difference, 76.5 [95% confidence interval, 41.4-111.6], P < .0001 versus Naïve), primary path length (431.2 [98.5-763.9], P = .001 versus Naïve), and more primary errors (5.7 [0.62-10.7], P = .017 versus Naïve) than Naïve mice on experimental day 3. Expression of MDMs in the hippocampus was significantly increased in TBI mice compared to Naïve mice (2.1 [0.6-3.7], P = .003 versus Naïve). Expression of monocyte chemotactic protein-1 (MCP1)-positive areas in the hippocampus was significantly increased in TBI mice compared to Naïve mice (0.38 [0.09-0.68], P = .007 versus Naïve). Immunoblotting indicated significantly increased expression of interleukin-1β in the hippocampus in TBI mice compared to Naïve mice (1.59 [0.08-3.1], P = .035 versus Naïve). In contrast, TBI mice pretreated with DEX were rescued from these changes and showed no significant difference from Naïve mice. Yohimbine, an α2 receptor antagonist, mitigated the effects of DEX (primary latency: 68.3 [36.5-100.1], P < .0001 versus TBI-DEX; primary path length: 414.9 [120.0-709.9], P = .0002 versus DEX; primary errors: 6.6 [2.1-11.2], P = .0005 versus TBI-DEX; expression of MDMs: 2.9 [1.4-4.4], P = .0001 versus TBI-DEX; expression of MCP1: 0.4 [0.05-0.67], P = .017 versus TBI-DEX; expression of interleukin-1β: 1.8 [0.34-3.35], P = .01 versus TBI-DEX). CONCLUSIONS Preanesthetic treatment with DEX suppressed infiltration of MDMs in the hippocampus and ameliorated PND in TBI model mice. Preanesthetic treatment with DEX appears to suppress infiltration of MDMs in the hippocampus and may lead to new treatments for PND in patients with a history of TBI.
Collapse
Affiliation(s)
- Natsumi Kii
- From the Department of Anesthesiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | |
Collapse
|
7
|
Dashniani MG, Burjanadze MA, Chkhikvishvili NC, Solomonia RO, Kandashvili M, Naneishvili TL, Beselia GV, Kruashvili LB, Chighladze MR. Modulation of spatial memory and expression of hippocampal neurotransmitter receptors by selective lesion of medial septal cholinergic and GABAergic neurons. Exp Brain Res 2020; 238:2385-2397. [PMID: 32770352 DOI: 10.1007/s00221-020-05889-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
The medial septum (MS) is an important modulator of hippocampal function. The degree of damage in which the particular set of septo-hippocampal projections contributes to the deficits of spatial memory with concomitant changes of hippocampal receptors expression has not been studied till present. Therefore, we investigated spatial memory and the expression level of cholinergic (α7 nACh and M1), GABAergic (α1 subunit of GABAA) and glutamatergic (NR2B subunit of NMDA and GluR 1 subunit of AMPA) receptors in the hippocampus following selective lesions of cholinergic and GABAergic septo-hippocampal projection. Learning process and long-term spatial memory were assessed using a Morris water maze. The obtained results revealed that in contrast to cholinergic lesions, rats with MS GABAergic lesions exhibit a retention deficit in 3 days after training. Western blot analyses revealed the MS cholinergic lesions have significant effect on the expression level of the M1 mACh receptors, while MS GABAergic lesions induce dramatic modulations of hippocampal glutamatergic, cholinergic and GABAergic receptors expression. These results for the first time demonstrated that selective lesions of MS cholinergic and GABAergic neurons differentially affect long-term spatial memory and the memory deficit after MS GABAergic lesion is paralleled with significant changes of hippocampal glutamate, GABA and acetylcholine receptors expression.
Collapse
Affiliation(s)
- Manana G Dashniani
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.
| | - Maia A Burjanadze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Nino C Chkhikvishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Revaz O Solomonia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
- Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Manana Kandashvili
- Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Temur L Naneishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Gela V Beselia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
- Department of Physiology and Pharmacology, Petre Shotadze Tbilisi Medical Academy, 0144, Tbilisi, Georgia
| | - Lali B Kruashvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Mariam R Chighladze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| |
Collapse
|
8
|
Buck SA, Baratta AM, Pocivavsek A. Exposure to elevated embryonic kynurenine in rats: Sex-dependent learning and memory impairments in adult offspring. Neurobiol Learn Mem 2020; 174:107282. [PMID: 32738461 PMCID: PMC7506508 DOI: 10.1016/j.nlm.2020.107282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 12/14/2022]
Abstract
Distinct abnormalities in kynurenine pathway (KP) metabolism have been reported in various psychiatric disorders, including schizophrenia (SZ). Kynurenic acid (KYNA), a neuroactive metabolite of the KP, is elevated in individuals diagnosed with SZ and has been linked to cognitive impairments seen in the disorder. To further understand the role of KYNA in SZ etiology, we developed a prenatal insult model where kynurenine (100 mg/day) is fed to pregnant Wistar rats from embryonic day (ED) 15 to ED 22. As sex differences in the prevalence and severity of SZ have been observed, we presently investigated the impact of prenatal kynurenine exposure on KP metabolism and spatial learning and memory in male and female offspring. Specifically, brain tissue and plasma from offspring (control: ECon; kynurenine-treated: EKyn) in prepuberty (postnatal day (PD) 21), adolescence (PD 32-35), and adulthood (PD 56-85) were collected. Separate cohorts of adult offspring were tested in the Barnes maze to assess hippocampus- and prefrontal cortex-mediated learning and memory. Plasma tryptophan, kynurenine, and KYNA were unchanged between ECon and EKyn offspring across all three ages. Hippocampal and frontal cortex KYNA were elevated in male EKyn offspring only in adulthood, compared to ECon, while brain KYNA levels were unchanged in adult females. Male EKyn offspring were significantly impaired during acquisition of the Barnes maze and during reversal learning in the task. In female EKyn offspring, learning and memory remained relatively intact. Taken together, our data demonstrate that exposure to elevated kynurenine during the last week of gestation results in intriguing sex differences and further support the EKyn model as an attractive tool to study the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Silas A Buck
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Annalisa M Baratta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| |
Collapse
|
9
|
Meden A, Knez D, Malikowska-Racia N, Brazzolotto X, Nachon F, Svete J, Sałat K, Grošelj U, Gobec S. Structure-activity relationship study of tryptophan-based butyrylcholinesterase inhibitors. Eur J Med Chem 2020; 208:112766. [PMID: 32919297 DOI: 10.1016/j.ejmech.2020.112766] [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: 07/20/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 12/19/2022]
Abstract
A series of tryptophan-based selective nanomolar butyrylcholinesterase (BChE) inhibitors was designed and synthesized. Compounds were optimized in terms of potency, selectivity, and synthetic accessibility. The crystal structure of the inhibitor 18 in complex with BChE revealed the molecular basis for its low nanomolar inhibition (IC50 = 2.8 nM). The favourable in vitro results enabled a first-in-animal in vivo efficacy and safety trial, which demonstrated a positive impact on fear-motivated and spatial long-term memory retrieval without any concomitant adverse motor effects. Altogether, this research culminated in a handful of new lead compounds with promising potential for symptomatic treatment of patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Anže Meden
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia
| | - Natalia Malikowska-Racia
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland
| | - Xavier Brazzolotto
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Florian Nachon
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91223, Brétigny sur Orge, France
| | - Jurij Svete
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Večna Pot 113, SI-1000, Ljubljana, Slovenia
| | - Kinga Sałat
- Department of Pharmacodynamics, Chair of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688, Krakow, Poland
| | - Uroš Grošelj
- University of Ljubljana, Faculty of Chemistry and Chemical Technology, Večna Pot 113, SI-1000, Ljubljana, Slovenia.
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
10
|
Macheda T, Roberts KN, Morganti JM, Braun DJ, Bachstetter AD. Optimization and validation of a modified radial-arm water maze protocol using a murine model of mild closed head traumatic brain injury. PLoS One 2020; 15:e0232862. [PMID: 32810143 PMCID: PMC7433858 DOI: 10.1371/journal.pone.0232862] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/27/2020] [Indexed: 11/25/2022] Open
Abstract
Cognitive impairments can be a significant problem after a traumatic brain injury (TBI), which affects millions worldwide each year. There is a need for establish reproducible cognitive assays in rodents to better understand disease mechanisms and to develop therapeutic interventions towards treating TBI-induced impairments. Our goal was to validate and standardize the radial arm water maze (RAWM) test as an assay to screen for cognitive impairments caused by TBI. RAWM is a visuo-spatial learning test, originally designed for use with rats, and later adapted for mice. The present study investigates whether test procedures, such us the presence of extra-maze cues influences learning and memory performance. C57BL/6 mice were tested in an 8-arm RAWM using a four-day protocol. We demonstrated that two days of training, exposing the mice to extra-maze cues and a visible platform, influenced learning and memory performance. Mice that did not receive training performed poorer compared to mice trained. To further validate our RAWM protocol, we used scopolamine. We, also, demonstrated that a single mild closed head injury (CHI) caused deficits in this task at two weeks post-CHI. Our data supported the use of 7 trials per day and a spaced training protocol as key factor to unmask memory impairment following CHI. Here, we provide a detailed standard operating procedure for RAWM test, which can be applied to a variety of mouse models including neurodegenerative diseases and pathology, as well as when pharmacological approaches are used.
Collapse
Affiliation(s)
- Teresa Macheda
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Kelly N. Roberts
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
| | - Josh M. Morganti
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - David J. Braun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
| | - Adam D. Bachstetter
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States of America
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
11
|
Chen WN, Yeong KY. Scopolamine, a Toxin-Induced Experimental Model, Used for Research in Alzheimer’s Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:85-93. [DOI: 10.2174/1871527319666200214104331] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Scopolamine as a drug is often used to treat motion sickness. Derivatives of scopolamine
have also found applications as antispasmodic drugs among others. In neuroscience-related research, it
is often used to induce cognitive disorders in experimental models as it readily permeates the bloodbrain
barrier. In the context of Alzheimer’s disease, its effects include causing cholinergic dysfunction
and increasing amyloid-β deposition, both of which are hallmarks of the disease. Hence, the application
of scopolamine in Alzheimer’s disease research is proven pivotal but seldom discussed. In this review,
the relationship between scopolamine and Alzheimer’s disease will be delineated through an
overall effect of scopolamine administration and its specific mechanisms of action, discussing mainly
its influences on cholinergic function and amyloid cascade. The validity of scopolamine as a model of
cognitive impairment or neurotoxin model will also be discussed in terms of advantages and limitations
with future insights.
Collapse
Affiliation(s)
- Win Ning Chen
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
12
|
Dysregulation of Epigenetic Control Contributes to Schizophrenia-Like Behavior in Ebp1 +/- Mice. Int J Mol Sci 2020; 21:ijms21072609. [PMID: 32283721 PMCID: PMC7178112 DOI: 10.3390/ijms21072609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 11/17/2022] Open
Abstract
Dysregulation of epigenetic machinery can cause a variety of neurological disorders associated with cognitive abnormalities. In the hippocampus of postmortem Schizophrenia (SZ) patients, the most notable finding is the deregulation of GAD67 along with differential regulation of epigenetic factors associated with glutamate decarboxylase 67 (GAD67) expression. As we previously reported, ErbB3-binding protein 1 (EBP1) is a potent epigenetic regulator. EBP1 can induce repression of Dnmt1, a well-studied transcriptional repressor of GAD67. In this study, we investigated whether EBP1 contributes to the regulation of GAD67 expression in the hippocampus, controlling epigenetic machinery. In accordance with SZ-like behaviors in Ebp1(+/−) mice, heterozygous deletion of EBP1 led to a dramatic reduction of GAD67 expression, reflecting an abnormally high level of Dnmt1. Moreover, we found that EBP1 binds to the promoter region of HDAC1, which leads to histone deacetylation of GAD67, and suppresses histone deacetylase 1 (HDAC1) expression, inversely mirroring an unusually high level of HDAC1 in Ebp1(+/−) mice. However, EBP1 mutant (p.Glu 183 Ter) found in SZ patients did not elevate the expression of GAD67, failing to suppress Dnmt1 and/or HDAC1 expression. Therefore, this data supports the hypothesis that a reduced amount of EBP1 may contribute to an etiology of SZ due to a loss of transcriptional inhibition of epigenetic repressors, leading to a decreased expression of GAD67.
Collapse
|
13
|
Santillán-Urquiza MA, Herrera-Ruiz M, Zamilpa A, Jiménez-Ferrer E, Román-Ramos R, Alegría-Herrera EY, Tortoriello J. Galphimia glauca and Natural Galphimines Block Schizophrenia-Like Symptoms Induced with Apomorphine and MK-801 in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:8404258. [PMID: 31428176 PMCID: PMC6679897 DOI: 10.1155/2019/8404258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/07/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Galphimia glauca has been used for many years in Mexican Traditional Medicine to calm "insane people." Triterpenes, known as galphimines, were identified in this species. One of them, Galphimine-B (G-B), acts selectively on dopaminergic neurons by antagonizing the effect of glutamate on NMDA receptors. The objective of this study was to evaluate the effect of G. glauca methanolic extract (GgMeOH), a Galphimine-Rich Fraction (GRF), as well as the galphimines G-A, G-B, and G-E, on the acute psychosis induced by Apomorphine (APO) in mice and on schizophrenia-like symptoms induced by subchronic administration of MK-801. METHOD On the first day, ICR male mice were given GgMeOH, GRF, or one of the galphimines. On day two, animals were treated with APO, and on day 3, they were subjected to behavioral tests. In a second test, MK-801 was administered daily for 28 days. In this case, animals were treated daily with G. glauca products from day 9 to day 28 and then subjected to behavioral tests (passive avoidance test, open field test, forced swimming test, and social interaction test). RESULTS The increased number of stereotyped behaviors and grooming behaviors induced with APO were counteracted by all of the experimental treatments. MK-801 induced an increase in immobility time, which was blocked with G-B; GRF counteracted the decreased social interaction, and GgMeOH and GRF prevented the memory loss induced by MK-801. CONCLUSION G. glauca and their derivatives products (GRF and galphimines) were able to interact with the dopaminergic and glutamatergic drugs and to block different behaviors associated with some of the positive, negative, and cognitive symptoms of induced schizophrenia in mice. It is necessary to continue with this research, in order to identify their mechanism of action.
Collapse
Affiliation(s)
- Mayra Alejandra Santillán-Urquiza
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Enrique Jiménez-Ferrer
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Rubén Román-Ramos
- Laboratorio de Farmacología, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Mexico City, Mexico
| | - Elian Yuritzi Alegría-Herrera
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| | - Jaime Tortoriello
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS), Argentina 1, 62790 Xochitepec, Morelos, Mexico
| |
Collapse
|
14
|
Duggal P, Mehan S. Neuroprotective Approach of Anti-Cancer Microtubule Stabilizers Against Tauopathy Associated Dementia: Current Status of Clinical and Preclinical Findings. J Alzheimers Dis Rep 2019; 3:179-218. [PMID: 31435618 PMCID: PMC6700530 DOI: 10.3233/adr-190125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuronal microtubule (MT) tau protein provides cytoskeleton to neuronal cells and plays a vital role including maintenance of cell shape, intracellular transport, and cell division. Tau hyperphosphorylation mediates MT destabilization resulting in axonopathy and neurotransmitter deficit, and ultimately causing Alzheimer’s disease (AD), a dementing disorder affecting vast geriatric populations worldwide, characterized by the existence of extracellular amyloid plaques and intracellular neurofibrillary tangles in a hyperphosphorylated state. Pre-clinically, streptozotocin stereotaxically mimics the behavioral and biochemical alterations similar to AD associated with tau pathology resulting in MT assembly defects, which proceed neuropathological cascades. Accessible interventions like cholinesterase inhibitors and NMDA antagonist clinically provides only symptomatic relief. Involvement of microtubule stabilizers (MTS) prevents tauopathy particularly by targeting MT oriented cytoskeleton and promotes polymerization of tubulin protein. Multiple in vitro and in vivo research studies have shown that MTS can hold substantial potential for the treatment of AD-related tauopathy dementias through restoration of tau function and axonal transport. Moreover, anti-cancer taxane derivatives and epothiolones may have potential to ameliorate MT destabilization and prevent the neuronal structural and functional alterations associated with tauopathies. Therefore, this current review strictly focuses on exploration of various clinical and pre-clinical features available for AD to understand the neuropathological mechanisms as well as introduce pharmacological interventions associated with MT stabilization. MTS from diverse natural sources continue to be of value in the treatment of cancer, suggesting that these agents have potential to be of interest in the treatment of AD-related tauopathy dementia in the future.
Collapse
Affiliation(s)
- Pallavi Duggal
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
15
|
Manyes L, Holst S, Lozano M, Santos E, Fernandez-Medarde A. Spatial learning and long-term memory impairments in RasGrf1 KO, Pttg1 KO, and double KO mice. Brain Behav 2018; 8:e01089. [PMID: 30259712 PMCID: PMC6236249 DOI: 10.1002/brb3.1089] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/12/2018] [Accepted: 07/01/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND RasGrf1 is a guanine-nucleotide releasing factor that enhances Ras activity. Human PTTG1 is an oncoprotein found in pituitary tumors and later identified as securin, a protein isolated from yeast with a reported role in chromosome separation. It has been suggested that RasGrf1 is an important upstream component of signal transduction pathways regulating Pttg1 expression and controlling beta cell development and their physiological response. At memory formation level, there are contradictory data regarding the role of RasGrf1, while Pttg1 has not been previously studied. Both proteins are expressed in the mammalian hippocampus, which is one of the key brain areas for spatial learning and memory. OBJECTIVE The aim of this work was to study a potential link between RasGrf1 and Pttg1 in memory formation. METHOD Spatial learning and memory test in the Pttg1 KO, RasGrf1 KO, and Pttg1-RasGrf1 double KO and their correspondent WT mice using a Barnes maze. RESULTS In comparison with the WT control mice, Pttg1 KO mice learned how to solve the task in a less efficient way, suggesting problems in memory consolidation. RasGrf1 KO mice performance was similar to controls, and they learned to use the best searching strategy. Double KO mice reached a better spatial learning level than WT. CONCLUSION A role for Pttg1 in memory consolidation/formation is suggested, while our RasGrf1 KO mice do not show hippocampus associated memory defects.
Collapse
Affiliation(s)
- Lara Manyes
- Lab 1, Cancer Research Center, CSIC-Universidad de Salamanca & CIBERONC, Salamanca, Spain.,Laboratory of Food Sciences and Toxicology, Faculty of Pharmacy, Universitat de València, València, Spain
| | - Sarah Holst
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Lozano
- Laboratory of Food Sciences and Toxicology, Faculty of Pharmacy, Universitat de València, València, Spain
| | - Eugenio Santos
- Lab 1, Cancer Research Center, CSIC-Universidad de Salamanca & CIBERONC, Salamanca, Spain
| | | |
Collapse
|