1
|
Nakano M, Tsuchida T, Mitsuishi Y, Nishimura M. Nicotinic acetylcholine receptor activation induces BACE1 transcription via the phosphorylation and stabilization of nuclear SP1. Neurosci Res 2024; 203:28-41. [PMID: 38110001 DOI: 10.1016/j.neures.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Abstract
Epidemiological studies have shown that cigarette smoking increases the risk of Alzheimer disease. However, inconsistent results have been reported regarding the effects of smoking or nicotine on brain amyloid β (Aβ) deposition. In this study, we found that stimulation of the nicotinic acetylcholine receptor (nAChR) increased Aβ production in mouse brains and cultured neuronal cells. nAChR activation triggered the MEK/ERK pathway, which then phosphorylated and stabilized nuclear SP1. Upregulated SP1 acted on two recognition motifs in the BACE1 gene to induce its transcription, resulting in enhanced Aβ production. Mouse brain microdialysis revealed that nAChR agonists increased Aβ levels in the interstitial fluid of the cerebral cortex but caused no delay of Aβ clearance. In vitro assays indicated that nicotine inhibited Aβ aggregation. We also found that nicotine modified the immunoreactivity of anti-Aβ antibodies, possibly through competitive inhibition and Aβ conformation changes. Using anti-Aβ antibody that was carefully selected to avoid these effects, we found that chronic nicotine treatment in Aβ precursor protein knockin mice increased the Aβ content but did not visibly change the aggregated Aβ deposition in the brain. Thus, nicotine influences brain Aβ deposition in the opposite direction, thereby increasing Aβ production and inhibiting Aβ aggregation.
Collapse
Affiliation(s)
- Masaki Nakano
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan.
| | - Tomohiro Tsuchida
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Yachiyo Mitsuishi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Shiga 520-2192, Japan; Department of Neurology, Yoka Municipal Hospital, Hyogo 667-8555, Japan.
| |
Collapse
|
2
|
Balasubramaniam M, Ganne A, Mainali N, Pahal S, Ayyadevara S, Shmookler Reis RJ. Alzheimer's-specific brain amyloid interactome: Neural-network analysis of intra-aggregate crosslinking identifies novel drug targets. iScience 2024; 27:108745. [PMID: 38274404 PMCID: PMC10809092 DOI: 10.1016/j.isci.2023.108745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/15/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by peri-neuronal amyloid plaque and intra-neuronal neurofibrillary tangles. These aggregates are identified by the immunodetection of "seed" proteins (Aβ1-42 and hyperphosphorylated tau, respectively), but include many other proteins incorporated nonrandomly. Using click-chemistry intra-aggregate crosslinking, we previously modeled amyloid "contactomes" in SY5Y-APPSw neuroblastoma cells, revealing that aspirin impedes aggregate growth and complexity. By an analogous strategy, we now construct amyloid-specific aggregate interactomes of AD and age-matched-control hippocampi. Comparing these interactomes reveals AD-specific interactions, from which neural-network (NN) analyses predict proteins with the highest impact on pathogenic aggregate formation and/or stability. RNAi knockdowns of implicated proteins, in C. elegans and human-cell-culture models of AD, validated those predictions. Gene-Ontology meta-analysis of AD-enriched influential proteins highlighted the involvement of mitochondrial and cytoplasmic compartments in AD-specific aggregation. This approach derives dynamic consensus models of aggregate growth and architecture, implicating highly influential proteins as new targets to disrupt amyloid accrual in the AD brain.
Collapse
Affiliation(s)
| | - Akshatha Ganne
- Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Nirjal Mainali
- Bioinformatics Program, University of Arkansas for Medical Sciences and University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Sonu Pahal
- Bioinformatics Program, University of Arkansas for Medical Sciences and University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Srinivas Ayyadevara
- Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- McClellan Veterans Medical Center, Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Robert J. Shmookler Reis
- Department of Geriatrics, Reynolds Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- McClellan Veterans Medical Center, Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| |
Collapse
|
3
|
Regulation of the Soluble Amyloid Precursor Protein α (sAPPα) Levels by Acetylcholinesterase and Brain-Derived Neurotrophic Factor in Lung Cancer Cell Media. Int J Mol Sci 2022; 23:ijms231810746. [PMID: 36142659 PMCID: PMC9500850 DOI: 10.3390/ijms231810746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
In comparing two human lung cancer cells, we previously found lower levels of acetylcholinesterase (AChE) and intact amyloid-β40/42 (Aβ), and higher levels of mature brain-derived neurotrophic factor (mBDNF) in the media of H1299 cells as compared to A549 cell media. In this study, we hypothesized that the levels of soluble amyloid precursor protein α (sAPPα) are regulated by AChE and mBDNF in A549 and H1299 cell media. The levels of sAPPα were higher in the media of H1299 cells. Knockdown of AChE led to increased sAPPα and mBDNF levels and correlated with decreased levels of intact Aβ40/42 in A549 cell media. AChE and mBDNF had opposite effects on the levels of Aβ and sAPPα and were found to operate through a mechanism involving α-secretase activity. Treatment with AChE decreased sAPPα levels and simultaneously increased the levels of intact Aβ40/42 suggesting a role of the protein in shifting APP processing away from the non-amyloidogenic pathway and toward the amyloidogenic pathway, whereas treatment with mBDNF led to opposite effects on those levels. We also show that the levels of sAPPα are regulated by protein kinase C (PKC), extracellular signal-regulated kinase (ERK)1/2, phosphoinositide 3 Kinase (PI3K), but not by protein kinase A (PKA).
Collapse
|
4
|
Alzheimer’s Disease and Toxins Produced by Marine Dinoflagellates: An Issue to Explore. Mar Drugs 2022; 20:md20040253. [PMID: 35447926 PMCID: PMC9029327 DOI: 10.3390/md20040253] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/10/2022] Open
Abstract
This paper examined the toxins naturally produced by marine dinoflagellates and their effects on increases in β-amyloid plaques along with tau protein hyperphosphorylation, both major drivers of Alzheimer’s disease (AD). This approach is in line with the demand for certain natural compounds, namely those produced by marine invertebrates that have the potential to be used in the treatment of AD. Current advances in AD treatment are discussed as well as the main factors that potentially affect the puzzling global AD pattern. This study focused on yessotoxins (YTXs), gymnodimine (GYM), spirolides (SPXs), and gambierol, all toxins that have been shown to reduce β-amyloid plaques and tau hyperphosphorylation, thus preventing the neuronal or synaptic dysfunction that ultimately causes the cell death associated with AD (or other neurodegenerative diseases). Another group of toxins described, okadaic acid (OA) and its derivatives, inhibit protein phosphatase activity, which facilitates the presence of phosphorylated tau proteins. A few studies have used OA to trigger AD in zebrafish, providing an opportunity to test in vivo the effectiveness of new drugs in treating or attenuating AD. Constraints on the production of marine toxins for use in these tests have been considered. Different lines of research are anticipated regarding the action of the two groups of toxins.
Collapse
|
5
|
Ren M, Lotfipour S, Leslie F. Unique effects of nicotine across the lifespan. Pharmacol Biochem Behav 2022; 214:173343. [PMID: 35122768 PMCID: PMC8904294 DOI: 10.1016/j.pbb.2022.173343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/23/2022]
Abstract
Smoking remains the leading cause of preventable death in the United States. Although combustible cigarettes are largely being replaced by tobacco-free products, nicotine use continues to increase in vulnerable populations, including youth, adolescents, and pregnant women. Nicotine exerts unique effects on specific brain regions during distinct developmental periods due to the dynamic expression of nicotinic acetylcholine receptors (nAChRs) throughout the lifespan. Nicotine exposure is a health concern not only for adults but also has neurotoxic effects on the fetus, newborn, child, and adolescent. In this review, we aim to highlight the dynamic roles of nAChRs throughout gestation, adolescence, and adulthood. We also provide clinical and preclinical evidence of the neurodevelopmental, cognitive, and behavioral consequences of nicotine exposure at different developmental periods. This comprehensive review highlights unique effects of nicotine throughout the lifespan to help elucidate interventions and public health measures to protect sensitive populations from nicotine exposure.
Collapse
Affiliation(s)
- Michelle Ren
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA.
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Frances Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
6
|
Genetic deletion of α7 nicotinic acetylcholine receptors induces an age-dependent Alzheimer's disease-like pathology. Prog Neurobiol 2021; 206:102154. [PMID: 34453977 DOI: 10.1016/j.pneurobio.2021.102154] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/29/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
The accumulation of amyloid-beta peptide (Aβ) and the failure of cholinergic transmission are key players in Alzheimer's disease (AD). However, in the healthy brain, Aβ contributes to synaptic plasticity and memory acting through α7 subtype nicotinic acetylcholine receptors (α7nAChRs). Here, we hypothesized that the α7nAChR deletion blocks Aβ physiological function and promotes a compensatory increase in Aβ levels that, in turn, triggers an AD-like pathology. To validate this hypothesis, we studied the age-dependent phenotype of α7 knock out mice. We found that α7nAChR deletion caused an impairment of hippocampal synaptic plasticity and memory at 12 months of age, paralleled by an increase of Amyloid Precursor Protein expression and Aβ levels. This was accompanied by other classical AD features such as a hyperphosphorylation of tau at residues Ser 199, Ser 396, Thr 205, a decrease of GSK-3β at Ser 9, the presence of paired helical filaments and neurofibrillary tangles, neuronal loss and an increase of GFAP-positive astrocytes. Our findings suggest that α7nAChR malfunction might precede Aβ and tau pathology, offering a different perspective to interpret the failure of anti-Aβ therapies against AD and to find novel therapeutical approaches aimed at restoring α7nAChRs-mediated Aβ function at the synapse.
Collapse
|
7
|
Loser D, Hinojosa MG, Blum J, Schaefer J, Brüll M, Johansson Y, Suciu I, Grillberger K, Danker T, Möller C, Gardner I, Ecker GF, Bennekou SH, Forsby A, Kraushaar U, Leist M. Functional alterations by a subgroup of neonicotinoid pesticides in human dopaminergic neurons. Arch Toxicol 2021; 95:2081-2107. [PMID: 33778899 PMCID: PMC8166715 DOI: 10.1007/s00204-021-03031-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/17/2021] [Indexed: 12/16/2022]
Abstract
Neonicotinoid pesticides, originally developed to target the insect nervous system, have been reported to interact with human receptors and to activate rodent neurons. Therefore, we evaluated in how far these compounds may trigger signaling in human neurons, and thus, affect the human adult or developing nervous system. We used SH-SY5Y neuroblastoma cells as established model of nicotinic acetylcholine receptor (nAChR) signaling. In parallel, we profiled dopaminergic neurons, generated from LUHMES neuronal precursor cells, as novel system to study nAChR activation in human post-mitotic neurons. Changes of the free intracellular Ca2+ concentration ([Ca2+]i) were used as readout, and key findings were confirmed by patch clamp recordings. Nicotine triggered typical neuronal signaling responses that were blocked by antagonists, such as tubocurarine and mecamylamine. Pharmacological approaches suggested a functional expression of α7 and non-α7 nAChRs on LUHMES cells. In this novel test system, the neonicotinoids acetamiprid, imidacloprid, clothianidin and thiacloprid, but not thiamethoxam and dinotefuran, triggered [Ca2+]i signaling at 10-100 µM. Strong synergy of the active neonicotinoids (at low micromolar concentrations) with the α7 nAChR-positive allosteric modulator PNU-120596 was observed in LUHMES and SH-SY5Y cells, and specific antagonists fully inhibited such signaling. To provide a third line of evidence for neonicotinoid signaling via nAChR, we studied cross-desensitization: pretreatment of LUHMES and SH-SY5Y cells with active neonicotinoids (at 1-10 µM) blunted the signaling response of nicotine. The pesticides (at 3-30 µM) also blunted the response to the non-α7 agonist ABT 594 in LUHMES cells. These data show that human neuronal cells are functionally affected by low micromolar concentrations of several neonicotinoids. An effect of such signals on nervous system development is a toxicological concern.
Collapse
Affiliation(s)
- Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Maria G Hinojosa
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Jonathan Blum
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Jasmin Schaefer
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | - Markus Brüll
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Ylva Johansson
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Karin Grillberger
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Timm Danker
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- NMI TT GmbH, 72770, Reutlingen, Germany
| | - Clemens Möller
- Life Sciences Faculty, Albstadt-Sigmaringen University, 72488, Sigmaringen, Germany
| | - Iain Gardner
- CERTARA UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Gerhard F Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | | | - Anna Forsby
- Department of Biochemistry and Biophysics, Stockholm University, 106 91, Stockholm, Sweden
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.
| |
Collapse
|
8
|
He W, Tu M, Du Y, Li J, Pang Y, Dong Z. Nicotine Promotes AβPP Nonamyloidogenic Processing via RACK1-Dependent Activation of PKC in SH-SY5Y-AβPP695 Cells. J Alzheimers Dis 2020; 75:451-460. [PMID: 32250310 DOI: 10.3233/jad-200003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Accumulation of amyloid-β (Aβ) peptides, generated from amyloid-β precursor protein (AβPP) amyloidogenic processing, is one of the most salient disease hallmarks of Alzheimer's disease (AD). Nicotine is able to promote α-secretase-mediated AβPP nonamyloidogenic processing and increase the release of sAβPPα and C-terminal fragment of 83 amino acids (C83). However, the potential molecular mechanism remains elusive. OBJECTIVE The aim of the present study was to investigate the effect of nicotine on AβPP processing in SH-SY5Y cells that stably express human Swedish mutant AβPP695 (SH-SY5Y-AβPP695). METHODS The expression of AβPP and its C-terminal fragments including C99, C89, and C83, was measured in SH-SY5Y-AβPP695 cells treated with nicotine for 6 h. Protein kinase C (PKC) antagonist Ro30-8220 or agonist PMA was used to determine the role of PKC in AβPP processing. Lentivirus-mediated shRNA targeting receptor for activated C-kinase 1 (RACK1) gene was added into the media to knockdown RACK1 expression, and then AβPP processing was examined. RESULTS The results showed that 6 h of nicotine exposure increased the expression of α-secretase (ADAM10) and C83 in a dose dependent manner. While the β-secretase (BACE1), AβPP amyloidogenic processing products C89 and C99 as well as Aβ peptides (including Aβ40 and Aβ42) remained unchanged. We also found that nicotine elevated the expression of phosphorylated PKC (P-PKC) and RACK1 on the cytomembrane. PKC antagonist Ro30-8220 treatment prevented the increase of ADAM10 and C83 by nicotine. Genetic knockdown RACK1 significantly inhibited P-PKC, and consequently abolished the increase of ADAM10 and C83 by nicotine. CONCLUSION Taken together, these results indicate that nicotine effectively promotes AβPP nonamyloidogenic processing via RACK1-dependent activation of PKC in SH-SY5Y-AβPP695 cells and could be a potential molecule for AD treatment.
Collapse
Affiliation(s)
- Wenting He
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Man Tu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yehong Du
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Junjie Li
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yayan Pang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhifang Dong
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Alzheimer's Disease Therapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1195:105-116. [PMID: 32468465 DOI: 10.1007/978-3-030-32633-3_15] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) was first described and diagnosed by Dr. Alois Alzheimer in 1906 (Hippius and Neundorfer, Dialogues Clin Neurosc 5:101-108, 2003). According to World Health Organization (WHO), AD is the most common cause of dementia, accounting for as many as 60-70% of senile dementia cases and affecting 47.5 million people worldwide (data from 2015) (Dementia Fact Sheet No 362. http://who.int/mediacentre/factsheets/fs362/en/ ). The median survival time after the onset of dementia ranges from 3.3 to 11.7 years (Todd et al. Int J Geriatr Psychiatry 28:1109-1124, 2013). AD is characterized as a severe, chronic, incurable, and progressive neurodegenerative disorder, associated with memory loss and cognition impairment accompanied by abnormal behavior and personality changes (Godyn et al. Pharmacol Rep 68:127-138, 2016). AD is characterized by neuronal death, which usually correlates with the appearance of key neuropathological changes, including acetylcholine deficiency, glutamate excitotoxicity, extracellular deposition of β-amyloid (Aβ plaques), intracellular neurofibrillary tangles by hyperphosphorylated tau protein deposits, neuroinflammation, and widespread neuronal loss (Godyn et al. Pharmacol Rep 68:127-138, 2016; Graham et al. Annu Rev. Med 68:413-430, 2017). The discovery of the degeneration of cholinergic neurons and the reduction of acetylcholine levels in postmortem studies of patients resulted in the use of drugs that leads to the increase of acetylcholine levels in brain (Dubois et al. Lacet Neurol 13:614-629, 2014). At present there is no preventative or curative treatment that interferes with the development of the disease. However, in recent years progress was made in the development of cholinergic drugs which have a positive effect on disease progression. Nowadays, specific drugs that can inhibit the enzyme that degrades acetylcholine are used. The development of new effective drugs involves a difficult and time-consuming process, accompanied by a very high failure rate. In the absence of effective therapies, the estimated number of people with dementia will reach 115 to 131, five million by 2050 (Dubois et al. Lacet Neurol 13:614-629, 2014; Cummings et al. Alzheimers Res Ther 6:37, 2014). Novel therapies and new targets required for developing more effective drugs for the treatment of AD patients are urgently needed.
Collapse
|
10
|
Lanni C, Fagiani F, Racchi M, Preda S, Pascale A, Grilli M, Allegri N, Govoni S. Beta-amyloid short- and long-term synaptic entanglement. Pharmacol Res 2019; 139:243-260. [DOI: 10.1016/j.phrs.2018.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/06/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
|
11
|
Sharma R, Razdan K, Bansal Y, Kuhad A. Rollercoaster ride of kynurenines: steering the wheel towards neuroprotection in Alzheimer's disease. Expert Opin Ther Targets 2018; 22:849-867. [PMID: 30223691 DOI: 10.1080/14728222.2018.1524877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is associated with cerebral cognitive deficits exhibiting two cardinal hallmarks: accruement of extracellular amyloid plaques and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein. The currently accessible therapeutic armamentarium merely provides symptomatic relief. Therefore, the cry for prospective neuroprotective strategies seems to be the need of the hour. Areas covered: This review comprehensively establishes correlation between kynurenine pathway (KP) metabolites and AD with major emphasis on its two functionally contrasting neuroactive metabolites i.e. kynurenic acid (KYNA) and quinolinic acid (QUIN) and enlists various clinical studies which hold a potential for future therapeutics in AD. Also, major hypotheses of AD and mechanisms underlying them have been scrutinized with the aim to brush up the readers with basic pathology of AD. Expert opinion: KP is unique in itself as it holds two completely different domains i.e. neurotoxic QUIN and neuroprotective KYNA and disrupted equilibrium between the two has a hand in neurodegeneration. KYNA has long been demonstrated to be neuroprotective but lately being disparaged for cognitive side effects. But we blaze a trail by amalgamating the pharmacological mechanistic studies of KYNA in kinship with α7nAChRs, NMDARs and GABA which lends aid in favour of KA.
Collapse
Affiliation(s)
- Radhika Sharma
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Karan Razdan
- b Pharmaceutics division , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Yashika Bansal
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| | - Anurag Kuhad
- a Pharmacology Research Laboratory , University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University , Chandigarh , India
| |
Collapse
|
12
|
Shmukler YB, Nikishin DA. Ladder-Shaped Ion Channel Ligands: Current State of Knowledge. Mar Drugs 2017; 15:E232. [PMID: 28726749 PMCID: PMC5532674 DOI: 10.3390/md15070232] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/07/2017] [Accepted: 07/14/2017] [Indexed: 12/20/2022] Open
Abstract
Ciguatoxins (CTX) and brevetoxins (BTX) are polycyclic ethereal compounds biosynthesized by the worldwide distributed planktonic and epibenthic dinoflagellates of Gambierdiscus and Karenia genera, correspondingly. Ciguatera, evoked by CTXs, is a type of ichthyosarcotoxism, which involves a variety of gastrointestinal and neurological symptoms, while BTXs cause so-called neurotoxic shellfish poisoning. Both types of toxins are reviewed together because of similar mechanisms of their action. These are the only molecules known to activate voltage-sensitive Na⁺-channels in mammals through a specific interaction with site 5 of its α-subunit and may compete for it, which results in an increase in neuronal excitability, neurotransmitter release and impairment of synaptic vesicle recycling. Most marine ciguatoxins potentiate Nav channels, but a considerable number of them, such as gambierol and maitotoxin, have been shown to affect another ion channel. Although the extrinsic function of these toxins is probably associated with the function of a feeding deterrent, it was suggested that their intrinsic function is coupled with the regulation of photosynthesis via light-harvesting complex II and thioredoxin. Antagonistic effects of BTXs and brevenal may provide evidence of their participation as positive and negative regulators of this mechanism.
Collapse
Affiliation(s)
- Yuri B Shmukler
- Group of Embryophysiology, N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26, Vavilov st, 119334 Moscow, Russia.
| | - Denis A Nikishin
- Group of Embryophysiology, N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26, Vavilov st, 119334 Moscow, Russia.
| |
Collapse
|
13
|
Cao G, Su P, Zhang S, Guo L, Zhang H, Liang Y, Qin C, Zhang W. Ginsenoside Re reduces Aβ production by activating PPARγ to inhibit BACE1 in N2a/APP695 cells. Eur J Pharmacol 2016; 793:101-108. [PMID: 27840193 DOI: 10.1016/j.ejphar.2016.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/23/2016] [Accepted: 11/03/2016] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by β-amyloid protein (Aβ) deposition. Reducing the Aβ load may be a new perspective for AD treatment. Ginsenoside Re is an extract from Panax notoginseng, which is a well-known traditional Chinese medicine that has been used for the treatment of various diseases for years. Ginsenoside Re has been reported to decrease Aβ in Alzheimer's disease animal models, but the mechanism has not been fully elucidated. In the present study, we investigated the mechanism of ginsenoside Re. Our results showed that ginsenoside Re decreased the Aβ levels in N2a/APP695 cells. Aβ peptides are generated by β-secretase (β-site amyloid precursor protein cleaving enzyme 1 (BACE1)) and γ-secretase. We found that ginsenoside Re decreased the BACE1 mRNA and protein levels and inhibited BACE1 activity in the N2a/APP695 cells. Peroxisome proliferator-activated receptor-γ (PPARγ) is a transcription factor that regulates the activity of the BACE1 promoter, and activating PPARγ can inhibit BACE1. The results also showed that ginsenoside Re significantly increased the PPARγ protein and mRNA levels. These effects of ginsenoside Re on BACE1 could be effectively inhibited by the PPARγ antagonist GW9662. These findings indicate that ginsenoside Re inhibits BACE1 through activation of PPARγ, which ultimately reduces the generation of Aβ1-40 and Aβ1-42. Therefore, ginsenoside Re may be a promising agent for the modulation of Aβ-related pathology in AD.
Collapse
Affiliation(s)
- Guoqiong Cao
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Ping Su
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Shuai Zhang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Limin Guo
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Haijing Zhang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Yuexia Liang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Chunxia Qin
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China
| | - Wensheng Zhang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100088, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100088, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China; National & Local United Engineering Research Center for Sanqi Resources Protection and Utilization Technology, Kunming 650000, China.
| |
Collapse
|
14
|
Nalivaeva NN, Turner AJ. AChE and the amyloid precursor protein (APP) – Cross-talk in Alzheimer's disease. Chem Biol Interact 2016; 259:301-306. [DOI: 10.1016/j.cbi.2016.04.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/18/2016] [Accepted: 04/04/2016] [Indexed: 01/27/2023]
|
15
|
Ovsepian SV, O'Leary VB, Zaborszky L. Cholinergic Mechanisms in the Cerebral Cortex: Beyond Synaptic Transmission. Neuroscientist 2016; 22:238-51. [PMID: 26002948 PMCID: PMC4681696 DOI: 10.1177/1073858415588264] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Functional overviews of cholinergic mechanisms in the cerebral cortex have traditionally focused on the release of acetylcholine with modulator and transmitter effects. Recently, however, data have emerged that extend the role of acetylcholine and cholinergic innervations to a range of housekeeping and metabolic functions. These include regulation of amyloid precursor protein (APP) processing with production of amyloid β (Aβ) and other APP fragments and control of the phosphorylation of microtubule-associated protein (MAP) tau. Evidence has been also presented for receptor-ligand like interactions of cholinergic receptors with soluble Aβ peptide and MAP tau, with modulator and signaling effects. Moreover, high-affinity binding of Aβ to the neurotrophin receptor p75 (p75NTR) enriched in basalo-cortical cholinergic projections has been implicated in clearance of Aβ and nucleation of amyloid plaques. Here, we critically evaluate these unorthodox cholinergic mechanisms and discuss their role in neuronal physiology and the biology of Alzheimer's disease.
Collapse
Affiliation(s)
- Saak V Ovsepian
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany Faculty of Science and Health, School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Valerie B O'Leary
- Institute of Radiation Biology, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
16
|
RamaKrishnan AM, Sankaranarayanan K. Understanding autoimmunity: The ion channel perspective. Autoimmun Rev 2016; 15:585-620. [PMID: 26854401 DOI: 10.1016/j.autrev.2016.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 01/29/2016] [Indexed: 12/11/2022]
Abstract
Ion channels are integral membrane proteins that orchestrate the passage of ions across the cell membrane and thus regulate various key physiological processes of the living system. The stringently regulated expression and function of these channels hold a pivotal role in the development and execution of various cellular functions. Malfunction of these channels results in debilitating diseases collectively termed channelopathies. In this review, we highlight the role of these proteins in the immune system with special emphasis on the development of autoimmunity. The role of ion channels in various autoimmune diseases is also listed out. This comprehensive review summarizes the ion channels that could be used as molecular targets in the development of new therapeutics against autoimmune disorders.
Collapse
Affiliation(s)
| | - Kavitha Sankaranarayanan
- AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chrompet, Chennai 600 044, India.
| |
Collapse
|
17
|
Gonçalves R, Vasques J, Trindade P, Serfaty C, Campello-Costa P, Faria-Melibeu A. Nicotine-induced plasticity in the retinocollicular pathway: Evidence for involvement of amyloid precursor protein. Neuroscience 2016; 313:1-9. [DOI: 10.1016/j.neuroscience.2015.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 10/22/2022]
|
18
|
Stivala CE, Benoit E, Aráoz R, Servent D, Novikov A, Molgó J, Zakarian A. Synthesis and biology of cyclic imine toxins, an emerging class of potent, globally distributed marine toxins. Nat Prod Rep 2015; 32:411-35. [PMID: 25338021 DOI: 10.1039/c4np00089g] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
From a small group of exotic compounds isolated only two decades ago, Cyclic Imine (CI) toxins have become a major class of marine toxins with global distribution. Their distinct chemical structure, biological mechanism of action, and intricate chemistry ensures that CI toxins will continue to be the subject of fascinating fundamental studies in the broad fields of chemistry, chemical biology, and toxicology. The worldwide occurrence of potent CI toxins in marine environments, their accumulation in shellfish, and chemical stability are important considerations in assessing risk factors for human health. This review article aims to provide an account of chemistry, biology, and toxicology of CI toxins from their discovery to the present day.
Collapse
Affiliation(s)
- Craig E Stivala
- Department of Chemistry, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Kuboyama T, Lee YA, Nishiko H, Tohda C. Inhibition of clathrin-mediated endocytosis prevents amyloid β-induced axonal damage. Neurobiol Aging 2015; 36:1808-19. [DOI: 10.1016/j.neurobiolaging.2015.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 01/05/2015] [Accepted: 02/05/2015] [Indexed: 01/15/2023]
|
20
|
Berk C, Paul G, Sabbagh M. Investigational drugs in Alzheimer's disease: current progress. Expert Opin Investig Drugs 2014; 23:837-46. [PMID: 24702504 DOI: 10.1517/13543784.2014.905542] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Alzheimer's disease is a progressive neurodegenerative disorder affecting millions of people worldwide. Yet, this disease is presently incurable and treatable only in terms of modest delay of symptomatic progression. The need for more effective pharmacological intervention is becoming more pronounced as the patient population increases. AREAS COVERED This paper outlines and evaluates the current landscape of interventions in early phases of clinical study. Data and analysis for this review were procured from PubMed, clinicaltrials.gov, review of posters, abstracts and presentations from American Neurological Association, American Academy of Neurology meetings, Alzheimer's Association International Conference and Clinical Trials on Alzheimer's disease. Keywords and criteria searched included: Phase 0, I, and II trials related to Alzheimer's disease, amyloid-β, anti-tau, monoclonal antibodies and metabolism. EXPERT OPINION The development of novel pharmacological interventions would be more fruitful if multitarget therapies were introduced, and unexplored mechanisms of action were expanded upon. Additionally, there is a rationale for intervening earlier in the disease, perhaps preceding or at the advent of symptoms.
Collapse
Affiliation(s)
- Camryn Berk
- Banner Sun Health Research Institute, The Cleo Roberts Center for Clinical Research , 10515 West Santa Fe Drive, Sun City, AZ 85351 , USA +1 623 832 6500 ; +1 623 832 6504 ;
| | | | | |
Collapse
|
21
|
Yang R, Lu Y, Liu J. Identification of tanshinone IIA as a natural monoacylglycerol lipase inhibitor by combined in silico and in vitro approach. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00186a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tanshinone IIA was identified as a novel natural MAGL inhibitor by an in silico approach combined with in vitro experimental analysis.
Collapse
Affiliation(s)
- Ruolin Yang
- Institute of Medical Science
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
- P. R. China
| | - Yang Lu
- Institute of Medical Science
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
- P. R. China
| | - Jianhua Liu
- Institute of Medical Science
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200025
- P. R. China
| |
Collapse
|
22
|
Chang RCC, Ho YS, Wong S, Gentleman SM, Ng HK. Neuropathology of cigarette smoking. Acta Neuropathol 2014; 127:53-69. [PMID: 24240736 DOI: 10.1007/s00401-013-1210-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/05/2013] [Indexed: 02/02/2023]
Abstract
It is well established that cigarette smoking is hazardous to health and is a risk factor for many chronic diseases. However, its impact on the brain, whether it be from prenatal exposure to maternal cigarette smoking, cerebrovascular disease, Alzheimer's disease (AD) or Parkinson's disease, is still not very clear. Neuroimaging and neuropathological investigations suggest that there are heterogeneous effects of cigarette smoking on the brain. On the one hand, it is quite clear that cigarette smoking causes damage to endothelial cells, resulting in increased risk of cerebrovascular disease. On the other hand, it seems to be associated with different Alzheimer's pathologies in post-mortem brains and experimental models, despite the fact that epidemiological studies clearly indicate a positive correlation between cigarette smoking and increased risk for AD. Interestingly, cigarette smoking appears to be associated with reduced Parkinson's pathology in post-mortem brains. However, although nicotine in cigarettes may have some neuroprotective actions, the effects of all the other toxic compounds in cigarettes cannot be ignored. It is, therefore, our aim to summarize what is known about the neuropathology of cigarette smoking and, in particular, its implications for neurodegenerative diseases.
Collapse
Affiliation(s)
- Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, Department of Anatomy, LKS Faculty of Medicine, The University of Hong Kong, Rm. L1-49, Laboratory Block, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong,
| | | | | | | | | |
Collapse
|
23
|
He N, Wang Z, Wang Y, Shen H, Yin M. ZY-1, a novel nicotinic analog, promotes proliferation and migration of adult hippocampal neural stem/progenitor cells. Cell Mol Neurobiol 2013; 33:1149-57. [PMID: 24057433 DOI: 10.1007/s10571-013-9981-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/24/2013] [Indexed: 12/18/2022]
Abstract
Neural stem/progenitor cells (NSPCs) of the subgranular zone have been implicated in cognitive processes, which represent a potentially important source of regenerative medicine for the treatment of neurodegenerative diseases such as Alzheimer's disease (AD). In our previous studies, ZY-1, a novel nicotinic analog, improved cognitive function in transgenic mice model of AD. However, the effect of ZY-1 on the NSPCs remains unclear. Here, we show that ZY-1 significantly increased proliferation and migration of NSPCs, but failed to affect NSPCs differentiation in vitro. Furthermore, during the proliferative period, ZY-1 enhanced intracellular reactive oxygen species (ROS) levels. Meanwhile, ZY-1 also inhibited the levels of Aβ42-induced ROS. Our data indicate that ZY-1 regulates adult hippocampal neurogenesis in vitro, at least partly due to modulating intracellular ROS levels. These results, taken together with those of our previous studies, suggest that ZY-1 might have a potential therapeutic effect for the treatment of AD.
Collapse
Affiliation(s)
- Na He
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | | | | | | | | |
Collapse
|
24
|
Appleby BS, Nacopoulos D, Milano N, Zhong K, Cummings JL. A review: treatment of Alzheimer's disease discovered in repurposed agents. Dement Geriatr Cogn Disord 2013; 35:1-22. [PMID: 23307039 DOI: 10.1159/000345791] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIMS Many compounds that have already been approved for alternate diagnoses have been studied in relation to Alzheimer's disease (AD). The purpose of this review is to summarize these studies and discuss the rationale and benefits of repurposing drugs for AD treatment. METHODS Studies of drugs related to AD treatment that were relevant to a disease-modifying mechanism of action (MOA) and are already approved by the Food and Drug Administration for non-AD diagnoses were collected from PubMed. RESULTS Many drugs already approved for the treatment of other diseases have been studied in relation to AD treatment. Numerous drugs with known toxicity profiles have the potential to be repurposed as a treatment for AD. CONCLUSION Known MOA, toxicology, and pharmacodynamic profiles would accelerate the process and increase the odds of finding a more timely disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Brian S Appleby
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
25
|
Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer's disease: Past, present and future. Neuropharmacology 2013; 76 Pt A:27-50. [PMID: 23891641 DOI: 10.1016/j.neuropharm.2013.07.004] [Citation(s) in RCA: 526] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 06/26/2013] [Accepted: 07/02/2013] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. The etiology is multifactorial, and pathophysiology of the disease is complex. Data indicate an exponential rise in the number of cases of AD, emphasizing the need for developing an effective treatment. AD also imposes tremendous emotional and financial burden to the patient's family and community. The disease has been studied over a century, but acetylcholinesterase inhibitors and memantine are the only drugs currently approved for its management. These drugs provide symptomatic improvement alone but do less to modify the disease process. The extensive insight into the molecular and cellular pathomechanism in AD over the past few decades has provided us significant progress in the understanding of the disease. A number of novel strategies that seek to modify the disease process have been developed. The major developments in this direction are the amyloid and tau based therapeutics, which could hold the key to treatment of AD in the near future. Several putative drugs have been thoroughly investigated in preclinical studies, but many of them have failed to produce results in the clinical scenario; therefore it is only prudent that lessons be learnt from the past mistakes. The current rationales and targets evaluated for therapeutic benefit in AD are reviewed in this article. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- R Anand
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamilnadu, India.
| | | | | |
Collapse
|
26
|
Nie H, Wang Z, Zhao W, Lu J, Zhang C, Lok K, Wang Y, Shen H, Xu Z, Yin M. New nicotinic analogue ZY-1 enhances cognitive functions in a transgenic mice model of Alzheimer's disease. Neurosci Lett 2013; 537:29-34. [PMID: 23340201 DOI: 10.1016/j.neulet.2013.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/18/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder marked by progressive loss of memory and cognitive function. One of the new approaches for treating AD is direct stimulation of nicotinic acetylcholine receptors (nAChRs) in the brain. α4β2-nAChR agonists have shown promising potential in preclinical cognition models of AD. The present report describes the pharmacological properties of ZY-1, a new nicotinic analogue that activates α4β2-nAChR. We describe in detail the binding profile and pharmacological effects of ZY-1 on transgenic AD mice. ZY-1 has high affinity to α4β2-nAChR. In a Morris water maze test, ZY-1 significantly decreases the escape latency and increases both the times in the platform quadrant and the times of platform crossing of transgenic mice. ZY-1 enhances cognitive functions in transgenic mice models of AD. As a novel nicotinic analogue, ZY-1 deserves further study as a potential candidate against AD.
Collapse
Affiliation(s)
- Huizhen Nie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Paris D, Beaulieu-Abdelahad D, Abdullah L, Bachmeier C, Ait-Ghezala G, Reed J, Verma M, Crawford F, Mullan M. Anti-inflammatory activity of anatabine via inhibition of STAT3 phosphorylation. Eur J Pharmacol 2013. [DOI: 10.1016/j.ejphar.2012.11.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Conotoxins that confer therapeutic possibilities. Mar Drugs 2012; 10:1244-1265. [PMID: 22822370 PMCID: PMC3397437 DOI: 10.3390/md10061244] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/24/2012] [Accepted: 05/24/2012] [Indexed: 12/19/2022] Open
Abstract
Cone snails produce a distinctive repertoire of venom peptides that are used both as a defense mechanism and also to facilitate the immobilization and digestion of prey. These peptides target a wide variety of voltage- and ligand-gated ion channels, which make them an invaluable resource for studying the properties of these ion channels in normal and diseased states, as well as being a collection of compounds of potential pharmacological use in their own right. Examples include the United States Food and Drug Administration (FDA) approved pharmaceutical drug, Ziconotide (Prialt®; Elan Pharmaceuticals, Inc.) that is the synthetic equivalent of the naturally occurring ω-conotoxin MVIIA, whilst several other conotoxins are currently being used as standard research tools and screened as potential therapeutic drugs in pre-clinical or clinical trials. These developments highlight the importance of driving conotoxin-related research. A PubMed query from 1 January 2007 to 31 August 2011 combined with hand-curation of the retrieved articles allowed for the collation of 98 recently identified conotoxins with therapeutic potential which are selectively discussed in this review. Protein sequence similarity analysis tentatively assigned uncharacterized conotoxins to predicted functional classes. Furthermore, conotoxin therapeutic potential for neurodegenerative disorders (NDD) was also inferred.
Collapse
|
29
|
Scerri C, Stewart CA, Balfour DJK, Breen KC. Nicotine modifies in vivo and in vitro rat hippocampal amyloid precursor protein processing in young but not old rats. Neurosci Lett 2012; 514:22-6. [PMID: 22381398 DOI: 10.1016/j.neulet.2012.02.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/08/2012] [Accepted: 02/13/2012] [Indexed: 11/30/2022]
Abstract
Previous studies have shown that administration of nicotine modifies the expression and secretion of amyloid precursor protein (APP) in various cell lines. The present study investigated the extent to which chronic subcutaneous nicotine administration influences APP levels and processing in cerebral cortex, striatum and hippocampus of young and old rat brains. The results showed that constant nicotine infusion (0.25 or 4.00mg/kg/day) increased the levels of particulate APP (APPp) but not secreted APP (APPs) in the hippocampus of young rats in vivo. This response to nicotine was not observed in the striatum or cerebral cortex of young rats or in any of the brain regions examined in old animals. Subsequent in vitro analysis demonstrated that nicotine enhanced the release of APPs from hippocampal slice preparations and that this increase was attenuated by mecamylamine, a non-selective nicotinic acetylcholine receptor (nAChR) antagonist. The in vitro effect of nicotine on APPs was age-related, being only detected from hippocampal slices derived from the young but not the older animals. These results suggest that nicotine modulates APP expression and secretion in the hippocampus and that the responses observed to the drug are age-dependent being only detected in younger rats.
Collapse
Affiliation(s)
- Charles Scerri
- Division of Neuroscience, Medical Research Institute, University of Dundee, Ninewells Hospital & Medical School, Dundee DD1 9SY, UK.
| | | | | | | |
Collapse
|
30
|
Fisher A. Cholinergic modulation of amyloid precursor protein processing with emphasis on M1 muscarinic receptor: perspectives and challenges in treatment of Alzheimer’s disease. J Neurochem 2011; 120 Suppl 1:22-33. [DOI: 10.1111/j.1471-4159.2011.07507.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
31
|
Ginzkey C, Stueber T, Friehs G, Koehler C, Hackenberg S, Richter E, Hagen R, Kleinsasser NH. Analysis of nicotine-induced DNA damage in cells of the human respiratory tract. Toxicol Lett 2011; 208:23-9. [PMID: 22001448 DOI: 10.1016/j.toxlet.2011.09.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 09/12/2011] [Accepted: 09/28/2011] [Indexed: 01/09/2023]
Abstract
Epithelium of the upper and lower airways is a common origin of tobacco-related cancer. The main tobacco alkaloid nicotine may be associated with tumor progression. The potential of nicotine in inducing DNA mutations as a step towards cancer initiation is still controversially discussed. Different subtypes of nicotinic acetylcholine receptors (nAChR) are expressed in human nasal mucosa and a human bronchial cell line representing respiratory mucosa as a possible target for receptor-mediated pathways. In the present study, both cell systems were investigated with respect to DNA damage induced by nicotine and its mechanisms. Specimens of human nasal mucosa were harvested during surgery of the nasal air passage. After enzymatic digestion over night, single cells were exposed to an increasing nicotine concentration between 0.001 mM and 4.0mM. In a second step co-incubation was performed using the antioxidant N-acetylcysteine (NAC) and the nAChR antagonist mecamylamine. DNA damage was assessed using the alkali version of the comet assay. Dose finding experiments for mecamylamine to evaluate the maximal inhibitory effect were performed in the human bronchial cell line BEAS-2B with an increasing mecamylamine concentration and a constant nicotine concentration. The influence of nicotine in the apoptotic pathway was evaluated in BEAS-2B cells with the TUNEL assay combined with flow cytometry. After 1h of nicotine exposure with 0.001, 0.01, 0.1, 1.0 and 4.0mM, significant DNA damage was determined at 1.0mM. Further co-incubation experiments with mecamylamine and NAC were performed using 1.0mM of nicotine. The strongest inhibitory effect was measured at 1.0mM mecamylamine and this concentration was used for co-incubation. Both, the antioxidant NAC at a concentration of 1.0mM, based on the literature, as well as the receptor antagonist were capable of complete inhibition of the nicotine-induced DNA migration in the comet assay. A nicotine-induced increase or decrease in apoptosis as assessed by the TUNEL assay in BEAS-2B could not be detected. These results support the hypothesis that oxidative stress is responsible for nicotine-induced DNA damage. Similar results exist for other antioxidants in different cell systems. The decrease in DNA damage after co-incubation with a nAChR antagonist indicates a receptor-dependent pathway of induction for oxidative stress. Further investigations concerning pathways of receptor-mediated DNA damage via nAChR, the role of reactive oxygen species and apoptosis in this cell system will elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Christian Ginzkey
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, Julius-Maximilian University Wuerzburg, Josef-Schneider-Str. 11, D-97080 Wuerzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Paris D, Beaulieu-Abdelahad D, Bachmeier C, Reed J, Ait-Ghezala G, Bishop A, Chao J, Mathura V, Crawford F, Mullan M. Anatabine lowers Alzheimer's Aβ production in vitro and in vivo. Eur J Pharmacol 2011; 670:384-91. [PMID: 21958873 DOI: 10.1016/j.ejphar.2011.09.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/29/2011] [Accepted: 09/07/2011] [Indexed: 11/30/2022]
Abstract
Brain Aβ accumulation represents a key pathological hallmark in Alzheimer's disease. In this study, we investigated the impact of anatabine, a minor alkaloid present in plants of the Solanacea family on Aβ production in vitro using a cell line overexpressing the human amyloid precursor protein (APP) and in vivo using a transgenic mouse model of Alzheimer's disease. In vitro, anatabine lowers Aβ₁₋₄₀ and Aβ₁₋₄₂ levels in a dose dependent manner and reduces sAPPβ production without impacting sAPPα levels suggesting that anatabine lowers Aβ production by mainly impacting the β-cleavage of APP. Additionally, we show that anatabine lowers NFκB activation at doses that inhibit Aβ production in vitro. Since NFκB is known to regulate BACE-1 expression (the rate limiting enzyme responsible for Aβ production), we determined the impact of anatabine on BACE-1 transcription. We show that anatabine inhibits BACE-1 transcription and reduces BACE-1 protein levels in human neuronal like SHSY-5Y cells suggesting that the Aβ lowering properties of anatabine are mediated via a regulation of BACE-1 expression. In vivo, we show that an acute treatment with anatabine for four days significantly lowers brain soluble Aβ₁₋₄₀ and Aβ₁₋₄₂ levels in a transgenic mouse model of Alzheimer's disease. Altogether our data suggest that anatabine may represent an interesting compound for regulating brain Aβ accumulation.
Collapse
Affiliation(s)
- Daniel Paris
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Alonso E, Vale C, Vieytes MR, Laferla FM, Giménez-Llort L, Botana LM. 13-Desmethyl spirolide-C is neuroprotective and reduces intracellular Aβ and hyperphosphorylated tau in vitro. Neurochem Int 2011; 59:1056-65. [PMID: 21907746 DOI: 10.1016/j.neuint.2011.08.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 08/10/2011] [Accepted: 08/17/2011] [Indexed: 12/22/2022]
Abstract
Spirolides are marine compounds of the cyclic imine group. Although the mechanism of action is not fully elucidated yet, cholinergic (muscarinic and nicotinic) receptors have been proposed as the main targets of these toxins. In this study we examined the effect of 13-desmethyl spirolide-C (SPX) on amyloid-beta (Aβ) accumulation and tau hyperphosphorylation in a neuronal model from triple transgenic mice (3xTg) for Alzheimer disease (AD). In vitro treatment of 3xTg cortical neurons with SPX reduced intracellular Aβ accumulation and the levels of phosphorylated tau. SPX treatment did not affect the steady-state levels of neither the M1 and M2 muscarinic nor the α7 nicotinic acetylcholine receptors (AChRs), while it decreased the amplitude of acetylcholine-evoked responses and increased ACh (acetylcholine) levels in 3xTg neurons. Additionally, SPX treatment decreased the levels of two protein kinases involved in tau phosphorylation, glycogen synthase kinase 3β (GSK-3β) and extracellular-regulated kinase (ERK). Also SPX abolished the glutamate-induced neurotoxicity in both control and 3xTg neurons. The results presented here constitute the first report indicating that exposure of 3xTg neurons to nontoxic concentrations of SPX produces a simultaneous reduction in the main pathological characteristics of AD. In spite of the few reports analyzing the mode of action of the toxin we suggest that SPX could ameliorate AD pathology increasing the intracellular ACh levels and simultaneously diminishing the levels of kinases involved in tau phosphorylation.
Collapse
Affiliation(s)
- Eva Alonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27003 Lugo, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Alonso E, Vale C, Vieytes MR, Laferla FM, Giménez-Llort L, Botana LM. The cholinergic antagonist gymnodimine improves Aβ and tau neuropathology in an in vitro model of Alzheimer disease. Cell Physiol Biochem 2011; 27:783-94. [PMID: 21691095 DOI: 10.1159/000330086] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2011] [Indexed: 12/13/2022] Open
Abstract
Gymnodimine (GYM) is a marine phycotoxin with a macrocyclic imine structure, isolated from extracts of the dinoflagellate Karenia selliformis known to act as a cholinergic antagonist with subtype selectivity. However, no data on the chronic effects of this compound has been reported so far. In this work, we evaluated the effect of long term exposure of cortical neurons to gymnodimine in the progress of Alzheimer disease (AD) pathology in vitro. Treatment of cortical neurons with 50 nM gymnodimine decreased the intracellular amyloid beta (Aβ) accumulation and the levels of the hyperphosphorylated isoforms of tau protein recognized by AT8 and AT100 antibodies. These results are suggested to be mediated by the increase in the inactive isoform of the glycogen synthase kinase-3 (phospho GSK-3 Ser9), the decrease in the levels of the active isoform of the ERK1/2 kinase and the increase in acetylcholine (Ach) synthesis elicited by long term exposure of cortical neurons to the toxin. Moreover, gymnodimine decreased glutamate-induced neurotoxicity in vitro. Altogether these results indicate that the marine phycotoxin gymnodimine may constitute a valuable tool for the development of drugs to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Eva Alonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27003 Lugo, Spain
| | | | | | | | | | | |
Collapse
|
35
|
A comparative study of dynamic NMR spectroscopy in analysis of selected N-alkyl-, N-acyl-, and halogenated cytisine derivatives. J Mol Struct 2011. [DOI: 10.1016/j.molstruc.2010.10.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
36
|
Keowkase R, Aboukhatwa M, Adam BL, Beach JW, Terry AV, Buccafussco JJ, Luo Y. Neuroprotective effects and mechanism of cognitive-enhancing choline analogs JWB 1-84-1 and JAY 2-22-33 in neuronal culture and Caenorhabditis elegans. Mol Neurodegener 2010; 5:59. [PMID: 21162742 PMCID: PMC3017027 DOI: 10.1186/1750-1326-5-59] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/16/2010] [Indexed: 11/12/2022] Open
Abstract
Background Our previous work indicated that novel analogs of choline have cytoprotective effects in vitro that might be useful in neurodegenerative conditions such as Alzheimer's disease (AD). Furthermore, two lead compounds (JWB1-84-1 and JAY2-22-33) from a library of more than 50 improved cognitive performances in a transgenic mouse model of AD. The purpose of these experiments was to more specifically investigate the neuroprotective capabilities of these lead compounds both in vitro and in vivo. Results We used N2a cells which express a Swedish mutation in the amyloid precursor protein and presenilin 1 genes to investigate the effect of JWB1-84-1 and JAY2-22-33 on β-amyloid (Aβ) levels and found that both compounds significantly reduced Aβ levels. JWB1-84-1 and JAY2-22-33 also protected rat primary cortical neurons from Aβ toxicity. Subsequently, we utilized the nematode Caenorhabditis elegans (C. elegans) as an in vivo model organism to identify potential molecular targets of these compounds. In the C. elegans model of Aβ toxicity, human Aβ is expressed intracellularly in the body wall muscle. The expression and subsequent aggregation of Aβ in the muscle leads to progressive paralysis. Conclusion We found that JAY2-22-33 (but not JWB1-84-1) significantly reduced Aβ toxicity by delaying paralysis and this protective effect required both the insulin signaling pathway and nicotinic acetylcholine receptors (nAChRs).
Collapse
Affiliation(s)
- Roongpetch Keowkase
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Nie HZ, Shi S, Lukas RJ, Zhao WJ, Sun YN, Yin M. Activation of α7 nicotinic receptor affects APP processing by regulating secretase activity in SH-EP1-α7 nAChR-hAPP695 cells. Brain Res 2010; 1356:112-20. [PMID: 20708605 DOI: 10.1016/j.brainres.2010.07.110] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 07/30/2010] [Accepted: 07/31/2010] [Indexed: 12/31/2022]
Abstract
Multiple lines of evidence have implicated that nicotinic acetylcholine receptor (nAChR) may be an important therapeutic target for the treatment of Alzheimer's disease (AD). Although there are reports suggesting a link between alpha7 nAChR subtype and AD, there has been little report on the mechanism. The present study investigates whether and how α7 nAChR activation affects APP695 processing in SH-EP1 cell model. Cell line co-expressing α7 nAChR gene and human amyloid precursor protein 695 (hAPP695) gene were constructed by stable transfection. Expression of β-amyloid, α-form of secreted APP (αAPPs) and APP1695 was measured by ELISA, western blotting and real-time PCR respectively. Additionally, α, β, and γ-secretase activities were also analyzed in constructed SH-EP1-α7 nAChR-hAPP695 cell line. The results showed that SH-EP1-α7 nAChR-hAPP695 cell line, expressing both hAPP695 gene and α7 nAChR subtype gene, was constructed successfully. The secreted Aβ was decreased and αAPPs was significantly increased by non-selective nAChR agonist nicotine (10 μM) and specific α7 nAChR agonist GTS-21 (1 μM), and APP expression was not affected. Furthermore, specific α7 nAChR antagonist methyllycaconitine (MLA) reversed the alterations induced by activation of α7 nAChR. CTF-α was increased and CTF-γ was decreased when treated with nicotine (10 μM). In addition, the results of enymatic activity analysis showed that nicotine (1μM) and GTS-21 (0.1, 1 μM) decreased γ-secretase activity, but has no effects on α-secretase activity and β-secretase activity. Our findings demonstrate that, through regulating γ-secretase activity, α7 nAChR activation reduces APP processing in amyloidogenic pathway, and at the same time enhances APP processing in non-amyloidogenic pathway. The constructed SH-EP1-α7 nAChR-hAPP695 cell line might be useful for screening specific nAChR agonists against AD.
Collapse
Affiliation(s)
- Hui Zhen Nie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | | | | | | | | | | |
Collapse
|
38
|
Amyloid precursor protein gene mutated at Swedish 670/671 sites in vitro induces changed expression of nicotinic acetylcholine receptors and neurotoxicity. Neurochem Int 2010; 57:647-54. [PMID: 20696195 DOI: 10.1016/j.neuint.2010.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Revised: 07/16/2010] [Accepted: 08/02/2010] [Indexed: 12/21/2022]
Abstract
In order to investigate the influence of amyloid precursor protein (APP) over-expression on the levels of nicotinic acetylcholine receptors (nAChRs), the pCDNA 3.0 carrying the Swedish 670/671 APP double mutation (APP(SWE)) gene was transfected into human neuroblastoma (SH-SY5Y) cells and primary culture of rat hippocampal neurons. The mRNA level of APP, and nAChR α3, α4 and α7 subunits were detected by real-time PCR, and their corresponding proteins as well as α-secreted APP (αAPPs) by Western blotting. [3H]Epibatidine binding sites were measured by the receptor binding assay. The results showed that significantly concomitant increases in mRNA and protein levels of SH-SY5Y cells and primary cultured neurons transfected with APP(SWE) were observed. Interestingly, a decreased αAPPs level was detected in both cells treated with APP(SWE) transfection. In addition, decreases in mRNA and protein levels of α3 nAChR subunit in SH-SY5Y cells or α4 subunit in primary cultured neurons with APP(SWE) transfection were observed. For α7 nAChR, the increased protein and mRNA levels were found in SH-SY5Y cells and primary cultured neurons with APP(SWE) transfection. The number of cholinergic receptor binding site of [3H]epibatidine was decreased in the SH-SY5Y cells transfected with APP(SWE). Elevations in the activities of AChE and BuChE and in the level of lipid peroxidation were detected in both types of cultured cells transfected with APP(SWE). These results indicated that the over-expression of APP(SWE) gene can influence the expression of nAChRs and resulted in neurotoxicity, in which this process might play an important role in the pathogenesis of Alzheimer's disease.
Collapse
|
39
|
Martorana A, Esposito Z, Koch G. Beyond the cholinergic hypothesis: do current drugs work in Alzheimer's disease? CNS Neurosci Ther 2010; 16:235-45. [PMID: 20560995 DOI: 10.1111/j.1755-5949.2010.00175.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by memory and cognitive loss, and represents the leading cause of dementia in elderly people. Besides the complex biochemical processes involved in the neuronal degeneration (formation of senile plaques containing Abeta peptides, and development of neurofibrillary tangles), other molecular and neurochemical alterations, like cholinergic deficit due to basal forebrain degeneration, also occur. Because acetylcholine has been demonstrated to be involved in cognitive processes, the idea to increase acetylcholine levels to restore cognitive deficits has gained interest (the so-called cholinergic hypothesis). This has led to the development of drugs able to prevent acetylcholine hydrolysis (acetylcholinesterase inhibitors). However, the analysis of clinical efficacy of these drugs in alleviating symptoms of dementia showed unsatisfactory results. Despite such critical opinions on the efficacy of these drugs, it should be said that acetylcholinesterase inhibitors, and for some aspects memantine also, improve memory and other cognitive functions throughout most of the duration of the disease. The pharmacological activity of these drugs suggests an effect beyond the mere increase of acetylcholine levels. These considerations are in agreement with the idea that cognitive decline is the result of a complex and not fully elucidated interplay among different neurotransmitters. The role of each of the neurotransmitters implicated has to be related to a cognitive process and as a consequence to its decline. The current review aims to highlight the positive role of cholinergic drugs in alleviating cognitive deficits during wake as well as sleep. Moreover, we suggest that future therapeutic approaches have to be developed to restore the complex interplay between acetylcholine and other neurotransmitters systems, such as dopamine, serotonin, noradrenaline, or glutamate, that are likely involved in the progressive deterioration of several cognitive functions such as attention, memory, and learning.
Collapse
Affiliation(s)
- Alessandro Martorana
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Roma "Tor Vergata", Rome, Italy.
| | | | | |
Collapse
|
40
|
Stevanović ID, Jovanović MD, Čolić M, Jelenković A, Bokonjić D, Ninković M. Nitric oxide synthase inhibitors protect cholinergic neurons against AlCl3 excitotoxicity in the rat brain. Brain Res Bull 2010; 81:641-6. [DOI: 10.1016/j.brainresbull.2010.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 01/06/2010] [Indexed: 10/20/2022]
|
41
|
The effect of aminoguanidine, an inducible nitric oxide synthase inhibitor, on AlCl3 toxicity in the rat hippocampus. ARCH BIOL SCI 2010. [DOI: 10.2298/abs1004981s] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The presented experiment was carried out to determine the effectiveness of
the inducible nitric oxide synthase inhibitor - aminoguanidine in modulating
the toxicity of aluminum chloride on the nitrite levels, malondialdehyde
concentration, reduced glutathione content, as well as cytochrome c oxidase
activity of Wistar rats. The animals were killed 3 h and 30 days after
treatment and the hippocampus was removed. The biochemical results show that
aluminum acts as a pro-oxidant, while aminoguanidine exerts an antioxidant
action in aluminum chloride-treated animals. We have also applied
immunohistochemical techniques to identify iNOS expression after the
treatment. Our data suggest that aminoguanidine can be effective in the
protection of toxicity induced by aluminum chloride.
Collapse
|
42
|
Ondrejcak T, Klyubin I, Hu NW, Barry AE, Cullen WK, Rowan MJ. Alzheimer's disease amyloid beta-protein and synaptic function. Neuromolecular Med 2009; 12:13-26. [PMID: 19757208 DOI: 10.1007/s12017-009-8091-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 08/25/2009] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized neuropathologically by the deposition of different forms of amyloid beta-protein (A beta) including variable amounts of soluble species that correlate with severity of dementia. The extent of synaptic loss in the brain provides the best morphological correlate of cognitive impairment in clinical AD. Animal research on the pathophysiology of AD has therefore focussed on how soluble A beta disrupts synaptic mechanisms in vulnerable brain regions such as the hippocampus. Synaptic plasticity in the form of persistent activity-dependent increases or decreases in synaptic strength provide a neurophysiological substrate for hippocampal-dependent learning and memory. Acute treatment with human-derived or chemically prepared soluble A beta that contains certain oligomeric assemblies, potently and selectively disrupts synaptic plasticity causing inhibition of long-term potentiation (LTP) and enhancement of long-term depression (LTD) of glutamatergic transmission. Over time these and related actions of A beta have been implicated in reducing synaptic integrity. This review addresses the involvement of neurotransmitter intercellular signaling in mediating or modulating the synaptic plasticity disrupting actions of soluble A beta, with particular emphasis on the different roles of glutamatergic and cholinergic mechanisms. There is growing evidence to support the view that NMDA and possibly nicotinic receptors are critically involved in mediating the disruptive effect of A beta and that targeting muscarinic receptors can indirectly modulate A beta's actions. Such studies should help inform ongoing and future clinical trials of drugs acting through the glutamatergic and cholinergic systems.
Collapse
Affiliation(s)
- Tomas Ondrejcak
- Department of Pharmacology and Therapeutics, Biotechnology Building and Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | | | | | | | | | | |
Collapse
|
43
|
Golde TE, Petrucelli L, Lewis J. Targeting Abeta and tau in Alzheimer's disease, an early interim report. Exp Neurol 2009; 223:252-66. [PMID: 19716367 DOI: 10.1016/j.expneurol.2009.07.035] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/25/2009] [Accepted: 07/23/2009] [Indexed: 12/28/2022]
Abstract
The amyloid beta (Abeta) and tau proteins, which misfold, aggregate, and accumulate in the Alzheimer's disease (AD) brain, are implicated as central factors in a complex neurodegenerative cascade. Studies of mutations that cause early onset AD and promote Abeta accumulation in the brain strongly support the notion that inhibiting Abeta aggregation will prevent AD. Similarly, genetic studies of frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17 MAPT) showing that mutations in the MAPT gene encoding tau lead to abnormal tau accumulation and neurodegeneration. Such genetic studies clearly show that tau dysfunction and aggregation can be central to neurodegeneration, however, most likely in a secondary fashion in relation to AD. Additional pathologic, biochemical, and modeling studies further support the concept that Abeta and tau are prime targets for disease modifying therapies in AD. Treatment strategies aimed at preventing the aggregation and accumulation of Abeta, tau, or both proteins should therefore be theoretically possible, assuming that treatment can be initiated before either irreversible damage is present or downstream, self-sustaining, pathological cascades have been initiated. Herein, we will review recent advances and also potential setbacks with respect to the myriad of therapeutic strategies that are designed to slow down, prevent, or clear the accumulation of either "pathological" Abeta or tau. We will also discuss the need for thoughtful prioritization with respect to clinical development of the preclinically validated modifiers of Abeta and tau pathology. The current number of candidate therapies targeting Abeta is becoming so large that a triage process is clearly needed to insure that resources are invested in a way such that the best candidates for disease modifying therapy are rapidly moved toward clinical trials. Finally, we will discuss the challenges for an appropriate "triage" after potential disease modifying therapies targeting tau and Abeta have entered clinical trials.
Collapse
Affiliation(s)
- Todd E Golde
- Department of Neuroscience, College of Medicine, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL 32224, USA.
| | | | | |
Collapse
|
44
|
Dome P, Lazary J, Kalapos MP, Rihmer Z. Smoking, nicotine and neuropsychiatric disorders. Neurosci Biobehav Rev 2009; 34:295-342. [PMID: 19665479 DOI: 10.1016/j.neubiorev.2009.07.013] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 07/23/2009] [Accepted: 07/30/2009] [Indexed: 12/20/2022]
Abstract
Tobacco smoking is an extremely addictive and harmful form of nicotine (NIC) consumption, but unfortunately also the most prevalent. Although disproportionately high frequencies of smoking and its health consequences among psychiatric patients are widely known, the neurobiological background of this epidemiological association is still obscure. The diverse neuroactive effects of NIC and some other major tobacco smoke constituents in the central nervous system may underlie this association. This present paper summarizes the pharmacology of NIC and its receptors (nAChR) based on a systematic review of the literature. The role of the brain's reward system(s) in NIC addiction and the results of functional and structural neuroimaging studies on smoking-related states and behaviors (i.e. dependence, craving, withdrawal) are also discussed. In addition, the epidemiological, neurobiological, and genetic aspects of smoking in several specific neuropsychiatric disorders are reviewed and the clinical relevance of smoking in these disease states addressed.
Collapse
Affiliation(s)
- Peter Dome
- Department of Clinical and Theoretical Mental Health, Kutvolgyi Clinical Center, Semmelweis University, Faculty of Medicine, Kutvolgyi ut 4, 1125 Budapest, Hungary.
| | | | | | | |
Collapse
|
45
|
Pákáski M, Hugyecz M, Sántha P, Jancsó G, Bjelik A, Domokos Á, Janka Z, Kálmán J. Capsaicin promotes the amyloidogenic route of brain amyloid precursor protein processing. Neurochem Int 2009; 54:426-30. [DOI: 10.1016/j.neuint.2009.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 01/13/2009] [Accepted: 01/20/2009] [Indexed: 01/11/2023]
|
46
|
Hunter S, Friedland RP, Brayne C. Time for a change in the research paradigm for Alzheimer's disease: the value of a chaotic matrix modeling approach. CNS Neurosci Ther 2009; 16:254-62. [PMID: 20002628 DOI: 10.1111/j.1755-5949.2009.00117.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The amyloid cascade hypothesis, based on the genetic data from early onset, familial forms of the disease, has been the dominant model for many years and involves over production and deposition of the beta amyloid protein as causal in the disease process. However, it does not apply very well to the more common, later onset, sporadic form of the disease, where a wider range of factors appear to be involved in disease progression. Over recent years, data illustrating reciprocal interactions between the amyloid precursor protein (APP) and its various metabolites with many factors involved in normal synaptic plasticity have emerged. These feedback relationships have the potential to affect the complex kinase cascades involved in every aspect of neuronal function. Further, data regarding the multiple roles of the presenilins have the potential to allow the over expression and deposition of the amyloid beta protein to be both a cause and consequence of disease progression, with relevance in both sporadic and familial of Alzheimer's disease (AD). Disease progression might be better explained by a chaotic matrix of factors and raises the question again whether AD should be approached as a single entity or as a syndrome, with important consequences for disease identification and treatment.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, Forvie Site, Robinson Way, Cambridge, United Kingdom.
| | | | | |
Collapse
|