201
|
Chiarini A, Armato U, Liu D, Dal Prà I. Calcium-Sensing Receptors of Human Neural Cells Play Crucial Roles in Alzheimer's Disease. Front Physiol 2016; 7:134. [PMID: 27199760 PMCID: PMC4844916 DOI: 10.3389/fphys.2016.00134] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/28/2016] [Indexed: 12/21/2022] Open
Abstract
In aged subjects, late-onset Alzheimer's disease (LOAD) starts in the lateral entorhinal allocortex where a failure of clearance mechanisms triggers an accumulation of neurotoxic amyloid-β42 oligomers (Aβ42-os). In neurons and astrocytes, Aβ42-os enhance the transcription of Aβ precursor protein (APP) and β-secretase/BACE1 genes. Thus, by acting together with γ-secretase, the surpluses of APP and BACE1 amplify the endogenous production of Aβ42-os which pile up, damage mitochondria, and are oversecreted. At the plasmalemma, exogenous Aβ42-os bind neurons' and astrocytes' calcium-sensing receptors (CaSRs) activating a set of intracellular signaling pathways which upkeep Aβ42-os intracellular accumulation and oversecretion by hindering Aβ42-os proteolysis. In addition, Aβ42-os accumulating in the extracellular milieu spread and reach mounting numbers of adjacent and remoter teams of neurons and astrocytes which in turn are recruited, again via Aβ42-os•CaSR-governed mechanisms, to produce and release additional Aβ42-os amounts. This relentless self-sustaining mechanism drives AD progression toward upper cortical areas. Later on accumulating Aβ42-os elicit the advent of hyperphosphorylated (p)-Tau oligomers which acting together with Aβ42-os and other glial neurotoxins cooperatively destroy wider and wider cognition-related cortical areas. In parallel, Aβ42-os•CaSR signals also elicit an excess production and secretion of nitric oxide and vascular endothelial growth factor-A from astrocytes, of Aβ42-os and myelin basic protein from oligodendrocytes, and of proinflammatory cytokines, nitric oxide and (likely) Aβ42-os from microglia. Activated astrocytes and microglia survive the toxic onslaught, whereas neurons and oligodendrocytes increasingly die. However, we have shown that highly selective allosteric CaSR antagonists (calcilytics), like NPS 2143 and NPS 89626, efficiently suppress all the neurotoxic effects Aβ42-os•CaSR signaling drives in cultured cortical untransformed human neurons and astrocytes. In fact, calcilytics increase Aβ42 proteolysis and discontinue the oversecretion of Aβ42-os, nitric oxide, and vascular endothelial growth factor-A from both astrocytes and neurons. Seemingly, calcilytics would also benefit the other types of glial cells and cerebrovascular cells otherwise damaged by the effects of Aβ42-os•CaSR signaling. Thus, given at amnestic minor cognitive impairment (aMCI) or initial symptomatic stages, calcilytics could prevent or terminate the propagation of LOAD neuropathology and preserve human neurons' viability and hence patients' cognitive abilities.
Collapse
Affiliation(s)
- Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical SchoolVerona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical SchoolVerona, Italy
| | - Daisong Liu
- Human Histology and Embryology Unit, University of Verona Medical SchoolVerona, Italy
- Proteomics Laboratory, Institute for Burn Research, Third Military Medical UniversityChongqing, China
| | - Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical SchoolVerona, Italy
| |
Collapse
|
202
|
Forloni G, Artuso V, La Vitola P, Balducci C. Oligomeropathies and pathogenesis of Alzheimer and Parkinson's diseases. Mov Disord 2016; 31:771-81. [DOI: 10.1002/mds.26624] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/13/2022] Open
Affiliation(s)
- Gianluigi Forloni
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| | | | - Pietro La Vitola
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| | - Claudia Balducci
- Departement of Neuroscience; IRCCS, Istituto di Ricerche Farmacologiche “Mario Negri,”; Milano Italy
| |
Collapse
|
203
|
Lykhmus O, Mishra N, Koval L, Kalashnyk O, Gergalova G, Uspenska K, Komisarenko S, Soreq H, Skok M. Molecular Mechanisms Regulating LPS-Induced Inflammation in the Brain. Front Mol Neurosci 2016; 9:19. [PMID: 27013966 PMCID: PMC4781876 DOI: 10.3389/fnmol.2016.00019] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/23/2016] [Indexed: 01/08/2023] Open
Abstract
Neuro-inflammation, one of the pathogenic causes of neurodegenerative diseases, is regulated through the cholinergic anti-inflammatory pathway via the α7 nicotinic acetylcholine receptor (α7 nAChR). We previously showed that either bacterial lipopolysaccharide (LPS) or immunization with the α7(1-208) nAChR fragment decrease α7 nAChRs density in the mouse brain, exacerbating chronic inflammation, beta-amyloid accumulation and episodic memory decline, which mimic the early stages of Alzheimer's disease (AD). To study the molecular mechanisms underlying the LPS and antibody effects in the brain, we employed an in vivo model of acute LPS-induced inflammation and an in vitro model of cultured glioblastoma U373 cells. Here, we report that LPS challenge decreased the levels of α7 nAChR RNA and protein and of acetylcholinesterase (AChE) RNA and activity in distinct mouse brain regions, sensitized brain mitochondria to the apoptogenic effect of Ca(2+) and modified brain microRNA profiles, including the cholinergic-regulatory CholinomiRs-132/212, in favor of anti-inflammatory and pro-apoptotic ones. Adding α7(1-208)-specific antibodies to the LPS challenge prevented elevation of both the anti-inflammatory and pro-apoptotic miRNAs while supporting the resistance of brain mitochondria to Ca(2+) and maintaining α7 nAChR/AChE decreases. In U373 cells, α7-specific antibodies and LPS both stimulated interleukin-6 production through the p38/Src-dependent pathway. Our findings demonstrate that acute LPS-induced inflammation induces the cholinergic anti-inflammatory pathway in the brain, that α7 nAChR down-regulation limits this pathway, and that α7-specific antibodies aggravate neuroinflammation by inducing the pro-inflammatory interleukin-6 and dampening anti-inflammatory miRNAs; however, these antibodies may protect brain mitochondria and decrease the levels of pro-apoptotic miRNAs, preventing LPS-induced neurodegeneration.
Collapse
Affiliation(s)
- Olena Lykhmus
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Nibha Mishra
- The Edmond and Lily Safra Center of Brain Science and The Alexander Silberman Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Lyudmyla Koval
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Olena Kalashnyk
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Galyna Gergalova
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Kateryna Uspenska
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Serghiy Komisarenko
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| | - Hermona Soreq
- The Edmond and Lily Safra Center of Brain Science and The Alexander Silberman Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Maryna Skok
- Laboratory of Cell Receptors Immunology, O. V. Palladin Institute of BiochemistryKyiv, Ukraine
| |
Collapse
|
204
|
The Transient Receptor Potential Melastatin 2 (TRPM2) Channel Contributes to β-Amyloid Oligomer-Related Neurotoxicity and Memory Impairment. J Neurosci 2016; 35:15157-69. [PMID: 26558786 DOI: 10.1523/jneurosci.4081-14.2015] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED In Alzheimer's disease, accumulation of soluble oligomers of β-amyloid peptide is known to be highly toxic, causing disturbances in synaptic activity and neuronal death. Multiple studies relate these effects to increased oxidative stress and aberrant activity of calcium-permeable cation channels leading to calcium imbalance. The transient receptor potential melastatin 2 (TRPM2) channel, a Ca(2+)-permeable nonselective cation channel activated by oxidative stress, has been implicated in neurodegenerative diseases, and more recently in amyloid-induced toxicity. Here we show that the function of TRPM2 is augmented by treatment of cultured neurons with β-amyloid oligomers. Aged APP/PS1 Alzheimer's mouse model showed increased levels of endoplasmic reticulum stress markers, protein disulfide isomerase and phosphorylated eukaryotic initiation factor 2α, as well as decreased levels of the presynaptic marker synaptophysin. Elimination of TRPM2 in APP/PS1 mice corrected these abnormal responses without affecting plaque burden. These effects of TRPM2 seem to be selective for β-amyloid toxicity, as ER stress responses to thapsigargin or tunicamycin in TRPM2(-/-) neurons was identical to that of wild-type neurons. Moreover, reduced microglial activation was observed in TRPM2(-/-)/APP/PS1 hippocampus compared with APP/PS1 mice. In addition, age-dependent spatial memory deficits in APP/PS1 mice were reversed in TRPM2(-/-)/APP/PS1 mice. These results reveal the importance of TRPM2 for β-amyloid neuronal toxicity, suggesting that TRPM2 activity could be potentially targeted to improve outcomes in Alzheimer's disease. SIGNIFICANCE STATEMENT Transient receptor potential melastatin 2 (TRPM2) is an oxidative stress sensing calcium-permeable channel that is thought to contribute to calcium dysregulation associated with neurodegenerative diseases, including Alzheimer's disease. Here we show that oligomeric β-amyloid, the toxic peptide in Alzheimer's disease, facilitates TRPM2 channel activation. In mice designed to model Alzheimer's disease, genetic elimination of TRPM2 normalized deficits in synaptic markers in aged mice. Moreover, the absence of TRPM2 improved age-dependent spatial memory deficits observed in Alzheimer's mice. Our results reveal the importance of TRPM2 for neuronal toxicity and memory impairments in an Alzheimer's mouse model and suggest that TRPM2 could be targeted for the development of therapeutic agents effective in the treatment of dementia.
Collapse
|
205
|
Takeda K, Yamaguchi Y, Hino M, Kato F. Potentiation of Acetylcholine-Mediated Facilitation of Inhibitory Synaptic Transmission by an Azaindolizione Derivative, ZSET1446 (ST101), in the Rat Hippocampus. J Pharmacol Exp Ther 2016; 356:445-55. [PMID: 26578264 DOI: 10.1124/jpet.115.229021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/16/2015] [Indexed: 03/08/2025] Open
Abstract
The integrity of the hippocampal network depends on the coordination of excitatory and inhibitory signaling, which are under dynamic control by various regulatory influences such as the cholinergic systems. ZSET1446 (ST101; spiro[imidazo[1,2-a]pyridine-3,2-indan]-2(3H)-one) is a newly synthesized azaindolizinone derivative that significantly improves learning deficits in various types of Alzheimer disease (AD) models in rats. We examined the effect of ZSET1446 on the nicotinic acetylcholine (ACh) receptor (nAChR)-mediated regulation of synaptic transmission in hippocampal slices of rats. ZSET1446 significantly potentiated the facilitatory effect of nicotine and ACh on the frequency of spontaneous postsynaptic currents (sPSCs) recorded in CA1 pyramidal neurons with a maximum effect at 100 pM (tested range, 10 pM-1000 pM). The basal sPSC frequency without ACh was not affected. Such potentiation by ZSET1446 was observed in both the pharmacologic isolations of inhibitory and excitatory sPSCs and markedly reduced by blockade of either α7 or α4β2 nAChRs. ZSET1446 did not affect ACh-activated inward currents or depolarization of interneurons in the stratum radiatum and the lacunosum moleculare. These results indicate that ZSET1446 potentiates the nicotine-mediated enhancement of synaptic transmission in the hippocampal neurons without affecting nAChRs themselves, providing a novel possible mechanism of procognitive action that might improve learning deficits in clinical therapy.
Collapse
Affiliation(s)
- Kentaro Takeda
- Central Research Laboratory, Zenyaku Kogyo Co., Ltd. (K.T., Y.Y., M.H.,) and Department of Neuroscience, Jikei University School of Medicine, Tokyo Japan (F.K.)
| | - Yoshimasa Yamaguchi
- Central Research Laboratory, Zenyaku Kogyo Co., Ltd. (K.T., Y.Y., M.H.,) and Department of Neuroscience, Jikei University School of Medicine, Tokyo Japan (F.K.)
| | - Masataka Hino
- Central Research Laboratory, Zenyaku Kogyo Co., Ltd. (K.T., Y.Y., M.H.,) and Department of Neuroscience, Jikei University School of Medicine, Tokyo Japan (F.K.)
| | - Fusao Kato
- Central Research Laboratory, Zenyaku Kogyo Co., Ltd. (K.T., Y.Y., M.H.,) and Department of Neuroscience, Jikei University School of Medicine, Tokyo Japan (F.K.)
| |
Collapse
|
206
|
Glimepiride protects neurons against amyloid-β-induced synapse damage. Neuropharmacology 2016; 101:225-36. [DOI: 10.1016/j.neuropharm.2015.09.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/04/2015] [Accepted: 09/28/2015] [Indexed: 12/30/2022]
|
207
|
Han SH, Park JC, Mook-Jung I. Amyloid β-interacting partners in Alzheimer's disease: From accomplices to possible therapeutic targets. Prog Neurobiol 2016; 137:17-38. [DOI: 10.1016/j.pneurobio.2015.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022]
|
208
|
Perforin Promotes Amyloid Beta Internalisation in Neurons. Mol Neurobiol 2016; 54:874-887. [DOI: 10.1007/s12035-016-9685-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022]
|
209
|
Ouach A, Pin F, Bertrand E, Vercouillie J, Gulhan Z, Mothes C, Deloye JB, Guilloteau D, Suzenet F, Chalon S, Routier S. Design of α7 nicotinic acetylcholine receptor ligands using the (het)Aryl-1,2,3-triazole core: Synthesis, in vitro evaluation and SAR studies. Eur J Med Chem 2016; 107:153-64. [DOI: 10.1016/j.ejmech.2015.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 10/22/2022]
|
210
|
Cao S, Yu L, Mao J, Wang Q, Ruan J. Uncovering the Molecular Mechanism of Actions between Pharmaceuticals and Proteins on the AD Network. PLoS One 2015; 10:e0144387. [PMID: 26650760 PMCID: PMC4674063 DOI: 10.1371/journal.pone.0144387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/17/2015] [Indexed: 11/18/2022] Open
Abstract
This study begins with constructing the mini metabolic networks (MMNs) of beta amyloid (Aβ) and acetylcholine (ACh) which stimulate the Alzheimer's Disease (AD). Then we generate the AD network by incorporating MMNs of Aβ and ACh, and other MMNs of stimuli of AD. The panel of proteins contains 49 enzymes/receptors on the AD network which have the 3D-structure in PDB. The panel of drugs is formed by 5 AD drugs and 5 AD nutraceutical drugs, and 20 non-AD drugs. All of these complexes formed by these 30 drugs and 49 proteins are transformed into dyadic arrays. Utilizing the prior knowledge learned from the drug panel, we propose a statistical classification (dry-lab). According to the wet-lab for the complex of amiloride and insulin degrading enzyme, and the complex of amiloride and neutral endopeptidase, we are confident that this dry-lab is reliable. As the consequences of the dry-lab, we discover many interesting implications. Especially, we show that possible causes of Tacrine, donepezil, galantamine and huperzine A cannot improve the level of ACh which is against to their original design purpose but they still prevent AD to be worse as Aβ deposition appeared. On the other hand, we recommend Miglitol and Atenolol as the safe and potent drugs to improve the level of ACh before Aβ deposition appearing. Moreover, some nutrients such as NADH and Vitamin E should be controlled because they may harm health if being used in wrong way and wrong time. Anyway, the insights shown in this study are valuable to be developed further.
Collapse
Affiliation(s)
- Shujuan Cao
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
| | - Liang Yu
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
| | - Jingyuan Mao
- Internal Medicine for the Heart, Tianjin University of Traditional Chinese Medicine, Tianjin, PRC
| | - Quan Wang
- National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Science, Beijing, PRC
- High Throughput Molecular Drug Discovery Center, Tianjin International Joint Academy of Biomedicine, TEDA, Tianjin, PRC
| | - Jishou Ruan
- College of Mathematical Sciences and LPMC, Nankai University, Tianjin, PRC
- State Key Laboratory for Medical Chemical and Biology, Nankai University, Tianjin, PRC
- * E-mail:
| |
Collapse
|
211
|
Puzzo D, Gulisano W, Arancio O, Palmeri A. The keystone of Alzheimer pathogenesis might be sought in Aβ physiology. Neuroscience 2015; 307:26-36. [PMID: 26314631 PMCID: PMC4591241 DOI: 10.1016/j.neuroscience.2015.08.039] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/15/2015] [Accepted: 08/18/2015] [Indexed: 01/17/2023]
Abstract
For several years Amyloid-beta peptide (Aβ) has been considered the main pathogenetic factor of Alzheimer's disease (AD). According to the so called Amyloid Cascade Hypothesis the increase of Aβ triggers a series of events leading to synaptic dysfunction and memory loss as well as to the structural brain damage in the later stage of the disease. However, several evidences suggest that this hypothesis is not sufficient to explain AD pathogenesis, especially considering that most of the clinical trials aimed to decrease Aβ levels have been unsuccessful. Moreover, Aβ is physiologically produced in the healthy brain during neuronal activity and it is needed for synaptic plasticity and memory. Here we propose a model interpreting AD pathogenesis as an alteration of the negative feedback loop between Aβ and its physiological receptors, focusing on alpha7 nicotinic acetylcholine receptors (α7-nAchRs). According to this vision, when Aβ cannot exert its physiological function a negative feedback mechanism would induce a compensatory increase of its production leading to an abnormal accumulation that reduces α7-nAchR function, leading to synaptic dysfunction and memory loss. In this perspective, the indiscriminate Aβ removal might worsen neuronal homeostasis, causing a further impoverishment of learning and memory. Even if further studies are needed to better understand and validate these mechanisms, we believe that to deepen the role of Aβ in physiological conditions might represent the keystone to elucidate important aspects of AD pathogenesis.
Collapse
Affiliation(s)
- D Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy.
| | - W Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| | - O Arancio
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, 630 West 168th Street, Columbia University, New York, NY 10032, USA
| | - A Palmeri
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, Viale A. Doria 6 (ed. 2), University of Catania, Catania 95125, Italy
| |
Collapse
|
212
|
BAG2 expression dictates a functional intracellular switch between the p38-dependent effects of nicotine on tau phosphorylation levels via the α7 nicotinic receptor. Exp Neurol 2015; 275 Pt 1:69-77. [PMID: 26496817 DOI: 10.1016/j.expneurol.2015.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/25/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
The histopathological hallmarks present in Alzheimer's disease (AD) brain are plaques of Aβ peptide, neurofibrillary tangles of hyperphosphorylated tau protein, and a reduction in nicotinic acetylcholine receptor (nAChR) levels. The role of nAChRs in AD is particularly controversial. Tau protein function is regulated by phosphorylation, and its hyperphosphorylated forms are significantly more abundant in AD brain. Little is known about the relationship between nAChR and phospho-tau degradation machinery. Activation of nAChRs has been reported to increase and decrease tau phosphorylation levels, and the mechanisms responsible for this discrepancy are not presently understood. The co-chaperone BAG2 is capable of regulating phospho-tau levels via protein degradation. In SH-SY5Y cell line and rat primary hippocampal cell culture low endogenous BAG2 levels constitute an intracellular environment conducive to nicotine-induced accumulation of phosphorylated tau protein. Further, nicotine treatment inhibited endogenous expression of BAG2, resulting in increased levels of phosphorylated tau indistinguishable from those induced by BAG2 knockdown. Conversely, overexpression of BAG2 is conducive to a nicotine-induced reduction in cellular levels of phosphorylated tau protein. In both cases the effect of nicotine was p38MAPK-dependent, while the α7 antagonist MLA was synthetic to nicotine treatment, either increasing levels of phospho-Tau in the absence of BAG2, or further decreasing the levels of phospho-Tau in the presence of BAG2. Taken together, these findings reconcile the apparently contradictory effects of nicotine on tau phosphorylation by suggesting a role for BAG2 as an important regulator of p38-dependent tau kinase activity and phospho-tau degradation in response to nicotinic receptor stimulation. Thus, we report that BAG2 expression dictates a functional intracellular switch between the p38-dependent functions of nicotine on tau phosphorylation levels via the α7 nicotinic receptor.
Collapse
|
213
|
John D, Berg DK. Long-lasting changes in neural networks to compensate for altered nicotinic input. Biochem Pharmacol 2015; 97:418-424. [PMID: 26206188 PMCID: PMC4600434 DOI: 10.1016/j.bcp.2015.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 07/07/2015] [Indexed: 11/21/2022]
Abstract
The nervous system must balance excitatory and inhibitory input to constrain network activity levels within a proper dynamic range. This is a demanding requirement during development, when networks form and throughout adulthood as networks respond to constantly changing environments. Defects in the ability to sustain a proper balance of excitatory and inhibitory activity are characteristic of numerous neurological disorders such as schizophrenia, Alzheimer's disease, and autism. A variety of homeostatic mechanisms appear to be critical for balancing excitatory and inhibitory activity in a network. These are operative at the level of individual neurons, regulating their excitability by adjusting the numbers and types of ion channels, and at the level of synaptic connections, determining the relative numbers of excitatory versus inhibitory connections a neuron receives. Nicotinic cholinergic signaling is well positioned to contribute at both levels because it appears early in development, extends across much of the nervous system, and modulates transmission at many kinds of synapses. Further, it is known to influence the ratio of excitatory-to-inhibitory synapses formed on neurons during development. GABAergic inhibitory neurons are likely to be key for maintaining network homeostasis (limiting excitatory output), and nicotinic signaling is known to prominently regulate the activity of several GABAergic neuronal subtypes. But how nicotinic signaling achieves this and how networks may compensate for the loss of such input are important questions remaining unanswered. These issues are reviewed.
Collapse
Affiliation(s)
- Danielle John
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357, United States; Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92093-0357, United States
| | - Darwin K Berg
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093-0357, United States; Kavli Institute for Brain and Mind, University of California, San Diego, La Jolla, CA 92093-0357, United States.
| |
Collapse
|
214
|
A novel nicotinic mechanism underlies β-amyloid-induced neurotoxicity. Neuropharmacology 2015; 97:457-63. [DOI: 10.1016/j.neuropharm.2015.04.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 04/09/2015] [Accepted: 04/26/2015] [Indexed: 10/23/2022]
|
215
|
Bertrand D, Lee CHL, Flood D, Marger F, Donnelly-Roberts D. Therapeutic Potential of α7 Nicotinic Acetylcholine Receptors. Pharmacol Rev 2015; 67:1025-73. [PMID: 26419447 DOI: 10.1124/pr.113.008581] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Progress in the fields of neuroscience and molecular biology has identified the forebrain cholinergic system as being important in many higher order brain functions. Further analysis of the genes encoding the nicotinic acetylcholine receptors (nAChRs) has highlighted, in particular, the role of α7 nAChRs in these higher order brain functions as evidenced by their peculiar physiologic and pharmacological properties. As this receptor has gained the attention of scientists from academia and industry, our knowledge of its roles in various brain and bodily functions has increased immensely. We have also seen the development of small molecules that have further refined our understanding of the roles of α7 nAChRs, and these molecules have begun to be tested in clinical trials for several indications. Although a large body of data has confirmed a role of α7 nAChRs in cognition, the translation of small molecules affecting α7 nAChRs into therapeutics has to date only progressed to the stage of testing in clinical trials. Notably, however, most recent human genetic and biochemical studies are further underscoring the crucial role of α7 nAChRs and associated genes in multiple organ systems and disease states. The aim of this review is to discuss our current knowledge of α7 nAChRs and their relevance as a target in specific functional systems and disease states.
Collapse
Affiliation(s)
- Daniel Bertrand
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Chih-Hung L Lee
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Dorothy Flood
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Fabrice Marger
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Diana Donnelly-Roberts
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| |
Collapse
|
216
|
The α7 nicotinic acetylcholine receptor: A mediator of pathogenesis and therapeutic target in autism spectrum disorders and Down syndrome. Biochem Pharmacol 2015; 97:363-377. [DOI: 10.1016/j.bcp.2015.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/04/2015] [Indexed: 01/06/2023]
|
217
|
Duan JJ, Lozada AF, Gou CY, Xu J, Chen Y, Berg DK. Nicotine recruits glutamate receptors to postsynaptic sites. Mol Cell Neurosci 2015; 68:340-9. [PMID: 26365992 DOI: 10.1016/j.mcn.2015.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/04/2015] [Accepted: 09/07/2015] [Indexed: 01/20/2023] Open
Abstract
Cholinergic neurons project throughout the nervous system and activate nicotinic receptors to modulate synaptic function in ways that shape higher order brain function. The acute effects of nicotinic signaling on long-term synaptic plasticity have been well-characterized. Less well understood is how chronic exposure to low levels of nicotine, such as those encountered by habitual smokers, can alter neural connections to promote addiction and other lasting behavioral effects. We show here that chronic exposure of hippocampal neurons in culture to low levels of nicotine recruits AMPA and NMDA receptors to the cell surface and sequesters them at postsynaptic sites. The receptors include GluA2-containing AMPA receptors, which are responsible for most of the excitatory postsynaptic current mediated by AMPA receptors on the neurons, and include NMDA receptors containing GluN1 and GluN2B subunits. Moreover, we find that the nicotine treatment also increases expression of the presynaptic component synapsin 1 and arranges it in puncta juxtaposed to the additional AMPA and NMDA receptor puncta, suggestive of increases in synaptic contacts. Consistent with increased synaptic input, we find that the nicotine treatment leads to an increase in the excitatory postsynaptic currents mediated by AMPA and NMDA receptors. Further, the increases skew the ratio of excitatory-to-inhibitory input that the cell receives, and this holds both for pyramidal neurons and inhibitory neurons in the hippocampal CA1 region. The GluN2B-containing NMDA receptor redistribution at synapses is associated with a significant increase in GluN2B phosphorylation at Tyr1472, a site known to prevent GluN2B endocytosis. These results suggest that chronic exposure to low levels of nicotine not only alters functional connections but also is likely to change excitability levels across networks. Further, it may increase the propensity for synaptic plasticity, given the increase in synaptic NMDA receptors.
Collapse
Affiliation(s)
- Jing-Jing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Neurobiology Section, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0357, United States
| | - Adrian F Lozada
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0357, United States
| | - Chen-Yu Gou
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Xu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuan Chen
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Darwin K Berg
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0357, United States.
| |
Collapse
|
218
|
Ullah G, Demuro A, Parker I, Pearson JE. Analyzing and Modeling the Kinetics of Amyloid Beta Pores Associated with Alzheimer's Disease Pathology. PLoS One 2015; 10:e0137357. [PMID: 26348728 PMCID: PMC4562663 DOI: 10.1371/journal.pone.0137357] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/14/2015] [Indexed: 11/24/2022] Open
Abstract
Amyloid beta (Aβ) oligomers associated with Alzheimer’s disease (AD) form Ca2+-permeable plasma membrane pores, leading to a disruption of the otherwise well-controlled intracellular calcium (Ca2+) homeostasis. The resultant up-regulation of intracellular Ca2+ concentration has detrimental implications for memory formation and cell survival. The gating kinetics and Ca2+ permeability of Aβ pores are not well understood. We have used computational modeling in conjunction with the ability of optical patch-clamping for massively parallel imaging of Ca2+ flux through thousands of pores in the cell membrane of Xenopus oocytes to elucidate the kinetic properties of Aβ pores. The fluorescence time-series data from individual pores were idealized and used to develop data-driven Markov chain models for the kinetics of the Aβ pore at different stages of its evolution. Our study provides the first demonstration of developing Markov chain models for ion channel gating that are driven by optical-patch clamp data with the advantage of experiments being performed under close to physiological conditions. Towards the end, we demonstrate the up-regulation of gating of various Ca2+ release channels due to Aβ pores and show that the extent and spatial range of such up-regulation increases as Aβ pores with low open probability and Ca2+ permeability transition into those with high open probability and Ca2+ permeability.
Collapse
Affiliation(s)
- Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, United States of America
- * E-mail:
| | - Angelo Demuro
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, United States of America
| | - Ian Parker
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, United States of America
| | - John E. Pearson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, United States of America
| |
Collapse
|
219
|
Sinkus ML, Graw S, Freedman R, Ross RG, Lester HA, Leonard S. The human CHRNA7 and CHRFAM7A genes: A review of the genetics, regulation, and function. Neuropharmacology 2015; 96:274-88. [PMID: 25701707 PMCID: PMC4486515 DOI: 10.1016/j.neuropharm.2015.02.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 01/16/2023]
Abstract
The human α7 neuronal nicotinic acetylcholine receptor gene (CHRNA7) is ubiquitously expressed in both the central nervous system and in the periphery. CHRNA7 is genetically linked to multiple disorders with cognitive deficits, including schizophrenia, bipolar disorder, ADHD, epilepsy, Alzheimer's disease, and Rett syndrome. The regulation of CHRNA7 is complex; more than a dozen mechanisms are known, one of which is a partial duplication of the parent gene. Exons 5-10 of CHRNA7 on chromosome 15 were duplicated and inserted 1.6 Mb upstream of CHRNA7, interrupting an earlier partial duplication of two other genes. The chimeric CHRFAM7A gene product, dupα7, assembles with α7 subunits, resulting in a dominant negative regulation of function. The duplication is human specific, occurring neither in primates nor in rodents. The duplicated α7 sequence in exons 5-10 of CHRFAM7A is almost identical to CHRNA7, and thus is not completely queried in high throughput genetic studies (GWAS). Further, pre-clinical animal models of the α7nAChR utilized in drug development research do not have CHRFAM7A (dupα7) and cannot fully model human drug responses. The wide expression of CHRNA7, its multiple functions and modes of regulation present challenges for study of this gene in disease. This article is part of the Special Issue entitled 'The Nicotinic Acetylcholine Receptor: From Molecular Biology to Cognition'.
Collapse
Affiliation(s)
- Melissa L Sinkus
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Sharon Graw
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA; Veterans Affairs Medical Research Center, Denver, CO 80262, USA.
| | - Randal G Ross
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Henry A Lester
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Sherry Leonard
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA; Veterans Affairs Medical Research Center, Denver, CO 80262, USA.
| |
Collapse
|
220
|
Abstract
Neurodegeneration correlates with Alzheimer's disease (AD) symptoms, but the molecular identities of pathogenic amyloid β-protein (Aβ) oligomers and their targets, leading to neurodegeneration, remain unclear. Amylospheroids (ASPD) are AD patient-derived 10- to 15-nm spherical Aβ oligomers that cause selective degeneration of mature neurons. Here, we show that the ASPD target is neuron-specific Na(+)/K(+)-ATPase α3 subunit (NAKα3). ASPD-binding to NAKα3 impaired NAKα3-specific activity, activated N-type voltage-gated calcium channels, and caused mitochondrial calcium dyshomeostasis, tau abnormalities, and neurodegeneration. NMR and molecular modeling studies suggested that spherical ASPD contain N-terminal-Aβ-derived "thorns" responsible for target binding, which are distinct from low molecular-weight oligomers and dodecamers. The fourth extracellular loop (Ex4) region of NAKα3 encompassing Asn(879) and Trp(880) is essential for ASPD-NAKα3 interaction, because tetrapeptides mimicking this Ex4 region bound to the ASPD surface and blocked ASPD neurotoxicity. Our findings open up new possibilities for knowledge-based design of peptidomimetics that inhibit neurodegeneration in AD by blocking aberrant ASPD-NAKα3 interaction.
Collapse
|
221
|
Abstract
Alzheimer's disease (AD) is a neurodegenerative illness associated with dementia and is most prevalent among the elderly population. Current medications can only treat symptoms. Alkaloids are structurally diverse and have been an important source of therapeutics for various brain disorders. Two US Food and Drug Administration (FDA)-approved acetylcholinesterase inhibitors for AD, galantamine and rivastigmine, are in fact alkaloids. In addition, clinical trials of four other extensively studied alkaloids-huperzine A, caffeine, nicotine, and indomethacin-have been conducted but do not convincingly demonstrate their clinical efficacy for AD. Interestingly, rhynchophylline, a known neuroprotective alkaloid, was recently discovered by in silico screening as an inhibitor of EphA4, a novel target for AD. Here, we review the pathophysiological mechanisms underlying AD, current treatment strategies, and therapeutic potential of several selected plant alkaloids in AD, highlighting their various drug targets and the key supportive preclinical and clinical studies. Future research should include more rigorous clinical studies of the most promising alkaloids, the further development of recently discovered candidate alkaloids, and the continual search for new alkaloids for relevant drug targets. It remains promising that an alkaloid drug candidate could significantly affect the progression of AD in addition to providing symptomatic relief.
Collapse
Affiliation(s)
- Yu Pong Ng
- Division of Life Science, Molecular Neuroscience Center, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Terry Cho Tsun Or
- Division of Life Science, Molecular Neuroscience Center, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, Molecular Neuroscience Center, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
222
|
Koukouli F, Maskos U. The multiple roles of the α7 nicotinic acetylcholine receptor in modulating glutamatergic systems in the normal and diseased nervous system. Biochem Pharmacol 2015. [PMID: 26206184 DOI: 10.1016/j.bcp.2015.07.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neuronal nicotinic acetylcholine receptors (nAChRs) play an important role in a variety of modulatory and regulatory processes including neurotransmitter release and synaptic transmission in various brain regions of the central nervous system (CNS). Glutamate is the principal excitatory neurotransmitter in the brain and the glutamatergic system participates in the pathophysiology of several neuropsychiatric disorders. Underpinning the importance of nAChRs, many studies demonstrated that nAChRs containing the α7 subunit facilitate glutamate release. Here, we review the currently available body of experimental evidence pertaining to α7 subunit containing nAChRs in their contribution to the modulation of glutamatergic neurotransmission, and we highlight the role of α7 in synaptic plasticity, the morphological and functional maturation of the glutamatergic system and therefore its important contribution in the modulation of neural circuits of the CNS.
Collapse
Affiliation(s)
- Fani Koukouli
- Institut Pasteur, Neurobiologie intégrative des systèmes cholinergiques, CNRS UMR 3571, Paris, France.
| | - Uwe Maskos
- Institut Pasteur, Neurobiologie intégrative des systèmes cholinergiques, CNRS UMR 3571, Paris, France.
| |
Collapse
|
223
|
Sadigh-Eteghad S, Talebi M, Mahmoudi J, Babri S, Shanehbandi D. Selective activation of α7 nicotinic acetylcholine receptor by PHA-543613 improves Aβ25–35-mediated cognitive deficits in mice. Neuroscience 2015; 298:81-93. [DOI: 10.1016/j.neuroscience.2015.04.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 12/22/2022]
|
224
|
Zhao LN, Zheng J, Chew LY, Mu Y. An Investigation on the Fundamental Interaction between Abeta Peptides and the AT-Rich DNA. J Phys Chem B 2015; 119:8247-59. [PMID: 26086541 DOI: 10.1021/acs.jpcb.5b00957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
DNA damage is ubiquitous in all mammalian cells with the occurrence of more than 60,000 times per day per cell. In particular, DNA damage in neurons is found to accumulate with age and has been suggested to interfere with the synthesis of functional proteins. Moreover, recent studies have found through transgenic mice that human amyloid precursor protein causes an increase in DNA double-strand breaks (DSBs) with the effect of a prolongation in DNA repair. It is surmised that amyloid β (Aβ) exacerbates the DNA DSBs in neurons, possibly engendering neuronal dysfunction as a result. However, a good understanding on the holistic interaction mechanisms and the manner in which Aβ intertwines with DNA damage is still in its infancy. In our study, we found that DNA with an AT-rich sequence has a very low binding affinity toward Aβ by means of molecular dynamics simulation. While we have pursued a particular sequence of DNA in this study, other DNA sequences are expected to affect the interaction and binding affinity between DNA and Aβ, and will be pursued in our further research. Nonetheless, we have uncovered favorable interaction between the positively charged side chain of Aβ and the two ends of DNA. The latest experiment reveals that many of the double-stranded breaks in neurons can be fixed via DNA repair mechanisms but not in the case that Aβs are present. It is found that the increased numbers of DSBs prevail in active neurons. Here, on the basis of the favorable interaction between Aβ and the two ends of DNA, we propose the possibility that Aβ prevents DNA repair via binding directly to the break ends of the DNA, which further exacerbates DNA damage. Moreover, we have found that the base pair oxygen of the DNA has a greater preference to form hydrogen bonds than the backbone oxygen with Aβ at the two ends. Thus, we postulate that Aβ could serve to prevent the repair of AT-rich DNA, and it is unlikely to cause its breakage or affect its binding toward histone. Another important observation from our study is that AT-rich DNA has very little or no influence on Aβ oligomerization. Finally, even though we do not observe any dramatic DNA conformational change in the presence of Aβ, we do observe an increase in diversity of the DNA structural parameters such as groove width, local base step, and torsional angles in lieu of Aβ interactions.
Collapse
Affiliation(s)
- Li Na Zhao
- †School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore.,‡School of Computer Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore.,§Bioinformatics Institute, 30 Biopolis Street, Singapore 138671
| | - Jie Zheng
- ‡School of Computer Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore.,∥Genome Institute of Singapore, A* STAR, 60 Biopolis Street, Singapore 138672
| | - Lock Yue Chew
- †School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore.,⊥Complexity Institute, Nanyang Technological University, 18 Nanyang Drive, Singapore
| | - Yuguang Mu
- #School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| |
Collapse
|
225
|
Wilcox KC, Marunde MR, Das A, Velasco PT, Kuhns BD, Marty MT, Jiang H, Luan CH, Sligar SG, Klein WL. Nanoscale Synaptic Membrane Mimetic Allows Unbiased High Throughput Screen That Targets Binding Sites for Alzheimer's-Associated Aβ Oligomers. PLoS One 2015; 10:e0125263. [PMID: 25928376 PMCID: PMC4415972 DOI: 10.1371/journal.pone.0125263] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 03/23/2015] [Indexed: 01/05/2023] Open
Abstract
Despite their value as sources of therapeutic drug targets, membrane proteomes are largely inaccessible to high-throughput screening (HTS) tools designed for soluble proteins. An important example comprises the membrane proteins that bind amyloid β oligomers (AβOs). AβOs are neurotoxic ligands thought to instigate the synapse damage that leads to Alzheimer's dementia. At present, the identities of initial AβO binding sites are highly uncertain, largely because of extensive protein-protein interactions that occur following attachment of AβOs to surface membranes. Here, we show that AβO binding sites can be obtained in a state suitable for unbiased HTS by encapsulating the solubilized synaptic membrane proteome into nanoscale lipid bilayers (Nanodiscs). This method gives a soluble membrane protein library (SMPL)--a collection of individualized synaptic proteins in a soluble state. Proteins within SMPL Nanodiscs showed enzymatic and ligand binding activity consistent with conformational integrity. AβOs were found to bind SMPL Nanodiscs with high affinity and specificity, with binding dependent on intact synaptic membrane proteins, and selective for the higher molecular weight oligomers known to accumulate at synapses. Combining SMPL Nanodiscs with a mix-incubate-read chemiluminescence assay provided a solution-based HTS platform to discover antagonists of AβO binding. Screening a library of 2700 drug-like compounds and natural products yielded one compound that potently reduced AβO binding to SMPL Nanodiscs, synaptosomes, and synapses in nerve cell cultures. Although not a therapeutic candidate, this small molecule inhibitor of synaptic AβO binding will provide a useful experimental antagonist for future mechanistic studies of AβOs in Alzheimer's model systems. Overall, results provide proof of concept for using SMPLs in high throughput screening for AβO binding antagonists, and illustrate in general how a SMPL Nanodisc system can facilitate drug discovery for membrane protein targets.
Collapse
Affiliation(s)
- Kyle C. Wilcox
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| | - Matthew R. Marunde
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| | - Aditi Das
- Department of Comparative Biosciences, University of Illinois—Urbana Champaign, Urbana, IL, United States of America
- Department of Biochemistry, University of Illinois—Urbana Champaign, Urbana, IL, United States of America
| | - Pauline T. Velasco
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| | - Benjamin D. Kuhns
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| | - Michael T. Marty
- Department of Chemistry, University of Illinois—Urbana Champaign, Urbana, IL, United States of America
| | - Haoming Jiang
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| | - Chi-Hao Luan
- High Throughput Analysis Laboratory and Department of Molecular Biosciences, Northwestern University, Evanston, IL, United States of America
| | - Stephen G. Sligar
- Department of Biochemistry, University of Illinois—Urbana Champaign, Urbana, IL, United States of America
- Department of Chemistry, University of Illinois—Urbana Champaign, Urbana, IL, United States of America
| | - William L. Klein
- Department of Neurobiology, Northwestern University, Evanston, IL, United States of America
| |
Collapse
|
226
|
Hao Y, Tang J, Wang K. Development of Automated Patch Clamp Assay for Evaluation of α7 Nicotinic Acetylcholine Receptor Agonists in Automated QPatch-16. Assay Drug Dev Technol 2015; 13:174-84. [PMID: 25880723 DOI: 10.1089/adt.2014.622] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The α7 nicotinic acetylcholine receptor (α7 nAChR) is an important and challenging target for drug discovery in the area of neuropsychiatric disorders. The current screening for chemicals targeting α7 nAChRs is primarily achieved by the use of low-throughput assay two-electrode voltage clamp (TEVC) in nonmammalian Xenopus oocytes. Automated patch clamp system has emerged as an attractive approach compared to conventional electrophysiology. To develop a mammalian cell-based functional assay in an automated electrophysiology system, we in this study generated a stable expression of α7 nAChRs in GH3 cells that originated from a rat pituitary tumor cell line and utilized automated QPatch-16 to test a set of tool compounds and chemicals identified as α7 agonists by TEVC. For the improvement of evaluating weak or partial α7 nAChRs agonists, we achieved enhancement of the signal-to-noise ratio by the addition of a positive allosteric modulator PNU-120596, which only activates α7 current in the presence of agonist. This improved assay was further validated by using known α7 partial agonists, such as RG3487, EVP-6124, and A-P90. Using this validated assay, we were able to identify a novel agonist 140507C that partially activates α7 nAChRs. Taken together, our results validate the use of QPatch-16 for evaluation α7 partial agonists, demonstrating its utility as an effective tool for α7 ion channel drug discovery.
Collapse
Affiliation(s)
- Yuchen Hao
- 1 Department of Molecular and Cellular Pharmacology, State Key Laboratory of Natural and Biomimetic Drugs, Peking University School of Pharmaceutical Sciences , Beijing, China
| | | | | |
Collapse
|
227
|
Virtual screening studies of Chinese medicine Coptidis Rhizoma as alpha7 nicotinic acetylcholine receptor agonists for treatment of Alzheimer’s disease. J Mol Struct 2015. [DOI: 10.1016/j.molstruc.2015.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
228
|
Beinat C, Reekie T, Banister SD, O'Brien-Brown J, Xie T, Olson TT, Xiao Y, Harvey A, O'Connor S, Coles C, Grishin A, Kolesik P, Tsanaktsidis J, Kassiou M. Structure-activity relationship studies of SEN12333 analogues: determination of the optimal requirements for binding affinities at α7 nAChRs through incorporation of known structural motifs. Eur J Med Chem 2015; 95:277-301. [PMID: 25827398 DOI: 10.1016/j.ejmech.2015.03.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 03/11/2015] [Accepted: 03/12/2015] [Indexed: 12/27/2022]
Abstract
Alpha7 nicotinic acetylcholine receptors (nAChRs) have implications in the regulation of cognitive processes such as memory and attention and have been identified as a promising therapeutic target for the treatment of the cognitive deficits associated with schizophrenia and Alzheimer's disease (AD). Structure affinity relationship studies of the previously described α7 agonist SEN12333 (8), have resulted in the identification of compound 45, a potent and selective agonist of the α7 nAChR with enhanced affinity and improved physicochemical properties over the parent compound (SEN12333, 8).
Collapse
Affiliation(s)
- Corinne Beinat
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia; Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tristan Reekie
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia
| | - Samuel D Banister
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Teresa Xie
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20057, USA
| | - Thao T Olson
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20057, USA
| | - Yingxian Xiao
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20057, USA
| | | | | | | | | | | | - John Tsanaktsidis
- CSIRO Materials Science & Engineering, Ian Wark Laboratory, Bayview Avenue, Clayton, Victoria 3168, Australia
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia; Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
229
|
Ezeani M, Omabe M. A New Perspective of Lysosomal Cation Channel-Dependent Homeostasis in Alzheimer's Disease. Mol Neurobiol 2015; 53:1672-1678. [PMID: 25691454 DOI: 10.1007/s12035-015-9108-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 01/20/2015] [Indexed: 01/08/2023]
Abstract
Studies have reported typically biophysical lysosomal cation channels including TPCs. Their plausible biological roles are being elucidated by pharmacological, genetic and conventional patch clamp procedures. The best characterized so far among these channels is the ML1 isoform of TRP. The reported TRPs and TPCs are bypass for cation fluxes and are strategic for homeostasis of ionic milieu of the acidic organelles they confine to. Ca(2+) homeostasis and adequate acidic pHL are critically influential for the regulation of a plethora of biological functions these intracellular cation channels perform. In lysosomal ion channel biology, we review: ML1 and TPC2 in Ca(2+) signaling, ML1 and TPC2 in pH(L) regulation. Using Aβ42 and tau proteins found along clathrin endolysosomal internalization pathway (Fig. 3), we proffer a mechanism of abnormal pH(L) and ML1/TPC2-dependent cation homeostasis in AD.
Collapse
Affiliation(s)
- Martin Ezeani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Sir Charles Tupper Medical Building, 5850 College Street, Halifax, B3H 4R2, Nova Scotia, Canada.
| | - Maxwell Omabe
- Cancer Research Unit, Saskatchewan Cancer Agency, Department of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
230
|
Dineley KT, Pandya AA, Yakel JL. Nicotinic ACh receptors as therapeutic targets in CNS disorders. Trends Pharmacol Sci 2015; 36:96-108. [PMID: 25639674 PMCID: PMC4324614 DOI: 10.1016/j.tips.2014.12.002] [Citation(s) in RCA: 355] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 01/30/2023]
Abstract
The neurotransmitter acetylcholine (ACh) can regulate neuronal excitability by acting on the cys-loop cation-conducting ligand-gated nicotinic ACh receptor (nAChR) channels. These receptors are widely distributed throughout the central nervous system (CNS), being expressed on neurons and non-neuronal cells, where they participate in a variety of physiological responses such as anxiety, the central processing of pain, food intake, nicotine seeking behavior, and cognitive functions. In the mammalian brain, nine different subunits have been found thus far, which assemble into pentameric complexes with much subunit diversity; however, the α7 and α4β2 subtypes predominate in the CNS. Neuronal nAChR dysfunction is involved in the pathophysiology of many neurological disorders. Here we will briefly discuss the functional makeup and expression of the nAChRs in mammalian brain, and their role as targets in neurodegenerative diseases (in particular Alzheimer's disease, AD), neurodevelopmental disorders (in particular autism and schizophrenia), and neuropathic pain.
Collapse
Affiliation(s)
- Kelly T Dineley
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch at Galveston (UTMB), Galveston, TX, USA
| | - Anshul A Pandya
- Chukchi Campus, Department of Bioscience, College of Rural and Community Development, University of Alaska Fairbanks, P.O. Box 297, Kotzebue, AK 99752-0297, USA
| | - Jerrel L Yakel
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health (NIEHS/NIH), Department of Health and Human Services (DHHS), F2-08, P.O. Box 12233, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
231
|
Nisbet RM, Polanco JC, Ittner LM, Götz J. Tau aggregation and its interplay with amyloid-β. Acta Neuropathol 2015; 129:207-20. [PMID: 25492702 PMCID: PMC4305093 DOI: 10.1007/s00401-014-1371-2] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/11/2014] [Accepted: 11/26/2014] [Indexed: 01/09/2023]
Abstract
Neurofibrillary tangles and amyloid plaques constitute the hallmark brain lesions of Alzheimer's disease (AD) patients. Tangles are composed of fibrillar aggregates of the microtubule-associated protein tau, and plaques comprise fibrillar forms of a proteolytic cleavage product, amyloid-β (Aβ). Although plaques and tangles are the end-stage lesions in AD, small oligomers of Aβ and tau are now receiving increased attention as they are shown to correlate best with neurotoxicity. One key question of debate, however, is which of these pathologies appears first and hence is upstream in the pathocascade. Studies suggest that there is an intense crosstalk between the two molecules and, based on work in animal models, there is increasing evidence that Aβ, at least in part, exerts its toxicity via tau, with the Src kinase Fyn playing a crucial role in this process. In other experimental paradigms, Aβ and tau have been found to exert both separate and synergistic modes of toxicity. The challenge, however, is to integrate these different scenarios into a coherent picture. Furthermore, the ability of therapeutic interventions targeting just one of these molecules, to successfully neutralize the toxicity of the other, needs to be ascertained to improve current therapeutic strategies, such as immunotherapy, for the treatment of AD. Although this article is not intended to provide a comprehensive review of the currently pursued therapeutic strategies, we will discuss what has been achieved by immunotherapy and, in particular, how the inherent limitations of this approach can possibly be overcome by novel strategies that involve single-chain antibodies.
Collapse
Affiliation(s)
- Rebecca M. Nisbet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Juan-Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Lars M. Ittner
- Dementia Research Unit, Wallace Wurth Building, The University of New South Wales, Sydney, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
232
|
Regulation of presynaptic Ca2+, synaptic plasticity and contextual fear conditioning by a N-terminal β-amyloid fragment. J Neurosci 2015; 34:14210-8. [PMID: 25339735 DOI: 10.1523/jneurosci.0326-14.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Soluble β-amyloid has been shown to regulate presynaptic Ca(2+) and synaptic plasticity. In particular, picomolar β-amyloid was found to have an agonist-like action on presynaptic nicotinic receptors and to augment long-term potentiation (LTP) in a manner dependent upon nicotinic receptors. Here, we report that a functional N-terminal domain exists within β-amyloid for its agonist-like activity. This sequence corresponds to a N-terminal fragment generated by the combined action of α- and β-secretases, and resident carboxypeptidase. The N-terminal β-amyloid fragment is present in the brains and CSF of healthy adults as well as in Alzheimer's patients. Unlike full-length β-amyloid, the N-terminal β-amyloid fragment is monomeric and nontoxic. In Ca(2+) imaging studies using a model reconstituted rodent neuroblastoma cell line and isolated mouse nerve terminals, the N-terminal β-amyloid fragment proved to be highly potent and more effective than full-length β-amyloid in its agonist-like action on nicotinic receptors. In addition, the N-terminal β-amyloid fragment augmented theta burst-induced post-tetanic potentiation and LTP in mouse hippocampal slices. The N-terminal fragment also rescued LTP inhibited by elevated levels of full-length β-amyloid. Contextual fear conditioning was also strongly augmented following bilateral injection of N-terminal β-amyloid fragment into the dorsal hippocampi of intact mice. The fragment-induced augmentation of fear conditioning was attenuated by coadministration of nicotinic antagonist. The activity of the N-terminal β-amyloid fragment appears to reside largely in a sequence surrounding a putative metal binding site, YEVHHQ. These findings suggest that the N-terminal β-amyloid fragment may serve as a potent and effective endogenous neuromodulator.
Collapse
|
233
|
Feuerbach D, Pezous N, Weiss M, Shakeri-Nejad K, Lingenhoehl K, Hoyer D, Hurth K, Bilbe G, Pryce CR, McAllister K, Chaperon F, Kucher K, Johns D, Blaettler T, Lopez Lopez C. AQW051, a novel, potent and selective α7 nicotinic ACh receptor partial agonist: pharmacological characterization and phase I evaluation. Br J Pharmacol 2015; 172:1292-304. [PMID: 25363835 DOI: 10.1111/bph.13001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/14/2014] [Accepted: 10/27/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of the α7 nicotinic ACh receptor (nACh receptor) is considered an attractive target for the treatment of cognitive impairment associated with neurological disorders. Here we describe the novel α7-nACh receptor agonist AQW051 as a promising drug candidate for this indication. EXPERIMENTAL APPROACH AQW051 was functionally characterized in vitro and cognitive effects evaluated in rodent behavioural models. Pharmacokinetics and tolerability were evaluated in three phase I placebo-controlled studies in 180 healthy subjects. KEY RESULTS In vitro, AQW051 bound with high affinity to α7-nACh receptors and stimulated calcium influx in cells recombinantly expressing the human α7-nACh receptor. In vivo, AQW051 demonstrated good oral bioavailability and rapid penetration into the rodent brain. AQW051 administered over a broad dose range facilitated learning/memory performance in the object recognition and social recognition test in mice and the water maze model in aged rats. Clinically, AQW051 was well tolerated in healthy young and elderly subjects, with an adverse event (AE) profile comparable with placebo. No serious AEs were reported and all AEs were either mild or moderate in severity at single oral doses up to 200 mg and multiple daily doses up to 75 mg. Once-daily oral administration of AQW051 resulted in continuous exposure and a two- to threefold accumulation compared with steady state was achieved by 1 week. CONCLUSIONS AND IMPLICATIONS These data support further development of AQW051 as a cognitive-enhancing agent, as a therapeutic, for example, in Alzheimer's disease or schizophrenia.
Collapse
|
234
|
Kamynina AV, Ponomareva EV, Koroev DO, Volkova TD, Kolykhalov IV, Selezneva ND, Gavrilova SI, Vol'pina OM. [The reduced level of antibodies to acetylcholine receptor alpha 7 fragment in the blood serum of patients with Alzheimer's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2015; 115:128-132. [PMID: 26978506 DOI: 10.17116/jnevro2015115112128-132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Determination of antibodies to neuronal membrane proteins in the blood serum of patients is of interest for diagnosis and optimization of treatment of Alzheimer's disease (AD). Authors studied the level of antibodies to acetylcholine receptor alpha 7 protein fragment (AChR), prion protein (РrР) and glycation end-products (RAGE) as well as to intracellular proteins nucleophosmin (Nuc) and survivin (Sur) in the serum of AD patients. MATERIAL AND METHODS Serum samples of 26 patients with probable AD and 13 healthy people were studied. Exposed sections of each protein were used for the choice of peptides for antibody visualization. ELIZA was a main method in this study. RESULTS AND CONCLUSION Antibodies to several proteins were identified but significant differences were found only for AChR-(173-193). The results demonstrated the involvement of AChR and AChR-antibodies in the development of AD. Determination of antibodies to AChR-(173-193) may be a marker of AD and a method for specifying the diagnosis of AD.
Collapse
Affiliation(s)
- A V Kamynina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| | | | - D O Koroev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| | - T D Volkova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| | | | | | | | - O M Vol'pina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow
| |
Collapse
|
235
|
Minniti AN, Arrazola MS, Bravo-Zehnder M, Ramos F, Inestrosa NC, Aldunate R. The protein oxidation repair enzyme methionine sulfoxide reductase a modulates Aβ aggregation and toxicity in vivo. Antioxid Redox Signal 2015; 22:48-62. [PMID: 24988428 PMCID: PMC4270145 DOI: 10.1089/ars.2013.5803] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
AIMS To examine the role of the enzyme methionine sulfoxide reductase A-1 (MSRA-1) in amyloid-β peptide (Aβ)-peptide aggregation and toxicity in vivo, using a Caenorhabditis elegans model of the human amyloidogenic disease inclusion body myositis. RESULTS MSRA-1 specifically reduces oxidized methionines in proteins. Therefore, a deletion of the msra-1 gene was introduced into transgenic C. elegans worms that express the Aβ-peptide in muscle cells to prevent the reduction of oxidized methionines in proteins. In a constitutive transgenic Aβ strain that lacks MSRA-1, the number of amyloid aggregates decreases while the number of oligomeric Aβ species increases. These results correlate with enhanced synaptic dysfunction and mislocalization of the nicotinic acetylcholine receptor ACR-16 at the neuromuscular junction (NMJ). INNOVATION This approach aims at modulating the oxidation of Aβ in vivo indirectly by dismantling the methionine sulfoxide repair system. The evidence presented here shows that the absence of MSRA-1 influences Aβ aggregation and aggravates locomotor behavior and NMJ dysfunction. The results suggest that therapies which boost the activity of the Msr system could have a beneficial effect in managing amyloidogenic pathologies. CONCLUSION The absence of MSRA-1 modulates Aβ-peptide aggregation and increments its deleterious effects in vivo.
Collapse
Affiliation(s)
- Alicia N Minniti
- 1 Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | | | | | | | | | | |
Collapse
|
236
|
Utility of Autoantibodies as Biomarkers for Diagnosis and Staging of Neurodegenerative Diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 122:1-51. [DOI: 10.1016/bs.irn.2015.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
237
|
Shen J, Wu J. Nicotinic Cholinergic Mechanisms in Alzheimer's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 124:275-92. [PMID: 26472533 DOI: 10.1016/bs.irn.2015.08.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by increased accumulation of Aβ and degeneration of cholinergic signaling between basal forebrain and hippocampus. Nicotinic acetylcholine receptors (nAChRs) are important mediators of cholinergic signaling in modulation of learning and memory function. Accumulating lines of evidence indicate that a nAChR subtype, α7 receptor (α7-nAChR), plays an important role in modulations of excitatory neurotransmitter release, improvement of learning and memory ability, and enhancement of cognitive function. Importantly, the expression and function of α7-nAChRs is altered in the brain of AD animal models and AD patients, suggesting that this nAChR subtype participates in AD pathogenesis and may serve as a novel therapeutic target for AD treatment. However, the mechanisms underlying the role of α7-nAChRs in AD pathogenesis are very complex, and either neuroprotective effects or neurotoxic effects may occur through the α7-nAChRs. These effects depend on the levels of α7-nAChR expression and function, disease stages, or the use of α7-nAChR agonists, antagonists, or allosteric modulators. In this chapter, we summarize recent progresses in the roles of α7-nAChRs played in AD pathogenesis and therapy.
Collapse
Affiliation(s)
- Jianxin Shen
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jie Wu
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong, China; Divisions of Neurology and Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix Arizona, USA.
| |
Collapse
|
238
|
Qi Y, Klyubin I, Harney SC, Hu N, Cullen WK, Grant MK, Steffen J, Wilson EN, Do Carmo S, Remy S, Fuhrmann M, Ashe KH, Cuello AC, Rowan MJ. Longitudinal testing of hippocampal plasticity reveals the onset and maintenance of endogenous human Aß-induced synaptic dysfunction in individual freely behaving pre-plaque transgenic rats: rapid reversal by anti-Aß agents. Acta Neuropathol Commun 2014; 2:175. [PMID: 25540024 PMCID: PMC4293804 DOI: 10.1186/s40478-014-0175-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 12/04/2014] [Indexed: 11/26/2022] Open
Abstract
Long before synaptic loss occurs in Alzheimer’s disease significant harbingers of disease may be detected at the functional level. Here we examined if synaptic long-term potentiation is selectively disrupted prior to extracellular deposition of Aß in a very complete model of Alzheimer’s disease amyloidosis, the McGill-R-Thy1-APP transgenic rat. Longitudinal studies in freely behaving animals revealed an age-dependent, relatively rapid-onset and persistent inhibition of long-term potentiation without a change in baseline synaptic transmission in the CA1 area of the hippocampus. Thus the ability of a standard 200 Hz conditioning protocol to induce significant NMDA receptor-dependent short- and long-term potentiation was lost at about 3.5 months of age and this deficit persisted for at least another 2–3 months, when plaques start to appear. Consistent with in vitro evidence for a causal role of a selective reduction in NMDA receptor-mediated synaptic currents, the deficit in synaptic plasticity in vivo was associated with a reduction in the synaptic burst response to the conditioning stimulation and was overcome using stronger 400 Hz stimulation. Moreover, intracerebroventricular treatment for 3 days with an N-terminally directed monoclonal anti- human Aß antibody, McSA1, transiently reversed the impairment of synaptic plasticity. Similar brief treatment with the BACE1 inhibitor LY2886721 or the γ-secretase inhibitor MRK-560 was found to have a comparable short-lived ameliorative effect when tracked in individual rats. These findings provide strong evidence that endogenously generated human Aß selectively disrupts the induction of long-term potentiation in a manner that enables potential therapeutic options to be assessed longitudinally at the pre-plaque stage of Alzheimer’s disease amyloidosis.
Collapse
|
239
|
Deardorff WJ, Shobassy A, Grossberg GT. Safety and clinical effects of EVP-6124 in subjects with Alzheimer's disease currently or previously receiving an acetylcholinesterase inhibitor medication. Expert Rev Neurother 2014; 15:7-17. [PMID: 25495510 DOI: 10.1586/14737175.2015.995639] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Alzheimer's disease (AD) is a prevalent and currently incurable brain disease whose impact will continue to rise as the population ages. With limited treatment options, a variety of experimental therapies are currently in clinical trials. EVP-6124 (encenicline) is an α7 nicotinic acetylcholine receptor partial agonist under investigation for the symptomatic treatment of AD. EVP-6124 activates the α7 nicotinic acetylcholine receptor at low nanomolar brain concentrations and improves memory performance in rats. Treatment with EVP-6124 in Phase I and II trials involving patients with mild-to-moderate AD was well tolerated and showed statistically significant improvements compared with placebo on cognitive and functional measures. Two Phase III trials under the title COGNITIV AD will assess the efficacy and tolerability of EVP-6124 in patients with mild-to-moderate AD. Based on the completed clinical trials and proposed mechanism of action, EVP-6124 would appear to be a good candidate for therapy in combination with cholinesterase inhibitors.
Collapse
|
240
|
Lombardo S, Maskos U. Role of the nicotinic acetylcholine receptor in Alzheimer's disease pathology and treatment. Neuropharmacology 2014; 96:255-62. [PMID: 25514383 DOI: 10.1016/j.neuropharm.2014.11.018] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/11/2014] [Accepted: 11/24/2014] [Indexed: 11/26/2022]
Abstract
Alzheimer's Disease (AD) is the major form of senile dementia, characterized by neuronal loss, extracellular deposits, and neurofibrillary tangles. It is accompanied by a loss of cholinergic tone, and acetylcholine (ACh) levels in the brain, which were hypothesized to be responsible for the cognitive decline observed in AD. Current medication is restricted to enhancing cholinergic signalling for symptomatic treatment of AD patients. The nicotinic acetylcholine receptor family (nAChR) and the muscarinic acetylcholine receptor family (mAChR) are the target of ACh in the brain. Both families of receptors are affected in AD. It was demonstrated that amyloid beta (Aβ) interacts with nAChRs. Here we discuss how Aβ activates or inhibits nAChRs, and how this interaction contributes to AD pathology. We will discuss the potential role of nAChRs as therapeutic targets. This article is part of the Special Issue entitled 'The Nicotinic Acetylcholine Receptor: From Molecular Biology to Cognition'.
Collapse
Affiliation(s)
- Sylvia Lombardo
- Département de Neuroscience, Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Paris Cedex 15, France; CNRS, UMR 3571, Paris, France.
| | - Uwe Maskos
- Département de Neuroscience, Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Paris Cedex 15, France; CNRS, UMR 3571, Paris, France
| |
Collapse
|
241
|
The Binding Receptors of Aβ: an Alternative Therapeutic Target for Alzheimer's Disease. Mol Neurobiol 2014; 53:455-471. [PMID: 25465238 DOI: 10.1007/s12035-014-8994-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/06/2014] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which causes the deterioration of memory and other cognitive abilities of the elderly. Previous lines of research have shown that Aβ is an essential factor in AD pathology and the soluble oligomeric species of Aβ peptide is presumed to be the drivers of synaptic impairment in AD. However, the exact mechanisms underlying Aβ-induced synapse dysfunction are still not fully understood. Recently, increasing evidence suggests that some potential receptors which bind specifically with Aβ may play important roles in inducing the toxicity of the neurons in AD pathology. These receptors include the cellular prion protein (PrPc), the α7 nicotinic acetylcholine receptor (α7nAChR), the p75 neurotrophin receptor (p75(NTR)), the beta-adrenergic receptors (β-ARs), the Eph receptors, the paired immunoglobulin-like receptor B (PirB), the PirB's human ortholog receptor (LilrB2), and the Fcγ receptor II-b (FcγRIIb). This review summarizes the characters of these prominent receptors and how the bindings of them with Aβ inhibit the LTP, decrease the number of dendritic spine, damage the neurons, and so on in AD pathogenesis. Blocking or rescuing these receptors may have significant importance for AD treatments.
Collapse
|
242
|
Kam TI, Gwon Y, Jung YK. Amyloid beta receptors responsible for neurotoxicity and cellular defects in Alzheimer's disease. Cell Mol Life Sci 2014; 71:4803-13. [PMID: 25151011 PMCID: PMC11113744 DOI: 10.1007/s00018-014-1706-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/24/2014] [Accepted: 08/13/2014] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease. Although a major cause of AD is the accumulation of amyloid-β (Aβ) peptide that induces neuronal loss and cognitive impairments, our understanding of its neurotoxic mechanisms is limited. Recent studies have identified putative Aβ-binding receptors that mediate Aβ neurotoxicity in cells and models of AD. Once Aβ interacts with a receptor, a toxic signal is transduced into neurons, resulting in cellular defects including endoplasmic reticulum stress and mitochondrial dysfunction. In addition, Aβ can also be internalized into neurons through unidentified Aβ receptors and induces malfunction of subcellular organelles, which explains some part of Aβ neurotoxicity. Understanding the neurotoxic signaling initiated by Aβ-receptor binding and cellular defects provide insight into new therapeutic windows for AD. In the present review, we summarize the findings on Aβ-binding receptors and the neurotoxicity of oligomeric Aβ.
Collapse
Affiliation(s)
- Tae-In Kam
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| | - Youngdae Gwon
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| | - Yong-Keun Jung
- Global Research Laboratory, School of Biological Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-747 Korea
| |
Collapse
|
243
|
Abstract
Alzheimer's disease is the most common neurodegenerative disorder in the world. Its most significant symptoms are memory loss and decrease in cognition. Alzheimer's disease is characterized by aggregation of two proteins in the brain namely Aβ (amyloid β) and tau. Recent evidence suggests that the interaction of soluble Aβ with nAChR (nicotinic acetylcholine receptors) contributes to disease progression. In this study, we determine the NMR structure of an Aβ17–34 peptide solubilized by the addition of two glutamic acids at each terminus. Our results indicate that the Aβ peptide adopts an α-helical structure for residues 19–26 and 28–33. The α-helical structure is broken around residues S26, N27 and K28, which form a kink in the helical conformation. This α-helix was not described earlier in an aqueous solution without organic solvents, and at physiological conditions (pH 7). These data are in agreement with Aβ adopting an α-helical conformation in the membrane before polymerizing into amyloid β-sheets and provide insight into the intermediate state of Aβ in Alzheimer's disease.
Collapse
|
244
|
Stancu IC, Vasconcelos B, Terwel D, Dewachter I. Models of β-amyloid induced Tau-pathology: the long and "folded" road to understand the mechanism. Mol Neurodegener 2014; 9:51. [PMID: 25407337 PMCID: PMC4255655 DOI: 10.1186/1750-1326-9-51] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 10/14/2014] [Indexed: 02/28/2023] Open
Abstract
The amyloid cascade hypothesis has been the prevailing hypothesis in Alzheimer’s Disease research, although the final and most wanted proof i.e. fully successful anti-amyloid clinical trials in patients, is still lacking. This may require a better in depth understanding of the cascade. Particularly, the exact toxic forms of Aβ and Tau, the molecular link between them and their respective contributions to the disease process need to be identified in detail. Although the lack of final proof has raised substantial criticism on the hypothesis per se, accumulating experimental evidence in in vitro models, in vivo models and from biomarkers analysis in patients supports the amyloid cascade and particularly Aβ-induced Tau-pathology, which is the focus of this review. We here discuss available models that recapitulate Aβ-induced Tau-pathology and review some potential underlying mechanisms. The availability and diversity of these models that mimic the amyloid cascade partially or more complete, provide tools to study remaining questions, which are crucial for development of therapeutic strategies for Alzheimer’s Disease.
Collapse
Affiliation(s)
| | | | | | - Ilse Dewachter
- Catholic University of Louvain, Institute of Neuroscience, Alzheimer Dementia, Av, E, Mounier 53, Av, Hippocrate 54, B-1200 Brussels, Belgium.
| |
Collapse
|
245
|
Tu S, Okamoto SI, Lipton SA, Xu H. Oligomeric Aβ-induced synaptic dysfunction in Alzheimer's disease. Mol Neurodegener 2014; 9:48. [PMID: 25394486 PMCID: PMC4237769 DOI: 10.1186/1750-1326-9-48] [Citation(s) in RCA: 414] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/08/2014] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating disease characterized by synaptic and neuronal loss in the elderly. Compelling evidence suggests that soluble amyloid-β peptide (Aβ) oligomers induce synaptic loss in AD. Aβ-induced synaptic dysfunction is dependent on overstimulation of N-methyl-D-aspartate receptors (NMDARs) resulting in aberrant activation of redox-mediated events as well as elevation of cytoplasmic Ca2+, which in turn triggers downstream pathways involving phospho-tau (p-tau), caspases, Cdk5/dynamin-related protein 1 (Drp1), calcineurin/PP2B, PP2A, Gsk-3β, Fyn, cofilin, and CaMKII and causes endocytosis of AMPA receptors (AMPARs) as well as NMDARs. Dysfunction in these pathways leads to mitochondrial dysfunction, bioenergetic compromise and consequent synaptic dysfunction and loss, impaired long-term potentiation (LTP), and cognitive decline. Evidence also suggests that Aβ may, at least in part, mediate these events by causing an aberrant rise in extrasynaptic glutamate levels by inhibiting glutamate uptake or triggering glutamate release from glial cells. Consequent extrasynaptic NMDAR (eNMDAR) overstimulation then results in synaptic dysfunction via the aforementioned pathways. Consistent with this model of Aβ-induced synaptic loss, Aβ synaptic toxicity can be partially ameliorated by the NMDAR antagonists (such as memantine and NitroMemantine). PSD-95, an important scaffolding protein that regulates synaptic distribution and activity of both NMDA and AMPA receptors, is also functionally disrupted by Aβ. PSD-95 dysregulation is likely an important intermediate step in the pathological cascade of events caused by Aβ. In summary, Aβ-induced synaptic dysfunction is a complicated process involving multiple pathways, components and biological events, and their underlying mechanisms, albeit as yet incompletely understood, may offer hope for new therapeutic avenues.
Collapse
Affiliation(s)
- Shichun Tu
- Neuroscience and Aging Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
246
|
Abstract
Alzheimer's disease (AD) is the most common form of dementia among older persons. Pathognomonic hallmarks of the disease include the development of amyloid senile plaques and deposits of neurofibrillary tangles. These changes occur in the brain long before the clinical manifestations of AD (cognitive impairment in particular) become apparent. Nicotinic acetylcholine receptors (AChRs), particularly the α7 subtype, are highly expressed in brain regions relevant to cognitive and memory functions and involved in the processing of sensory information. There is strong evidence that implicates the participation of AChRs in AD. This review briefly introduces current strategies addressing the pathophysiologic findings (amyloid-β-peptide plaques, neurofibrillary tangles) and then focuses on more recent efforts of pharmacologic intervention in AD, specifically targeted to the α7 AChR. Whereas cholinesterase inhibitors such as donepezil, galantamine, or rivastigmine, together with the non-competitive N-methyl-D-aspartate receptor antagonist memantine are at the forefront of present-day clinical intervention for AD, new insights into AChR molecular pharmacology are bringing other drugs, directed at AChRs, to center stage. Among these are the positive allosteric modulators that selectively target α7 AChRs and are aimed at unleashing the factors that hinder agonist-mediated, α7 AChR channel activation. This calls for more detailed knowledge of the distribution, functional properties, and involvement of AChRs in various signaling cascades-together with the corresponding abnormalities in all these properties-to be able to engineer strategies in drug design and evaluate the therapeutic possibilities of new compounds targeting this class of neurotransmitter receptors.
Collapse
|
247
|
Zheng X, Xie Z, Zhu Z, Liu Z, Wang Y, Wei L, Yang H, Yang H, Liu Y, Bi J. Methyllycaconitine alleviates amyloid-β peptides-induced cytotoxicity in SH-SY5Y cells. PLoS One 2014; 9:e111536. [PMID: 25360664 PMCID: PMC4216102 DOI: 10.1371/journal.pone.0111536] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 10/04/2014] [Indexed: 12/01/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic progressive neurodegenerative disorder. As the most common form of dementia, it affects more than 35 million people worldwide and is increasing. Excessive extracellular deposition of amyloid-β peptide (Aβ) is a pathologic feature of AD. Accumulating evidence indicates that macroautophagy is involved in the pathogenesis of AD, but its exact role is still unclear. Although major findings on the molecular mechanisms have been reported, there are still no effective treatments to prevent, halt, or reverse Alzheimer's disease. In this study, we investigated whether Aβ25–35 could trigger an autophagy process and inhibit the growth of SH-SY5Y cells. Furthermore, we examined the effect of methyllycaconitine (MLA) on the cytotoxity of Aβ25–35. MLA had a protective effect against cytotoxity of Aβ, which may be related to its inhibition of Aβ-induced autophagy and the involvement of the mammalian target of rapamycin pathway. Moreover, MLA had a good safety profile. MLA treatment may be a promising therapeutic tool for AD.
Collapse
Affiliation(s)
- XiaoLei Zheng
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - ZhaoHong Xie
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - ZhengYu Zhu
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - Zhen Liu
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - Yun Wang
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - LiFei Wei
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - Hui Yang
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - HongNa Yang
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - YiQing Liu
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
| | - JianZhong Bi
- Department of Neural Medicine, Second Hospital of Shandong University, Jinan, China
- Institute of Neurology, Shandong University, Jinan, China
- Key Laboratory of Translational Medicine on Neurological Degenerative Disease in Universities of Shandong (Shandong University), Jinan, China
- * E-mail: .
| |
Collapse
|
248
|
Deutsch SI, Burket JA, Benson AD. Targeting the α7 nicotinic acetylcholine receptor to prevent progressive dementia and improve cognition in adults with Down's syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:131-9. [PMID: 24865150 DOI: 10.1016/j.pnpbp.2014.05.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 12/11/2022]
Abstract
As persons with Down's syndrome (DS) age into the third decade and beyond, they develop Alzheimer's disease (AD)-like histopathological changes in brain and may manifest progressive worsening of adaptive functions. Increasingly, persons with DS have near-normal to normal life spans; thus, it has become a therapeutic imperative to preserve adaptive functions and ability to live as independently as possible in the least restrictive environment throughout adulthood. Data suggest that these histopathological changes and worsening adaptive functions result, at least in part, from the binding of the amyloidogenic Aβ1-42 peptide to α7 nicotinic acetylcholine receptors (α7nAChRs) on the surface of neurons, which can lead to the internalization of the tightly-bound complex and cell lysis. Pharmacotherapeutic targeting of the α7nAChR may inhibit the creation of the Aβ1-42-α7nAChR complex, which has been observed both intraneuronally and as a component of the amyloid plaque seen in AD. Additionally, selective α7nAChR agonists may improve memory and cognition independently of their potential ability to attenuate the cytotoxicity of Aβ1-42 and retard the deposition of amyloid plaques in adults with DS. However, there are conflicting data supporting an antagonist strategy to improve cognition in the presence of elevated levels of Aβ amyloidogenic peptides, as well as to prevent emergence of pyramidal neuron hyperexcitability. A major challenge to the implementation of clinical trials of targeted α7nAChR interventions in adults with DS will be the ability to detect medication-induced changes in cognition in the context of intellectual disability. The Review will consider some of the current evidence supporting both the role of the Aβ1-42-α7nAChR complex in the pathogenesis of the AD-like histopathology in adult persons with DS, and pharmacotherapeutic interventions with α7nAChR agonists.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences1, Eastern Virginia Medical School, Norfolk, VA, United States.
| | - Jessica A Burket
- Department of Psychiatry and Behavioral Sciences1, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Andrew D Benson
- Department of Psychiatry and Behavioral Sciences1, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
249
|
Paulo JA. Nicotine alters the proteome of two human pancreatic duct cell lines. JOP : JOURNAL OF THE PANCREAS 2014; 15:465-74. [PMID: 25262714 DOI: 10.6092/1590-8577/2559] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CONTEXT Cigarette smoking is a known risk factor of pancreatic disease. Nicotine--a major cigarette tobacco component--can traffic through the circulatory system and may induce fibrosis and metastasis, hallmarks of chronic pancreatitis and pancreatic adenocarcinoma, respectively. However, at the biomolecular level, particularly in pancreatic research, the effects of nicotine remain unresolved. METHODS The effects of nicotine on the proteomes of two pancreatic duct cell lines-an immortalized normal cell line (HPNE) and a cancer cell line (PanC1)- were investigated using mass spectrometry-based proteomics. For each cell line, the global proteomes of cells exposed to nicotine for 24 hrs were compared with untreated cells in triplicate using 6-plex tandem mass tag-based isobaric labeling techniques. RESULTS Over 5,000 proteins were detected per cell line. Of these, over 900 proteins were differentially abundant with statistical significance (corrected P-value <0.01) upon nicotine treatment, 57 of which were so in both cell lines. Amyloid precursor protein, previously observed to increase expression in pancreatic stellate cells when exposed to nicotine, was also up-regulated in both cell lines.In general, the two cell lines varied in the classes of proteins altered by nicotine treatment, supporting published evidence that nicotine may play different roles in the initiation and progression of pancreatic disease. CONCLUSIONS Understanding the underlying mechanisms associating nicotine with pancreatic function is paramount to intervention aiming to retard, arrest, or ameliorate pancreatic disease.
Collapse
Affiliation(s)
- Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, USA.
| |
Collapse
|
250
|
Bordia T, McGregor M, Papke RL, Decker MW, McIntosh JM, Quik M. The α7 nicotinic receptor agonist ABT-107 protects against nigrostriatal damage in rats with unilateral 6-hydroxydopamine lesions. Exp Neurol 2014; 263:277-84. [PMID: 25261754 DOI: 10.1016/j.expneurol.2014.09.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/05/2014] [Accepted: 09/16/2014] [Indexed: 02/01/2023]
Abstract
The finding that smoking is inversely correlated with Parkinson's disease and that nicotine attenuates nigrostriatal damage in Parkinsonian animals supports the idea that nicotine may be neuroprotective. Nicotine is thought to exert this effect by acting at nicotinic receptors (nAChRs), including the α7 subtype. The objective of this study was twofold: first, to test the protective potential of ABT-107, an agonist with high selectivity for α7 nAChRs; and second, to investigate its cellular mechanism of action. Rats were implanted with minipumps containing ABT-107 (0.25mg/kg/d). In addition, we tested the effect of nicotine (1mg/kg/d) as a positive control, and also DMXB (2mg/kg/d) which acts primarily with α7 but also α4β2* nAChRs. Two weeks after minipump placement, the rats were lesioned by unilateral administration of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle. Lesioning alone decreased contralateral forelimb use and adjusted stepping, two measures of Parkinsonism. ABT-107 and nicotine treatment significantly improved these behaviors at all weeks tested, with variable improvement with DMXB. We next investigated the cellular mechanism involved. The striatal dopamine transporter (DAT), a marker of dopaminergic integrity, was reduced ~70% with lesioning. ABT-107 or nicotine treatment significantly increased DAT levels in lesioned striatum; these drugs did not alter DAT levels in intact striatum. ABT-107 and nicotine also significantly improved basal dopamine release from lesioned striatum, as well as nicotine-stimulated dopamine release mediated via α4β2* and α6β2* nAChRs. These data suggest that α7 nAChR agonists may improve motor behaviors associated with nigrostriatal damage by enhancing striatal dopaminergic function.
Collapse
Affiliation(s)
- Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Matthew McGregor
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 3261, USA
| | - Michael W Decker
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL 60064-6125, USA
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA; Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA.
| |
Collapse
|