101
|
Majdi A, Sadigh-Eteghad S, Rahigh Aghsan S, Farajdokht F, Vatandoust SM, Namvaran A, Mahmoudi J. Amyloid-β, tau, and the cholinergic system in Alzheimer's disease: seeking direction in a tangle of clues. Rev Neurosci 2020; 31:391-413. [PMID: 32017704 DOI: 10.1515/revneuro-2019-0089] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/22/2019] [Indexed: 12/14/2022]
Abstract
The link between histopathological hallmarks of Alzheimer's disease (AD), i.e. amyloid plaques, and neurofibrillary tangles, and AD-associated cognitive impairment, has long been established. However, the introduction of interactions between amyloid-beta (Aβ) as well as hyperphosphorylated tau, and the cholinergic system to the territory of descriptive neuropathology has drastically changed this field by adding the theory of synaptic neurotransmission to the toxic pas de deux in AD. Accumulating data show that a multitarget approach involving all amyloid, tau, and cholinergic hypotheses could better explain the evolution of events happening in AD. Various species of both Aβ and tau could be traced in cholinergic neurons of the basal forebrain system early in the course of the disease. These molecules induce degeneration in the neurons of this system. Reciprocally, aberrant cholinergic system modulation promotes changes in amyloid precursor protein (APP) metabolism and tau phosphorylation, resulting in neurotoxicity, neuroinflammation, and neuronal death. Altogether, these changes may better correlate with the clinical findings and cognitive impairment detected in AD patients. Failure of several of Aβ- and tau-related therapies further highlights the need for special attention to molecules that target all of these mentioned pathologic changes. Another noteworthy fact here is that none of the popular hypotheses of AD such as amyloidopathy or tauopathy seem to be responsible for the changes observed in AD alone. Thus, the main culprit should be sought higher in the stream somewhere in APP metabolism or Wnt signaling in the cholinergic system of the basal forebrain. Future studies should target these pathological events.
Collapse
Affiliation(s)
- Alireza Majdi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Sepideh Rahigh Aghsan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Seyed Mehdi Vatandoust
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Ali Namvaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51368, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz 51368, Iran
| |
Collapse
|
102
|
Findley CA, Bartke A, Hascup KN, Hascup ER. Amyloid Beta-Related Alterations to Glutamate Signaling Dynamics During Alzheimer's Disease Progression. ASN Neuro 2020; 11:1759091419855541. [PMID: 31213067 PMCID: PMC6582288 DOI: 10.1177/1759091419855541] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) ranks sixth on the Centers for Disease Control and Prevention Top 10 Leading Causes of Death list for 2016, and the Alzheimer’s Association attributes 60% to 80% of dementia cases as AD related. AD pathology hallmarks include accumulation of senile plaques and neurofibrillary tangles; however, evidence supports that soluble amyloid beta (Aβ), rather than insoluble plaques, may instigate synaptic failure. Soluble Aβ accumulation results in depression of long-term potentiation leading to cognitive deficits commonly characterized in AD. The mechanisms through which Aβ incites cognitive decline have been extensively explored, with a growing body of evidence pointing to modulation of the glutamatergic system. The period of glutamatergic hypoactivation observed alongside long-term potentiation depression and cognitive deficits in later disease stages may be the consequence of a preceding period of increased glutamatergic activity. This review will explore the Aβ-related changes to the tripartite glutamate synapse resulting in altered cell signaling throughout disease progression, ultimately culminating in oxidative stress, synaptic dysfunction, and neuronal loss.
Collapse
Affiliation(s)
- Caleigh A Findley
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- 3 Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA.,4 Department of Molecular Biology, Microbiology & Biochemistry, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- 1 Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, USA.,2 Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
103
|
Functional characterization of multifunctional ligands targeting acetylcholinesterase and alpha 7 nicotinic acetylcholine receptor. Biochem Pharmacol 2020; 177:114010. [PMID: 32360492 DOI: 10.1016/j.bcp.2020.114010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/28/2020] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with cholinergic dysfunction, provoking memory loss and cognitive dysfunction in elderly patients. The cholinergic hypothesis provided over the years with molecular targets for developing palliative treatments for AD, acting on the cholinergic system, namely, acetylcholinesterase and α7 nicotinic acetylcholine receptor (α7 nAChR). In our synthetic work, we used "click-chemistry" to synthesize two Multi Target Directed Ligands (MTDLs) MB105 and MB118 carrying tacrine and quinuclidine scaffolds which are known for their anticholinesterase and α7 nAChR agonist activities, respectively. Both, MB105 and MB118, inhibit human acetylcholinesterase and human butyrylcholinesterase in the nanomolar range. Electrophysiological recordings on Xenopus laevis oocytes expressing human α7 nAChR showed that MB105 and MB118 acted as partial agonists of the referred nicotinic receptor, albeit, with different potencies despite their similar structure. The different substitution at C-3 on the 2,3-disubstituted quinuclidine scaffold may account for the significantly lower potency of MB118 compared to MB105. Electrophysiological recordings also showed that the tacrine precursor MB320 behaved as a competitive antagonist of human α7 nAChR, in the micromolar range, while the quinuclidine synthetic precursor MB099 acted as a partial agonist. Taken all together, MB105 behaved as a partial agonist of α7 nAChR at concentrations where it completely inhibited human acetylcholinesterase activity paving the way for the design of novel MTDLs for palliative treatment of AD.
Collapse
|
104
|
Finnie PSB, Nader K. Amyloid Beta Secreted during Consolidation Prevents Memory Malleability. Curr Biol 2020; 30:1934-1940.e4. [PMID: 32243855 DOI: 10.1016/j.cub.2020.02.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/21/2020] [Accepted: 02/26/2020] [Indexed: 12/21/2022]
Abstract
Memory allows organisms to predict future events based on their prior sampling of the world. Rather than faithfully encoding each detail of related episodes, the brain is thought to incrementally construct probabilistic estimates of environmental statistics that are re-evaluated each time relevant events are encountered [1]. When faced with evidence that does not adequately fit mnemonic predictions, a process called reconsolidation can alter relevant memories to better recapitulate ongoing experience [2]. Conversely, when an ongoing event matches well-established predictions, reactivated memories tend to remain stable [3, 4]. In part, the brain may confer selective mnemonic stability by shifting cell-intrinsic mechanisms of plasticity induction [5], which could serve to constrain maladaptive updating of reliably predictive representations during anomalous events. Based on evidence of decreased cognitive flexibility and restricted synaptic plasticity in later life [6], we hypothesized that some prevalent age-associated neurobiological changes might in fact contribute to mnemonic stability [7]. Specifically, we predicted that amyloid beta (Aβ)-a peptide that often accumulates in the brains of individuals expressing senescent dementia [8-10]-is required for memory stabilization. Indeed, we observe elevated soluble Aβx-42 concentrations in the amygdala shortly after young adult rats form reconsolidation-resistant auditory fear memories. Suppressing secretases required for Aβ production immediately after learning prevents mnemonic stabilization, rendering these memories vulnerable to disruption by post-reactivation amnestic treatments. Thus, the seemingly pathogenic Aβ42 peptide may serve an adaptive physiological function during memory consolidation by engaging mechanisms that protect reliably predictive representations against subsequent modification.
Collapse
Affiliation(s)
- Peter S B Finnie
- Psychology Department, McGill University, 1205 Avenue Dr. Penfield, Montreal, QC H3A 1B1, Canada.
| | - Karim Nader
- Psychology Department, McGill University, 1205 Avenue Dr. Penfield, Montreal, QC H3A 1B1, Canada.
| |
Collapse
|
105
|
Lin MW, Chen YH, Yang HB, Lin CC, Hung SY. Galantamine Inhibits Aβ 1-42-Induced Neurotoxicity by Enhancing α7nAChR Expression as a Cargo Carrier for LC3 Binding and Aβ 1-42 Engulfment During Autophagic Degradation. Neurotherapeutics 2020; 17:676-689. [PMID: 31823156 PMCID: PMC7283419 DOI: 10.1007/s13311-019-00803-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Despite Alzheimer's disease (AD) being the most common neurodegenerative disorder worldwide, no FDA-approved disease-modifying treatments have been approved for this condition since 2003. Neuronal-type alpha7 nicotinic acetylcholine receptors (α7nAChRs) play an essential role in cognitive functions, binding with extracellular β-amyloid (Aβ plaques) and inhibiting Aβ-induced neurotoxicity. α7nAChRs are impaired early in the course of AD; drugs targeting α7nAChRs are being hotly pursued as a treatment of AD. Encenicline, a partial selective agonist of α7nAChR and modulator of acetylcholine, failed in phase III trials because of gastrointestinal side effects. We, therefore, evaluated the efficacy of galantamine, a positive allosteric modulator at α7nAChRs and an acetylcholinesterase inhibitor, that has been used since 2000 as first-line treatment of mild-to-moderate dementia. This study highlights an important new benefit with galantamine. We found that galantamine inhibits Aβ1-42-induced apoptosis by activating the JNK signaling pathway, thus enhancing α7nAChR expression, and also inhibits the Akt pathway, which further increases autophagosome biogenesis and autophagy. These effects can be reproduced by α7nAChR overexpression in the absence of galantamine. Importantly, the α7 subunit protein sequence of α7nAChRs contains 3 LC3-interacting regions; our immunoprecipitation data show that α7 binds with the autophagosomal marker protein LC3. This is the first report to provide evidence showing that the cell surface receptor α7nAChR acts as a cargo carrier for LC3 binding for Aβ1-42 sequestration to autophagosomes, suggesting a novel mechanism for the neuroprotective efficacy of galantamine in AD.
Collapse
Affiliation(s)
- Ming-Wei Lin
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, Kaohsiung, 82445, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan
| | - Han-Ben Yang
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung, 40249, Taiwan
| | - Chi Chien Lin
- Department of Life Sciences, Institute of Biomedical Science, National Chung Hsing University, Taichung, 40249, Taiwan
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
- Division of Colorectal Surgery, China Medical University Hospital, Taichung, 40447, Taiwan.
| |
Collapse
|
106
|
Coughlin JM, Rubin LH, Du Y, Rowe SP, Crawford JL, Rosenthal HB, Frey SM, Marshall ES, Shinehouse LK, Chen A, Speck CL, Wang Y, Lesniak WG, Minn I, Bakker A, Kamath V, Smith GS, Albert MS, Azad BB, Dannals RF, Horti A, Wong DF, Pomper MG. High Availability of the α7-Nicotinic Acetylcholine Receptor in Brains of Individuals with Mild Cognitive Impairment: A Pilot Study Using 18F-ASEM PET. J Nucl Med 2020; 61:423-426. [PMID: 31420499 PMCID: PMC9374031 DOI: 10.2967/jnumed.119.230979] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2019] [Indexed: 01/05/2023] Open
Abstract
Emerging evidence supports a hypothesized role for the α7-nicotinic acetylcholine receptor (α7-nAChR) in the pathophysiology of Alzheimer's disease. 18F-ASEM (3-(1,4-diazabicyclo[3.2.2]nonan-4-yl)-6-18F-fluorodibenzo[b,d]thiophene 5,5-dioxide) is a radioligand for estimating the availability of α7-nAChR in the brain in vivo with PET. Methods: In this cross-sectional study, 14 patients with mild cognitive impairment (MCI), a prodromal stage to dementia, and 17 cognitively intact, elderly controls completed 18F-ASEM PET. For each participant, binding in each region of interest was estimated using Logan graphical analysis with a metabolite-corrected arterial input function. Results: Higher 18F-ASEM binding was observed in MCI patients than in controls across all regions, supporting higher availability of α7-nAChR in MCI. 18F-ASEM binding was not associated with verbal memory in this small MCI sample. Conclusion: These data support use of 18F-ASEM PET to examine further the relationship between α7-nAChR availability and MCI.
Collapse
Affiliation(s)
- Jennifer M Coughlin
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Leah H Rubin
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Medical Institutions, Baltimore, Maryland; and
| | - Yong Du
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Steven P Rowe
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jeffrey L Crawford
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Hailey B Rosenthal
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Sarah M Frey
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Erica S Marshall
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Laura K Shinehouse
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Allen Chen
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Caroline L Speck
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Yuchuan Wang
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Wojciech G Lesniak
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Il Minn
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Arnold Bakker
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Vidyulata Kamath
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Gwenn S Smith
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Babak Behnam Azad
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Robert F Dannals
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Andrew Horti
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Dean F Wong
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Martin G Pomper
- Department of Psychiatry, Johns Hopkins Medical Institutions, Baltimore, Maryland
- Department of Radiology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
107
|
Volloch V, Olsen B, Rits S. Alzheimer's Disease is Driven by Intraneuronally Retained Beta-Amyloid Produced in the AD-Specific, βAPP-Independent Pathway: Current Perspective and Experimental Models for Tomorrow. ANNALS OF INTEGRATIVE MOLECULAR MEDICINE 2020; 2:90-114. [PMID: 32617536 PMCID: PMC7331974 DOI: 10.33597/aimm.02-1007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A view of the origin and progression of Alzheimer's disease, AD, prevailing until now and formalized as the Amyloid Cascade Hypothesis theory, maintains that the disease is initiated by overproduction of beta-amyloid, Aβ, which is generated solely by the Aβ precursor protein, βAPP, proteolytic pathway and secreted from the cell. Consequent extracellular accumulation of Aβ triggers a cascade of molecular and cellular events leading to neurodegeneration that starts early in life, progresses as one prolonged process, builds up for decades, and culminates in symptomatic manifestations of the disease late in life. In this paradigm, a time window for commencement of therapeutic intervention is small and accessible only early in life. The outlook introduced in the present study is fundamentally different. It posits that the βAPP proteolytic/secretory pathway of Aβ production causes AD in humans no more than it does in either short- or long-lived non-human mammals that share this pathway with humans, accumulate beta-amyloid as they age, but do not develop the disease. Alzheimer's disease, according to this outlook, is driven by an additional powerful AD-specific pathway of Aβ production that operates in affected humans, is completely independent of the βAPP precursor, and is not available in non-human mammals. The role of the βAPP proteolytic pathway in the disease in humans is activation of this additional AD-specific Aβ production pathway. This occurs through accumulation of intracellular Aβ, primarily via ApoE-assisted cellular uptake of secreted beta-amyloid, but also through retention of a fraction of Aβ produced in the βAPP proteolytic pathway. With time, accumulated intracellular Aβ triggers mitochondrial dysfunction. In turn, cellular stresses associated with mitochondrial dysfunction, including ER stress, activate a second, AD-specific, Aβ production pathway: Asymmetric RNA-dependent βAPP mRNA amplification; animal βAPP mRNA is ineligible for this process. In this pathway, every conventionally produced βAPP mRNA molecule serves potentially as a template for production of severely 5'-truncated mRNA encoding not the βAPP but its C99 fragment (hence "asymmetric"), the immediate precursor of Aβ. Thus produced, N-terminal signal peptide-lacking C99 is processed not in the secretory pathway on the plasma membrane, but at the intracellular membrane sites, apparently in a neuron-specific manner. The resulting Aβ is, therefore, not secreted but is retained intraneuronally and accumulates rapidly within the cell. Increased levels of intracellular Aβ augment mitochondrial dysfunction, which, in turn, sustains the activity of the βAPP mRNA amplification pathway. These self-propagating mutual Aβ overproduction/mitochondrial dysfunction feedback cycles constitute a formidable two-stroke engine, an engine that drives Alzheimer's disease. The present outlook envisions Alzheimer's disorder as a two-stage disease. The first stage is a slow process of intracellular beta-amyloid accumulation. It results neither in significant neurodegenerative damage, nor in manifestation of the disease. The second stage commences with the activation of the βAPP mRNA amplification pathway shortly before symptomatic onset of the disease, sharply increases the rate of Aβ generation and the extent of its intraneuronal accumulation, produces significant damages, triggers AD symptoms, and is fast. In this paradigm, the time window of therapeutic intervention is wide open, and preventive treatment can be initiated any time, even late in life, prior to commencement of the second stage of the disease. Moreover, there are good reasons to believe that with a drug blocking the βAPP mRNA amplification pathway, it would be possible not only to preempt the disease but also to stop and to reverse it even when early AD symptoms have already manifested. There are numerous experimental models of AD, all based on a notion of the exceptionality of βAPP proteolytic/secretory pathway in Aβ production in the disease. However, with no drug even remotely effective in Alzheimer's disease, a long list of candidate drugs that succeeded remarkably in animal models, yet failed utterly in human clinical trials of potential AD drugs, attests to the inadequacy of currently employed AD models. The concept of a renewable supply of beta-amyloid, produced in the βAPP mRNA amplification pathway and retained intraneuronally in Alzheimer's disease, explains spectacular failures of both BACE inhibition and Aβ-immunotherapy in human clinical trials. This concept also forms the basis of a new generation of animal and cell-based experimental models of AD, described in the present study. These models incorporate Aβ- or C99-encoding mRNA amplification pathways of Aβ production, as well as intracellular retention of their product, and can support not only further investigation of molecular mechanisms of AD but also screening for and testing of candidate drugs aimed at therapeutic targets suggested by the present study.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, USA
| | - Bjorn Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, USA
| | - Sophia Rits
- Division of Molecular Medicine, Children’s Hospital, Boston, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, USA
| |
Collapse
|
108
|
Hoskin JL, Al-Hasan Y, Sabbagh MN. Nicotinic Acetylcholine Receptor Agonists for the Treatment of Alzheimer's Dementia: An Update. Nicotine Tob Res 2019; 21:370-376. [PMID: 30137524 DOI: 10.1093/ntr/nty116] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 08/18/2018] [Indexed: 01/08/2023]
Abstract
A significant portion of the clinical phenotype observed in Alzheimer's disease (AD) occurs through nicotinic acetylcholine receptors (nAChRs). Degeneration of cholinergic neurons, combined with aberrant nAChR expression and activation partially through amyloid-beta peptide (Aβ)-nAChR leads to upregulation of pro-inflammatory pathways and subsequently the progressive cognitive decline of AD. Interestingly, the cholinergic anti-inflammatory pathway is also mediated through nAChR particularly α7 nAChR. Thus, agonists of these receptors will likely exert pro-cognitive benefits through multiple mechanisms including stimulating the cholinergic pathway, modulating inflammation, and buffering the effects of amyloid. Despite this promising theoretical use, trials thus far have been complicated by adverse effects or minimal improvement. This review will provide an update on several pharmacological nAChR agonists tested in clinical trials and reasons that further investigation of nAChR agonists is merited. IMPLICATIONS nAChRs have consistently presented a promising theoretical use in the treatment of AD; however, trials thus far have been complicated by adverse effects or minimal improvement. This review will provide an update on several pharmacological nAChR agonists trialed and reasons that further investigation of nAChR agonists is merited.
Collapse
Affiliation(s)
| | | | - Marwan Noel Sabbagh
- Barrow Neurological Institute, Phoenix, AZ.,Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV
| |
Collapse
|
109
|
The Interplay between Ca 2+ Signaling Pathways and Neurodegeneration. Int J Mol Sci 2019; 20:ijms20236004. [PMID: 31795242 PMCID: PMC6928941 DOI: 10.3390/ijms20236004] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
Calcium (Ca2+) homeostasis is essential for cell maintenance since this ion participates in many physiological processes. For example, the spatial and temporal organization of Ca2+ signaling in the central nervous system is fundamental for neurotransmission, where local changes in cytosolic Ca2+ concentration are needed to transmit information from neuron to neuron, between neurons and glia, and even regulating local blood flow according to the required activity. However, under pathological conditions, Ca2+ homeostasis is altered, with increased cytoplasmic Ca2+ concentrations leading to the activation of proteases, lipases, and nucleases. This review aimed to highlight the role of Ca2+ signaling in neurodegenerative disease-related apoptosis, where the regulation of intracellular Ca2+ homeostasis depends on coordinated interactions between the endoplasmic reticulum, mitochondria, and lysosomes, as well as specific transport mechanisms. In neurodegenerative diseases, alterations-increased oxidative stress, energy metabolism alterations, and protein aggregation have been identified. The aggregation of α-synuclein, β-amyloid peptide (Aβ), and huntingtin all adversely affect Ca2+ homeostasis. Due to the mounting evidence for the relevance of Ca2+ signaling in neuroprotection, we would focus on the expression and function of Ca2+ signaling-related proteins, in terms of the effects on autophagy regulation and the onset and progression of neurodegenerative diseases.
Collapse
|
110
|
Hahm ET, Nagaraja RY, Waro G, Tsunoda S. Cholinergic Homeostatic Synaptic Plasticity Drives the Progression of Aβ-Induced Changes in Neural Activity. Cell Rep 2019; 24:342-354. [PMID: 29996096 DOI: 10.1016/j.celrep.2018.06.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/03/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
Homeostatic synaptic plasticity (HSP) is the ability of neurons to exert compensatory changes in response to altered neural activity. How pathologically induced activity changes are intertwined with HSP mechanisms is unclear. We show that, in cholinergic neurons from Drosophila, beta-amyloid (Aβ) peptides Aβ40 and Aβ42 both induce an increase in spontaneous activity. In a transgenic line expressing Aβ42, we observe that this early increase in spontaneous activity is followed by a dramatic reduction in spontaneous events, a progression that has been suggested to occur in cholinergic brain regions of mammalian models of Alzheimer's disease. We present evidence that the early enhancement in synaptic activity is mediated by the Drosophila α7 nicotinic acetylcholine receptor (nAChR) and that, later, Aβ42-induced inhibition of synaptic events is a consequence of Dα7-dependent HSP mechanisms induced by earlier hyperactivity. Thus, while HSP may initially be an adaptive response, it may also drive maladaptive changes and downstream pathologies.
Collapse
Affiliation(s)
- Eu-Teum Hahm
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Raghavendra Y Nagaraja
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Girma Waro
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, CO 80523, USA.
| |
Collapse
|
111
|
Godoy PA, Ramírez-Molina O, Fuentealba J. Exploring the Role of P2X Receptors in Alzheimer's Disease. Front Pharmacol 2019; 10:1330. [PMID: 31787900 PMCID: PMC6854869 DOI: 10.3389/fphar.2019.01330] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/18/2019] [Indexed: 01/01/2023] Open
Abstract
Several studies have pointed to soluble oligomers of beta amyloid peptide (SOAβ) as the principal neurotoxic agents responsible for the generation of synaptotoxic events that can explain the main symptoms of Alzheimer’s disease (AD). Among the toxic features associated with SOAβ, one of the most notorious is the formation of a non-selective pore-like structure in the plasma membrane, which may partly explain the overload of intracellular Ca2+. There is evidence that the pore causes leakage of key intracellular compounds, such as adenosine triphosphate (ATP), to the extracellular milieu. Extracellular ATP activates P2X receptors (P2XR), which are ligand-gated ion channels (LGICs) widely expressed in both neuron and glial cells and act as neuromodulators of synaptic activity by promoting Ca2+ entry and facilitating neurotransmitter release. There is abundant evidence correlating the overexpression of these receptors to neurodegenerative diseases, including AD, thus opening the possibility that P2XR could potentiate the toxic mechanisms induced by SOAβ and contribute to intracellular Ca2+ overload in neurons and other mechanisms related to glial activation and inflammation. In this review, we correlate scientific evidence related to the main toxic effects induced by SOAβ and those that are mediated by purinergic P2XR. The data suggest that these purinergic receptors participate in the deleterious cellular and molecular effects of SOAβ that lead to the pathogenesis of AD. This information sheds light on the participation of new components in SOAβ toxicity that could be interesting as pharmacological targets for the development of molecular or chemical compounds able to modulate them.
Collapse
Affiliation(s)
- Pamela Andrea Godoy
- Neuroactive Compounds Screening Laboratory,Departamento de Fisiología, Facultad de Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - Oscar Ramírez-Molina
- Neuroactive Compounds Screening Laboratory,Departamento de Fisiología, Facultad de Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Neuroactive Compounds Screening Laboratory,Departamento de Fisiología, Facultad de Cs. Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
112
|
Sakimoto Y, Kida H, Mitsushima D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons. FASEB J 2019; 33:14382-14393. [PMID: 31689120 PMCID: PMC6894079 DOI: 10.1096/fj.201801893rrr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although contextual learning requires plasticity at both excitatory and inhibitory (E/I) synapses in cornu ammonis 1 (CA1) neurons, the temporal dynamics across the neuronal population are poorly understood. Using an inhibitory avoidance task, we analyzed the dynamic changes in learning-induced E/I synaptic plasticity. The training strengthened GABAA receptor–mediated synapses within 1 min, peaked at 10 min, and lasted for over 60 min. The intracellular loop (Ser408−409) of GABAA receptor β3 subunit was also phosphorylated within 1 min of training. As the results of strengthening of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor–mediated synapses, CA1 pyramidal neurons exhibited broad diversity of E/I synaptic currents within 5 min. Moreover, presynaptic glutamate release probability at basal dendrites also increased within 5 min. To further quantify the diversified E/I synaptic currents, we calculated self-entropy (bit) for individual neurons. The neurons showed individual levels of the parameter, which rapidly increased within 1 min of training and maintained for over 60 min. These results suggest that learning-induced synaptic plasticity is critical immediately following encoding rather than during the retrieval phase of the learning. Understanding the temporal dynamics along with the quantification of synaptic diversity would be necessary to identify a failure point for learning-promoted plasticity in cognitive disorders.—Sakimoto, Y., Kida, H., Mitsushima, D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Yuya Sakimoto
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Hiroyuki Kida
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Dai Mitsushima
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
113
|
Zeng SL, Sudlow LC, Berezin MY. Using Xenopus oocytes in neurological disease drug discovery. Expert Opin Drug Discov 2019; 15:39-52. [PMID: 31674217 DOI: 10.1080/17460441.2020.1682993] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Introduction: Neurological diseases present a difficult challenge in drug discovery. Many of the current treatments have limited efficiency or result in a variety of debilitating side effects. The search of new therapies is of a paramount importance, since the number of patients that require a better treatment is growing rapidly. As an in vitro model, Xenopus oocytes provide the drug developer with many distinct advantages, including size, durability, and efficiency in exogenous protein expression. However, there is an increasing need to refine the recent breakthroughs.Areas covered: This review covers the usage and recent advancements of Xenopus oocytes for drug discovery in neurological diseases from expression and functional measurement techniques to current applications in Alzheimer's disease, painful neuropathies, and amyotrophic lateral sclerosis (ALS). The existing limitations of Xenopus oocytes in drug discovery are also discussed.Expert opinion: With the rise of aging population and neurological disorders, Xenopus oocytes, will continue to play an important role in understanding the mechanism of the disease, identification and validation of novel molecular targets, and drug screening, providing high-quality data despite the technical limitations. With further advances in oocytes-related techniques toward an accurate modeling of the disease, the diagnostics and treatment of neuropathologies will be becoming increasing personalized.
Collapse
Affiliation(s)
- Steven L Zeng
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Leland C Sudlow
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mikhail Y Berezin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
114
|
Sun JL, Stokoe SA, Roberts JP, Sathler MF, Nip KA, Shou J, Ko K, Tsunoda S, Kim S. Co-activation of selective nicotinic acetylcholine receptors is required to reverse beta amyloid-induced Ca 2+ hyperexcitation. Neurobiol Aging 2019; 84:166-177. [PMID: 31629115 DOI: 10.1016/j.neurobiolaging.2019.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/20/2022]
Abstract
Beta-amyloid (Aβ) peptide accumulation has long been implicated in the pathogenesis of Alzheimer's disease (AD). Hippocampal network hyperexcitability in the early stages of the disease leads to increased epileptiform activity and eventually cognitive decline. We found that acute application of 250 nM soluble Aβ42 oligomers increased Ca2+ activity in hippocampal neurons in parallel with a significant decrease in activity in Aβ42-treated interneurons. A potential target of Aβ42 is the nicotinic acetylcholine receptor (nAChR). Three major subtypes of nAChRs (α7, α4β2, and α3β4) have been reported in the human hippocampus. Simultaneous inhibition of both α7 and α4β2 nAChRs mimicked the Aβ42 effects on both excitatory and inhibitory neurons. However, inhibition of all 3 subtypes showed the opposite effect. Importantly, simultaneous activation of α7 and α4β2 nAChRs was required to reverse Aβ42-induced neuronal hyperexcitation. We suggest co-activation of α7 and α4β2 nAChRs is required to reverse Aβ42-induced Ca2+ hyperexcitation.
Collapse
Affiliation(s)
- Julianna L Sun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Sarah A Stokoe
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Jessica P Roberts
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Matheus F Sathler
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kaila A Nip
- Cellular and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA
| | - Jiayi Shou
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kaitlyn Ko
- Poudre High School, Fort Collins, CO, USA
| | - Susan Tsunoda
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA
| | - Seonil Kim
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Molecular, Cellular and Integrative Neurosciences Program, Fort Collins, CO, USA; Cellular and Molecular Biology Graduate Program, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
115
|
Computational Study of Natural Compounds for the Clearance of Amyloid-Βeta: A Potential Therapeutic Management Strategy for Alzheimer's Disease. Molecules 2019; 24:molecules24183233. [PMID: 31491967 PMCID: PMC6767296 DOI: 10.3390/molecules24183233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/19/2019] [Accepted: 08/27/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a widespread dynamic neurodegenerative malady. Its etiology is still not clear. One of the foremost pathological features is the extracellular deposits of Amyloid-beta (Aβ) peptides in senile plaques. The interaction of Aβ and the receptor for advanced glycation end products at the blood-brain barrier is also observed in AD, which not only causes the neurovascular anxiety and articulation of proinflammatory cytokines, but also directs reduction of cerebral bloodstream by upgrading the emission of endothelin-1 to induce vasoconstriction. In this process, RAGE is deemed responsible for the influx of Aβ into the brain through BBB. In the current study, we predicted the interaction potential of the natural compounds vincamine, ajmalicine and emetine with the Aβ peptide concerned in the treatment of AD against the standard control, curcumin, to validate the Aβ peptide–compounds results. Protein-protein interaction studies have also been carried out to see their potential to inhibit the binding process of Aβ and RAGE. Moreover, the current study verifies that ligands are more capable inhibitors of a selected target compared to positive control with reference to ΔG values. The inhibition of Aβ and its interaction with RAGE may be valuable in proposing the next round of lead compounds for effective Alzheimer’s disease treatment.
Collapse
|
116
|
Chhatwal JP, Schultz AP, Hedden T, Boot BP, Wigman S, Rentz D, Johnson KA, Sperling RA. Anticholinergic Amnesia is Mediated by Alterations in Human Network Connectivity Architecture. Cereb Cortex 2019; 29:3445-3456. [PMID: 30192928 PMCID: PMC6644870 DOI: 10.1093/cercor/bhy214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/12/2018] [Accepted: 08/08/2018] [Indexed: 11/13/2022] Open
Abstract
Disrupted cholinergic neurotransmission plays a central role in Alzheimer's disease, medication-induced memory impairment, and delirium. At the systems level, this suggests anticholinergic drugs may alter the activity and interplay of anatomically distributed neural networks critical for memory function. Using a network-sensitive imaging technique (functional connectivity MRI) and a double-blind, crossover design, we examined the consequences of anticholinergic drug administration on episodic memory and functional network architecture in a group of clinically normal elderly. We observed that low-dose scopolamine (0.2 mg IV) decreased episodic memory performance and selectively decreased connectivity strength in 3 of 7 cortical networks. Both memory and connectivity effects were independent of β-amyloid burden. Drug-induced connectivity changes within the Default and Salience networks, as well as reductions in the strength of anticorrelation between these 2 networks, were sufficient to fully statistically mediate the effects of scopolamine on memory performance. These results provide experimental support for the importance of the Default and Salience networks to memory performance and suggest scopolamine-induced amnesia is underpinned by disrupted connectivity within and between these 2 networks. More broadly, these results support the potential utility of fcMRI as tool examine the systems-level pharmacology of psychoactive drugs.
Collapse
Affiliation(s)
- Jasmeer P Chhatwal
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Trey Hedden
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, MA, USA
| | - Brendon P Boot
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Voyager Therapeutics, Cambridge, MA, USA
| | - Sarah Wigman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Dorene Rentz
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
- Department of Radiology, Massachusetts General Hospital, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA 02115, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, USA
| |
Collapse
|
117
|
Fabiani C, Antollini SS. Alzheimer's Disease as a Membrane Disorder: Spatial Cross-Talk Among Beta-Amyloid Peptides, Nicotinic Acetylcholine Receptors and Lipid Rafts. Front Cell Neurosci 2019; 13:309. [PMID: 31379503 PMCID: PMC6657435 DOI: 10.3389/fncel.2019.00309] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/25/2019] [Indexed: 12/17/2022] Open
Abstract
Biological membranes show lateral and transverse asymmetric lipid distribution. Cholesterol (Chol) localizes in both hemilayers, but in the external one it is mostly condensed in lipid-ordered microdomains (raft domains), together with saturated phosphatidyl lipids and sphingolipids (including sphingomyelin and glycosphingolipids). Membrane asymmetries induce special membrane biophysical properties and behave as signals for several physiological and/or pathological processes. Alzheimer’s disease (AD) is associated with a perturbation in different membrane properties. Amyloid-β (Aβ) plaques and neurofibrillary tangles of tau protein together with neuroinflammation and neurodegeneration are the most characteristic cellular changes observed in this disease. The extracellular presence of Aβ peptides forming senile plaques, together with soluble oligomeric species of Aβ, are considered the major cause of the synaptic dysfunction of AD. The association between Aβ peptide and membrane lipids has been extensively studied. It has been postulated that Chol content and Chol distribution condition Aβ production and posterior accumulation in membranes and, hence, cell dysfunction. Several lines of evidence suggest that Aβ partitions in the cell membrane accumulate mostly in raft domains, the site where the cleavage of the precursor AβPP by β- and γ- secretase is also thought to occur. The main consequence of the pathogenesis of AD is the disruption of the cholinergic pathways in the cerebral cortex and in the basal forebrain. In parallel, the nicotinic acetylcholine receptor has been extensively linked to membrane properties. Since its transmembrane domain exhibits extensive contacts with the surrounding lipids, the acetylcholine receptor function is conditioned by its lipid microenvironment. The nicotinic acetylcholine receptor is present in high-density clusters in the cell membrane where it localizes mainly in lipid-ordered domains. Perturbations of sphingomyelin or cholesterol composition alter acetylcholine receptor location. Therefore, Aβ processing, Aβ partitioning, and acetylcholine receptor location and function can be manipulated by changes in membrane lipid biophysics. Understanding these mechanisms should provide insights into new therapeutic strategies for prevention and/or treatment of AD. Here, we discuss the implications of lipid-protein interactions at the cell membrane level in AD.
Collapse
Affiliation(s)
- Camila Fabiani
- Instituto de Investigaciones Bioquímicas de Bahía Blanca CONICET-UNS, Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Silvia S Antollini
- Instituto de Investigaciones Bioquímicas de Bahía Blanca CONICET-UNS, Bahía Blanca, Argentina.,Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| |
Collapse
|
118
|
Abstract
Alterations in membrane proteins (MPs) and their regulated pathways have been established as cancer hallmarks and extensively targeted in clinical applications. However, the analysis of MP-interacting proteins and downstream pathways across human malignancies remains challenging. Here, we present a systematically integrated method to generate a resource of cancer membrane protein-regulated networks (CaMPNets), containing 63,746 high-confidence protein-protein interactions (PPIs) for 1962 MPs, using expression profiles from 5922 tumors with overall survival outcomes across 15 human cancers. Comprehensive analysis of CaMPNets links MP partner communities and regulated pathways to provide MP-based gene sets for identifying prognostic biomarkers and druggable targets. For example, we identify CHRNA9 with 12 PPIs (e.g., ERBB2) can be a therapeutic target and find its anti-metastasis agent, bupropion, for treatment in nicotine-induced breast cancer. This resource is a study to systematically integrate MP interactions, genomics, and clinical outcomes for helping illuminate cancer-wide atlas and prognostic landscapes in tumor homo/heterogeneity.
Collapse
|
119
|
Rodríguez-Soacha DA, Scheiner M, Decker M. Multi-target-directed-ligands acting as enzyme inhibitors and receptor ligands. Eur J Med Chem 2019; 180:690-706. [PMID: 31401465 DOI: 10.1016/j.ejmech.2019.07.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
Abstract
In this review, we present the latest advances in the field of multi-target-directed ligand (MTDL) design for the treatment of various complex pathologies of multifactorial origin. In particular, latest findings in the field of MTDL design targeting both an enzyme and a receptor are presented for different diseases such as Alzheimer's disease (AD), depression, addiction, glaucoma, non-alcoholic steatohepatitis and pain and inflammation. The ethology of the diseases is briefly described, with special emphasis on how the MTDL can evolve into novel therapies that replace the classic pharmacological dogma "one target one disease". Considering the current needs for therapy adherence improvement, it is exposed as from the medicinal chemistry, different molecular scaffolds are studied. With the use of structure activity relationship studies and molecular optimization, new hybrid molecules are generated with improved biological properties acting at two biologically very distinct targets.
Collapse
Affiliation(s)
- Diego Alejandro Rodríguez-Soacha
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Matthias Scheiner
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Michael Decker
- Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy and Food Chemistry, Julius Maximilian University of Würzburg, Am Hubland, 97074, Würzburg, Germany.
| |
Collapse
|
120
|
Rothbard JB, Kurnellas MP, Ousman SS, Brownell S, Rothbard JJ, Steinman L. Small Heat Shock Proteins, Amyloid Fibrils, and Nicotine Stimulate a Common Immune Suppressive Pathway with Implications for Future Therapies. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034223. [PMID: 30249602 DOI: 10.1101/cshperspect.a034223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The α7 nicotinic acetylcholine receptor (α7nAChR) is central to the anti-inflammatory function of the vagus nerve in a physiological mechanism termed the inflammatory reflex. Studies on the inflammatory reflex have been instrumental for the current development of the field of bioelectronic medicine. An independent investigation of the biological role of αB-crystallin (HspB5), the most abundant gene transcript present in active multiple sclerosis lesions in human brains, also led to α7nAChR. Induction of experimental autoimmune encephalomyelitis (EAE) in HspB5-/- mice results in greater paralytic signs, increased levels of proinflammatory cytokines, and T-lymphocyte activation relative to wild-type animals. Administration of HspB5 was therapeutic in animal models of multiple sclerosis, retinal and cardiac ischemia, and stroke. Structure-activity studies established that residues 73-92 were as potent as the parent protein, but only when it formed amyloid fibrils. Amyloid fibrils and small heat shock proteins (sHsps) selectively bound α7nAChR on peritoneal macrophages (MΦs) and B lymphocytes, converting the MΦs to an immune suppressive phenotype and mobilizing the migration of both cell types from the peritoneum to secondary lymph organs. Here, we review multiple aspects of this work, which may be of interest for developing future therapeutic approaches for multiple sclerosis and other disorders.
Collapse
Affiliation(s)
- Jonathan B Rothbard
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| | | | - Shalina S Ousman
- Department of Clinical Neurosciences, University of Calgary, Alberta T2N 1N4, Canada
| | - Sara Brownell
- School of Life Sciences, Arizona State University, Tempe, Arizona 85281
| | - Jesse J Rothbard
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| | - Lawrence Steinman
- Department of Neurology, Stanford University School of Medicine, Stanford, California 94305-5316
| |
Collapse
|
121
|
Chen XQ, Mobley WC. Alzheimer Disease Pathogenesis: Insights From Molecular and Cellular Biology Studies of Oligomeric Aβ and Tau Species. Front Neurosci 2019; 13:659. [PMID: 31293377 PMCID: PMC6598402 DOI: 10.3389/fnins.2019.00659] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/07/2019] [Indexed: 01/08/2023] Open
Abstract
Alzheimer disease (AD) represents an oncoming epidemic that without an effective treatment promises to exact extraordinary human and financial burdens. Studies of pathogenesis are essential for defining targets for discovering disease-modifying treatments. Past studies of AD neuropathology provided valuable, albeit limited, insights. Nevertheless, building on these findings, recent studies have provided an increasingly rich harvest of genetic, molecular and cellular data that are creating unprecedented opportunities to both understand and treat AD. Among the most significant are those documenting the presence within the AD brain of toxic oligomeric species of Aβ and tau. Existing data support the view that such species can propagate and spread within neural circuits. To place these findings in context we first review the genetics and neuropathology of AD, including AD in Down syndrome (AD-DS). We detail studies that support the existence of toxic oligomeric species while noting the significant unanswered questions concerning their precise structures, the means by which they spread and undergo amplification and how they induce neuronal dysfunction and degeneration. We conclude by offering a speculative synthesis for how oligomers of Aβ and tau initiate and drive pathogenesis. While 100 years after Alzheimer's first report there is much still to learn about pathogenesis and the discovery of disease-modifying treatments, the application of new concepts and sophisticated new tools are poised to deliver important advances for combatting AD.
Collapse
Affiliation(s)
- Xu-Qiao Chen
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - William C. Mobley
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
122
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
123
|
Cecon E, Dam J, Luka M, Gautier C, Chollet AM, Delagrange P, Danober L, Jockers R. Quantitative assessment of oligomeric amyloid β peptide binding to α7 nicotinic receptor. Br J Pharmacol 2019; 176:3475-3488. [PMID: 30981214 DOI: 10.1111/bph.14688] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Progressive dysfunction of cholinergic transmission is a well-known characteristic of Alzheimer's disease (AD). Amyloid β (Aβ) peptide oligomers are known to play a central role in AD and are suggested to impair the function of the cholinergic nicotinic ACh receptor α7 (α7nAChR). However, the mechanism underlying the effect of Aβ on α7nAChR function is not fully understood, limiting the therapeutic exploration of this observation in AD. Here, we aimed to detect and characterize Aβ binding to α7nAChR, including the possibility of interfering with this interaction for therapeutic purposes. EXPERIMENTAL APPROACH We developed a specific and quantitative time-resolved FRET (TR-FRET)-based binding assay for Aβ to α7nAChR and pharmacologically characterized this interaction. KEY RESULTS We demonstrated specific and high-affinity (low nanomolar) binding of Aβ to the orthosteric binding site of α7nAChR. Aβ binding was prevented and reversed by the well-characterized orthosteric ligands of α7nAChR (epibatidine, α-bungarotoxin, methylylcaconitine, PNU-282987, S24795, and EVP6124) and by the type II positive allosteric modulator (PAM) PNU-120596 but not by the type I PAM NS1738. CONCLUSIONS AND IMPLICATIONS Our TR-FRET Aβ binding assay demonstrates for the first time the specific binding of Aβ to α7nAChR, which will be a crucial tool for the development, testing, and selection of a novel generation of AD drug candidates targeting Aβ/α7nAChR complexes with high specificity and fewer side effects compared to currently approved α7nAChR drugs. LINKED ARTICLES This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Erika Cecon
- Inserm U1016, Institut Cochin, Dept Endocrinology, Metabolism and Diabetes, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Julie Dam
- Inserm U1016, Institut Cochin, Dept Endocrinology, Metabolism and Diabetes, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marine Luka
- Inserm U1016, Institut Cochin, Dept Endocrinology, Metabolism and Diabetes, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Clément Gautier
- Institut de Recherches SERVIER, Division Therapeutic Innovation in Neuropsychiatry, Croissy-sur-Seine, France
| | - Anne-Marie Chollet
- Institut de Recherches SERVIER, Division Therapeutic Innovation in Neuropsychiatry, Croissy-sur-Seine, France
| | - Philippe Delagrange
- Institut de Recherches SERVIER, Division Therapeutic Innovation in Neuropsychiatry, Croissy-sur-Seine, France
| | - Laurence Danober
- Institut de Recherches SERVIER, Division Therapeutic Innovation in Neuropsychiatry, Croissy-sur-Seine, France
| | - Ralf Jockers
- Inserm U1016, Institut Cochin, Dept Endocrinology, Metabolism and Diabetes, Paris, France.,CNRS UMR 8104, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
124
|
Lykhmus O, Koval L, Voytenko L, Uspenska K, Komisarenko S, Deryabina O, Shuvalova N, Kordium V, Ustymenko A, Kyryk V, Skok M. Intravenously Injected Mesenchymal Stem Cells Penetrate the Brain and Treat Inflammation-Induced Brain Damage and Memory Impairment in Mice. Front Pharmacol 2019; 10:355. [PMID: 31057400 PMCID: PMC6479176 DOI: 10.3389/fphar.2019.00355] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/21/2019] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is regarded as one of the pathogenic factors of Alzheimer disease (AD). Previously, we showed that mice regularly injected with bacterial lipopolysaccharide (LPS) possessed the AD-like symptoms like episodic memory decline, elevated amounts of amyloid beta (Aβ) peptide (1-42), and decreased levels of nicotinic acetylcholine receptors (nAChRs) in the brain. The use of mesenchymal stem cells (MSCs), which can differentiate into multiple cell types, including neurons, is an attractive idea of regenerative medicine, in particular, for neurodegenerative disorders like AD. In the present study, we aimed to investigate whether pathogenic effect of LPS on the brain and behavior of mice can be prevented or treated by injection of MSCs or MSC-produced soluble factors. Fluorescently-labeled MSCs, injected intravenously, were found in the brain blood vessels of LPS-treated mice. Mice co-injected with LPS and MSCs did not demonstrate episodic memory impairment, Aβ (1-42) accumulation, and nAChR decrease in the brain and brain mitochondria. Their mitochondria released less cytochrome c under the effect of Ca2+ compared to mitochondria of LPS-only-treated mice. Moreover, MSCs could reverse the pathogenic symptoms developed 3 weeks after LPS injection. Cultured MSCs produced IL-6 in response to LPS and MSCs effect in vivo was accompanied by additional stimulation of both micro- and macroglia. Xenogeneic (human) MSCs were almost as efficient as allogeneic (mouse) ones and regular injections of human MSC-conditioned medium also produced positive effect. These data allow suggesting MSCs as a potential therapeutic tool to cure neuroinflammation-related cognitive pathology.
Collapse
Affiliation(s)
- Olena Lykhmus
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| | - Lyudmyla Koval
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| | - Larysa Voytenko
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| | - Kateryna Uspenska
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| | - Serhiy Komisarenko
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| | - Olena Deryabina
- Department of Gene Technologies, State Institute of Genetic and Regenerative Medicine NAMS, Kyiv, Ukraine
| | - Nadia Shuvalova
- Department of Gene Technologies, State Institute of Genetic and Regenerative Medicine NAMS, Kyiv, Ukraine
| | - Vitalii Kordium
- Department of Gene Technologies, State Institute of Genetic and Regenerative Medicine NAMS, Kyiv, Ukraine.,Department of Cell Regulatory Mechanisms, Institute of Molecular Biology and Genetics NAS, Kyiv, Ukraine
| | - Alina Ustymenko
- Department of Gene Technologies, State Institute of Genetic and Regenerative Medicine NAMS, Kyiv, Ukraine
| | - Vitalii Kyryk
- Department of Gene Technologies, State Institute of Genetic and Regenerative Medicine NAMS, Kyiv, Ukraine
| | - Maryna Skok
- Laboratory of Cell Receptors Immunology, Palladin Institute of Biochemistry NAS, Kyiv, Ukraine
| |
Collapse
|
125
|
Li X, Cao C, Wei P, Xu M, Liu Z, Liu L, Zhong Y, Li R, Zhou Y, Yi T. Self-Assembly of Amphiphilic Peptides for Recognizing High Furin-Expressing Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2019; 11:12327-12334. [PMID: 30864434 DOI: 10.1021/acsami.9b01281] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Self-assembled nanostructures of amphiphilic peptides have a wide range of applications in bioimaging and delivery systems. In this study, we design and synthesize a biocompatible amphiphilic peptide (C-3) consisting of an RVRRFFF sequence and a nitrobenzoxadiazole fluorophore that can self-assemble into stable micelles for specifically detecting furin, a kind of proprotein convertase with promoting tumor progression. The self-assembly of C-3 with a β-sheet nanostructure is capable of a rapid and specific response to furin in only 5 min in aqueous solution because of the existence of the RVRR motif in the C-3 molecule. The C-3 nanostructures thus can selectively distinguish high furin-expressing cancer cells, like MDA-MB-231 cells, a kind of human breast cancer cells, from normal cells. Furthermore, the C-3 self-assembly can stay in living cells for a long time and are capable of durable detection of intracellular furin, being good for tracer analysis. To our knowledge, this is the first example of self-assembly of a soluble amphiphilic peptide that can selectively detect furin in high furin-expressing live cells.
Collapse
Affiliation(s)
- Xiang Li
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Chunyan Cao
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Peng Wei
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Mengyin Xu
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Zhongkuan Liu
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Lingyan Liu
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Yaping Zhong
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Ruohan Li
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| | - Yifeng Zhou
- School of Chemical and Environmental Engineering , Shanghai Institute of Technology , 100 Haiquan Road , Shanghai 201418 , P. R. China
| | - Tao Yi
- Department of Chemistry , Fudan University , 2005 Songhu Road , Shanghai 200438 , P. R. China
| |
Collapse
|
126
|
Kong D, Jin R, Zhao X, Li H, Yan X, Liu F, Sun P, Gao Y, Liang X, Lin Y, Lu G. Protein-Inorganic Hybrid Nanoflower-Rooted Agarose Hydrogel Platform for Point-of-Care Detection of Acetylcholine. ACS APPLIED MATERIALS & INTERFACES 2019; 11:11857-11864. [PMID: 30830739 DOI: 10.1021/acsami.8b21571] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Rapid and precise profiling of acetylcholine (ACh) has become important for diagnosing diseases and safeguarding health care because of its pivotal role in the central nervous system. Herein, we developed a new colorimetric sensor based on protein-inorganic hybrid nanoflowers as artificial peroxidase, comprising a test kit and a smartphone reader, which sensitively quantifies ACh in human serum. In this sensor, ACh indirectly triggered the substrate reaction with the help of a multienzyme system including acetylcholinesterase, choline oxidase, and mimic peroxidase (nanoflowers), accompanying the enhancement of absorbance intensity at 652 nm. Therefore, the multienzyme platform can be used to detect ACh via monitoring the change of the absorbance in a range from 0.0005 to 6.0 mmol L-1. It is worth mentioning that the platform was used to prepare a portable agarose gel-based kit for rapid qualitative monitoring of ACh. Coupling with ImageJ program, the image information of test kits can be transduced into the hue parameter, which provides a directly quantitative tool to identify ACh. Based on the advantages of simple operation, good selectivity, and low cost, the availability of a portable kit for point-of-care testing will achieve the needs of frequent screening and diagnostic tracking.
Collapse
Affiliation(s)
- Deshuai Kong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Rui Jin
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Xu Zhao
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Hongxia Li
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Xu Yan
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Fangmeng Liu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Peng Sun
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Yuan Gao
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Xishuang Liang
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| | - Yuehe Lin
- School of Mechanical and Materials Engineering , Washington State University , Pullman , Washington 99164 , United States
| | - Geyu Lu
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering , Jilin University , Changchun 130012 , People's Republic of China
| |
Collapse
|
127
|
Hashimoto S, Saido TC. Critical review: involvement of endoplasmic reticulum stress in the aetiology of Alzheimer's disease. Open Biol 2019; 8:rsob.180024. [PMID: 29695619 PMCID: PMC5936719 DOI: 10.1098/rsob.180024] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/03/2018] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) stress response is regarded as an important process in the aetiology of Alzheimer's disease (AD). The accumulation of pathogenic misfolded proteins and the disruption of intracellular calcium (Ca2+) signalling are considered to be fundamental mechanisms that underlie the induction of ER stress, leading to neuronal cell death. Indeed, a number of studies have proposed molecular mechanisms linking ER stress to AD pathogenesis based on results from in vitro systems and AD mouse models. However, stress responsivity was largely different between each mouse model, even though all of these models display AD-related pathologies. While several reports have shown elevated ER stress responses in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (Tg) AD mouse models, we and other groups, in contrast, observed no such ER stress response in APP-single-Tg or App-knockin mice. Therefore, it is debatable whether the ER stress observed in APP and PS1 double-Tg mice is due to AD pathology. From these findings, the roles of ER stress in AD pathogenesis needs to be carefully addressed in future studies. In this review, we summarize research detailing the relationship between ER stress and AD, and analyse the results in detail.
Collapse
Affiliation(s)
- Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
128
|
Dynamic effect of beta-amyloid 42 on cell mechanics. J Biomech 2019; 86:79-88. [DOI: 10.1016/j.jbiomech.2019.01.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 01/06/2023]
|
129
|
Volloch V, Rits-Volloch S. News from Mars: Two-Tier Paradox, Intracellular PCR, Chimeric Junction Shift, Dark Matter mRNA and Other Remarkable Features of Mammalian RNA-Dependent mRNA Amplification. Implications for Alzheimer's Disease, RNA-Based Vaccines and mRNA Therapeutics. ACTA ACUST UNITED AC 2019; 2:131-173. [PMID: 33942036 DOI: 10.33597/aimm.02-1009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular Biology, a branch of science established to examine the flow of information from "letters" encrypted into DNA structure to functional proteins, was initially defined by a concept of DNA-to-RNA-to-Protein information movement, a notion termed the Central Dogma of Molecular Biology. RNA-dependent mRNA amplification, a novel mode of eukaryotic protein-encoding RNA-to-RNA-to-Protein genomic information transfer, constitutes the extension of the Central Dogma in the context of mammalian cells. It was shown to occur in cellular circumstances requiring exceptionally high levels of production of specific polypeptides, e.g. globin chains during erythroid differentiation or defined secreted proteins in the context of extracellular matrix deposition. Its potency is reflected in the observed cellular levels of the resulting amplified mRNA product: At the peak of the erythroid differentiation, for example, the amount of globin mRNA produced in the amplification pathway is about 1500-fold higher than the amount of its conventionally generated counterpart in the same cells. The cellular enzymatic machinery at the core of this process, RNA-dependent RNA polymerase activity (RdRp), albeit in a non-conventional form, was shown to be constitutively and ubiquitously present, and RNA-dependent RNA synthesis (RdRs) appeared to regularly occur, in mammalian cells. Under most circumstances, the mammalian RdRp activity produces only short antisense RNA transcripts. Generation of complete antisense RNA transcripts and amplification of mRNA molecules require the activation of inducible components of the mammalian RdRp complex. The mechanism of such activation is not clear. The present article suggests that it is triggered by a variety of cellular stresses and occurs in the context of stress responses in general and within the framework of the integrated stress response (ISR) in particular. In this process, various cellular stresses activate, in a stress type-specific manner, defined members of the mammalian translation initiation factor 2α, eIF2α, kinase family: PKR, GCN2, PERK and HRI. Any of these kinases, in an activated form, phosphorylates eIF2α. This results in suppression of global cellular protein synthesis but also in activation of expression of select group of transcription factors including ATF4, ATF5 and CHOP. These transcription factors either function as inducible components of the RdRp complex or enable their expression. The assembly of the competent RdRp complex activates mammalian RNA-dependent mRNA amplification, which appears to be a two-tier process. Tier One is a "chimeric" pathway, named so because it results in an amplified chimeric mRNA molecule containing a fragment of the antisense RNA strand at its 5' terminus. Tier Two further amplifies one of the two RNA end products of the chimeric pathway and constitutes the physiologically occurring intracellular polymerase chain reaction, iPCR. Depending on the structure of the initial mRNA amplification progenitor, the chimeric pathway, Tier One, may result in multiple outcomes including chimeric mRNA that produces either a polypeptide identical to the original, conventional mRNA progenitor-encoded protein or only its C-terminal fragment, CTF. The chimeric RNA end product of Tier One may also produce a polypeptide that is non-contiguously encoded in the genome, activate translation from an open reading frame, which is "silent" in a conventionally transcribed mRNA, or initiate an abortive translation. In sharp contrast, regardless of the outcome of Tier One, the mRNA end product of Tier Two of mammalian mRNA amplification, the iPCR pathway, always produces a polypeptide identical to a conventional mRNA progenitor-encoded protein. This discordance is referred to as the Two-Tier Paradox and discussed in detail in the present article. On the other hand, both Tiers are similar in that they result in heavily modified mRNA molecules resistant to reverse transcription, undetectable by reverse transcription-based methods of sequencing and therefore constituting a proverbial "Dark Matter" mRNA, despite being highly ubiquitous. It appears that in addition to their other functions, the modifications of the amplified mRNA render it compatible, unlike the bulk of cellular mRNA, with phosphorylated eIF2α in translation, implying that in addition to being extraordinarily abundant due to the method of its generation, amplified mRNA is also preferentially translated under the ISR conditions, thus augmenting the efficiency of the amplification process. The vital importance of powerful mechanisms of amplification of protein-encoding genomic information in normal physiology is self-evident. Their malfunctions or misuse appear to be associated with two types of abnormalities, the deficiency of a protein normally produced by these mechanisms and the mRNA amplification-mediated overproduction of a protein normally not generated by such a process. Certain classes of beta-thalassemia exemplify the first type, whereas the second type is represented by overproduction of beta-amyloid in Alzheimer's disease. Moreover, the proposed mechanism of Alzheimer's disease allows a crucial and verifiable prediction, namely that the disease-causing intraneuronally retained variant of beta-amyloid differs from that produced conventionally by βAPP proteolysis in that it contains the additional methionine or acetylated methionine at its N-terminus. Because of its extraordinary evidential value as a natural reporter of the mRNA amplification pathway, this feature, if proven, would, arguably, constitute the proverbial Holy Grail not only for Alzheimer's disease but also for the mammalian RNA-dependent mRNA amplification field in general. Both examples are discussed in detail in the present article, which summarizes and systematizes our current understanding of the field and describes two categories of reporter constructs, one for the chimeric Tier of mRNA amplification, another for the iPCR pathway; both reporter types are essential for elucidating underlying molecular mechanisms. It also suggests, in light of the recently demonstrated feasibility of RNA-based vaccines, that the targeted intracellular amplification of exogenously introduced amplification-eligible antigen-encoding mRNAs via the induced or naturally occurring RNA-dependent mRNA amplification pathway could be of substantial benefit in triggering a fast and potent immune response and instrumental in the development of future vaccines. Similar approaches can also be effective in achieving efficient and sustained expression of exogenous mRNA in mRNA therapeutics.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children's Hospital, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, USA
| |
Collapse
|
130
|
Volloch V, Olsen B, Rits S. Alzheimer's Disease Prevention and Treatment: Case for Optimism. ACTA ACUST UNITED AC 2019; 2:115-130. [PMID: 33043322 DOI: 10.33597/aimm.02-1008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
A paradigm shift is under way in the Alzheimer's field. A view of Alzheimer's disease, AD, prevailing until now, the old paradigm, maintains that it is initiated and driven by the overproduction and extracellular accumulation of beta-amyloid, Aβ; a peptide assumed to be derived, both in health and disease, solely by proteolysis of its large precursor, βAPP. In AD, according to this view, Aβ overproduction-associated neurodegeneration begins early, accumulates throughout the lifespan, and manifests symptomatically late in life. A number of drugs, designed within the framework of exceptionality of the βAPP proteolytic/secretory pathway in Aβ production in Alzheimer's disease, achieved spectacular successes in treatment, even the reversal, of AD symptoms in animal models. Without exception, they all exhibited equally spectacular failures in human clinical trials. This paradigm provides few causes for optimism with regard to prevention and treatment of AD. In its context, the disease is considered untreatable in the symptomatic phase; even prodromal cases are assumed too advanced for treatment because Aβ-triggered damages have been accumulating for preceding decades, presumably starting in the early twenties and, to be effective, this is when therapeutic intervention should commence and continue for life. The new paradigm does not dispute the seminal role of Aβ in AD but posits that beta-amyloid produced in the βAPP proteolytic/secretory pathway causes AD in humans no more than it does in non-human mammals that share this pathway with humans, accumulate Aβ as they age, but do not develop the disease. Alzheimer's disease, according to this outlook, is driven by the AD-specific pathway of Aβ production, independent of βAPP and absent in animals. Its activation, late in life, occurs through accumulation, via both cellular uptake of secreted Aβ and neuronal retention of a fraction of beta-amyloid produced in the βAPP proteolytic pathway, of intraneuronal Aβ, which triggers mitochondrial dysfunction. Cellular stresses associated with mitochondrial dysfunction, or, probably, the integrated stress response, ISR, elicited by it, activate an AD-specific Aβ production pathway. In it, every conventionally produced βAPP mRNA molecule potentially serves repeatedly as a template for production of severely 5'-truncated mRNA encoding C99 fragment of βAPP, the immediate precursor of Aβ that is processed in a non-secretory pathway, apparently in a neuron-specific manner. The resulting intraneuronally retained Aβ augments mitochondrial dysfunction, which, in turn, sustains the activity of the βAPP mRNA amplification pathway. These self-propagating Aβ overproduction/mitochondrial dysfunction mutual feedback cycles constitute the engine that drives AD and ultimately triggers neuronal death. In this paradigm, preventive treatment can be initiated any time prior to commencement of βAPP mRNA amplification. Moreover, there are good reasons to believe that with a drug blocking the amplification pathway, it would be possible not only to preempt the disease but also stop and reverse it even when early AD symptoms are already manifested. Thus, the new paradigm introduces a novel theory of Alzheimer's disease. It explains the observed discordances, determines defined therapeutic targets, provides blueprints for a new generation of conceptually distinct AD models and specifies design of a reporter for the mRNA amplification pathway. Most importantly, it offers detailed guidance and tangible hope for prevention of the disease and its treatment at the early symptomatic stages.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, USA
| | - Bjorn Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, USA
| | - Sophia Rits
- Division of Molecular Medicine, Children's Hospital, Boston, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, USA
| |
Collapse
|
131
|
Lasala M, Fabiani C, Corradi J, Antollini S, Bouzat C. Molecular Modulation of Human α7 Nicotinic Receptor by Amyloid-β Peptides. Front Cell Neurosci 2019; 13:37. [PMID: 30800059 PMCID: PMC6376857 DOI: 10.3389/fncel.2019.00037] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/23/2019] [Indexed: 12/21/2022] Open
Abstract
Amyloid β peptide (Aβ) is a key player in the development of Alzheimer's disease (AD). It is the primary component of senile plaques in AD patients and is also found in soluble forms. Cholinergic activity mediated by α7 nicotinic receptors has been shown to be affected by Aβ soluble forms. To shed light into the molecular mechanism of this effect, we explored the direct actions of oligomeric Aβ1-40 and Aβ1-42 on human α7 by fluorescence spectroscopy and single-channel recordings. Fluorescence measurements using the conformational sensitive probe crystal violet (CrV) revealed that in the presence of Aβ α7 undergoes concentration-dependent conformational changes. Exposure of α7 to 100 pM Aβ changes CrV KD towards that of the desensitized state. However, α7 is still reactive to high carbamylcholine (Carb) concentrations. These observations are compatible with the induction of active/desensitized states as well as of a novel conformational state in the presence of both Aβ and Carb. At 100 nM Aβ, α7 adopts a resting-state-like structure which does not respond to Carb, suggesting stabilization of α7 in a blocked state. In real time, we found that Aβ is capable of eliciting α7 channel activity either in the absence or presence of the positive allosteric modulator (PAM) PNU-120596. Activation by Aβ is favored at picomolar or low nanomolar concentrations and is not detected at micromolar concentrations. At high Aβ concentrations, the mean duration of activation episodes elicited by ACh in the presence of PNU-120596 is significantly reduced, an effect compatible with slow open-channel block. We conclude that Aβ directly affects α7 function by acting as an agonist and a negative modulator. Whereas the capability of low concentrations of Aβ to activate α7 could be beneficial, the reduced α7 activity in the presence of higher Aβ concentrations or its long exposure may contribute to the cholinergic signaling deficit and may be involved in the initiation and development of AD.
Collapse
Affiliation(s)
- Matías Lasala
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Camila Fabiani
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Jeremías Corradi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Silvia Antollini
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| | - Cecilia Bouzat
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina
| |
Collapse
|
132
|
Liu J, Chang L, Song Y, Li H, Wu Y. The Role of NMDA Receptors in Alzheimer's Disease. Front Neurosci 2019; 13:43. [PMID: 30800052 PMCID: PMC6375899 DOI: 10.3389/fnins.2019.00043] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022] Open
Abstract
In Alzheimer’s disease (AD), early synaptic dysfunction is associated with the increased oligomeric amyloid-beta peptide, which causes NMDAR-dependent synaptic depression and spine elimination. Memantine, low-affinity NMDAR channel blocker, has been used in the treatment of moderate to severe AD. However, clear evidence is still deficient in demonstrating the underlying mechanisms and a relationship between NMDARs dysfunction and AD. This review focuses on not only changes in expression of different NMDAR subunits, but also some unconventional modes of NMDAR action.
Collapse
Affiliation(s)
- Jinping Liu
- School of Medicine, Tsinghua University, Beijing, China
| | - Lirong Chang
- Department of Anatomy, Ministry of Science and Technology Laboratory of Brain Disorders, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yizhi Song
- Department of Anatomy, Ministry of Science and Technology Laboratory of Brain Disorders, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Hui Li
- Department of Anatomy, Ministry of Science and Technology Laboratory of Brain Disorders, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yan Wu
- Department of Anatomy, Ministry of Science and Technology Laboratory of Brain Disorders, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
133
|
Ma KG, Qian YH. Alpha 7 nicotinic acetylcholine receptor and its effects on Alzheimer's disease. Neuropeptides 2019; 73:96-106. [PMID: 30579679 DOI: 10.1016/j.npep.2018.12.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/26/2018] [Accepted: 12/16/2018] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) is one of the major disabling and lethal diseases for aged individuals worldwide. To date, there are more than 10 hypotheses proposed for AD pathology. The beta-amyloid (Aβ) cascade hypothesis is the most widely accepted and proposes that the accumulation of Aβ in the brain is one potential mechanism for AD pathogenesis. Because some Aβ-overloaded patients do not have AD syndrome, this hypothesis is challenged from time to time. More recently, it has been shown that intracellular Aβ plays a key role in AD pathology. Aβ is internalized by receptors distributed on the cell membrane. Among these receptors, the alpha7 nicotinic acetylcholine receptor (α7 nAChR) has been shown to play an important role in AD. The α7 nAChR is a ligand-gated ion channel and is expressed in pivotal brain regions (e.g., the cerebral cortex and hippocampus) responsible for cognitive functions. The α7 nAChR is localized both presynaptically and postsynaptically, where it activates intracellular signaling cascades. Its agonist has been investigated in clinical studies to improve cognitive functions in AD. Although many studies have shown the importance of the α7 nAChR in AD, little is known regarding its role in AD pathology. Therefore, in the current review, we summarized the basic information regarding the structures and functions of the α7 nAChR, the distribution and expression of the α7 nAChR, and the role of the α7 nAChR in mediating Aβ internalization. We subsequently focused on introducing the comprehensive α7 nAChR related signaling pathways and how these signaling pathways are integrated with the α7 nAChR to play a role in AD. Finally, we stressed the AD therapy that targets the α7 nAChR.
Collapse
Affiliation(s)
- Kai-Ge Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China; Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yi-Hua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China; Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an 710061, China.
| |
Collapse
|
134
|
Hassan M, Shahzadi S, Raza H, Abbasi MA, Alashwal H, Zaki N, Moustafa AA, Seo SY. Computational investigation of mechanistic insights of Aβ42 interactions against extracellular domain of nAChRα7 in Alzheimer's disease. Int J Neurosci 2019; 129:666-680. [PMID: 30422726 DOI: 10.1080/00207454.2018.1543670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIM Amyloid beta (Aβ) 1-42, which is a basic constituent of amyloid plaques, binds with extracellular transmembrane receptor nicotine acetylcholine receptor α7 (nAChRα7) in Alzheimer's disease. MATERIALS AND METHODS In the current study, a computational approach was employed to explore the active binding sites of nAChRα7 through Aβ 1-42 interactions and their involvement in the activation of downstream signalling pathways. Sequential and structural analyses were performed on the extracellular part of nAChRα7 to identify its core active binding site. RESULTS Results showed that a conserved residual pattern and well superimposed structures were observed in all nAChRs proteins. Molecular docking servers were used to predict the common interactive residues in nAChRα7 and Aβ1-42 proteins. The docking profile results showed some common interactive residues such as Glu22, Ala42 and Trp171 may consider as the active key player in the activation of downstream signalling pathways. Moreover, the signal communication and receiving efficacy of best-docked complexes was checked through DynOmic online server. Furthermore, the results from molecular dynamic simulation experiment showed the stability of nAChRα7. The generated root mean square deviations and fluctuations (RMSD/F), solvent accessible surface area (SASA) and radius of gyration (Rg) graphs of nAChRα7 also showed its backbone stability and compactness, respectively. CONCLUSION Taken together, our predicted results intimated the structural insight on the molecular interactions of beta amyloid protein involved in the activation of nAChRα7 receptor. In future, a better understanding of nAChRα7 and their interconnected proteins signalling cascade may be consider as target to cure Alzheimer's disease.
Collapse
Affiliation(s)
- Mubashir Hassan
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| | - Saba Shahzadi
- b Institute of Molecular Science and Bioinformatics , Lahore , Pakistan.,c Department of Bioinformatics , Virtual University of Pakistan , Lahore , Pakistan
| | - Hussain Raza
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| | - Muhammad Athar Abbasi
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea.,d Department of Chemistry , Government College University , Lahore , Pakistan
| | - Hany Alashwal
- e Department of Computer Science and Software Engineering, College of Information Technology , United Arab Emirates University , Al-Ain , United Arab Emirates
| | - Nazar Zaki
- e Department of Computer Science and Software Engineering, College of Information Technology , United Arab Emirates University , Al-Ain , United Arab Emirates
| | - Ahmed A Moustafa
- f School of Social Sciences and Psychology.,g MARCS Institute for Brain and Behaviour , Western Sydney University , Sydney , New South Wales , Australia.,h Department of Social Sciences, College of Arts and Sciences , Qatar University , Doha , Qatar'
| | - Sung-Yum Seo
- a Department of Biological Sciences, College of Natural Sciences , Kongju National University , Gongju , Chungnam-do 32588 , Republic of Korea
| |
Collapse
|
135
|
Yao D, Zhao W, Zhang L, Tian Y. A ratiometric electrochemical strategy for sensitive determination of Furin activity based on dual signal amplification and antifouling nanosurfaces. Analyst 2018; 142:4215-4220. [PMID: 29058010 DOI: 10.1039/c7an01295k] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Developing a sensitive and accurate method for Furin activity is still the bottleneck for understanding the role played by Furin in cell-surface systems and even in Alzheimer's disease. In this work, a ratiometric electrochemical biosensor was developed for sensitive and accurate determination of Furin activity in the cell based on dual signal amplification stemming from a peptide with multiple response sites and the antifouling gold nano-bellflowers (GBFs). A new peptide, HS-CMRVRR↓YKDFDFG (P3), was designed for the first time to be selectively cleaved by Furin at site↓. More importantly, this peptide P3 constitutes three amino acid residues with the -COOH group subsequently used to bind with the response molecule of ferrocene, and can remarkably improve the determination sensitivity by about 2.3 fold. Meanwhile, GBFs stabilized by PEG were taken as a second element to magnify the signal of the ferrocene group via a large ratio surface area and good conductivity, as well as an antibiofouling nanosurface to reduce the biofouling of the electrode surface in cells. This double amplification strategy can greatly enhance the sensitivity of Furin detection by 6.5-fold, which is favorable for detection of low amounts of Furin. In addition, 5'-MB-GGCGCGA(T)13-SH-3' was co-assembled as an inner reference to provide a built-in element to correct the determination error resulting from a complicated analysis environment. Finally, this sensitive and accurate Furin biosensor was successfully applied to detect Furin activity in Furin overexpressed U251 and MDA-MB-468 cells. As far as we know, this is the first report to mention an electrochemical strategy to detect Furin activity in cells.
Collapse
Affiliation(s)
- Dazhi Yao
- Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Dong Chuan Road 500, Shanghai 200241, P.R. China.
| | | | | | | |
Collapse
|
136
|
Mulcahy MJ, Paulo JA, Hawrot E. Proteomic Investigation of Murine Neuronal α7-Nicotinic Acetylcholine Receptor Interacting Proteins. J Proteome Res 2018; 17:3959-3975. [PMID: 30285449 DOI: 10.1021/acs.jproteome.8b00618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.
Collapse
Affiliation(s)
- Matthew J Mulcahy
- Division of Biology and Biological Engineering , California Institute of Technology , 1200 East California Boulevard , Pasadena , California 91125-2900 , United States.,Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Edward Hawrot
- Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
137
|
Toglia P, Demuro A, Mak DOD, Ullah G. Data-driven modeling of mitochondrial dysfunction in Alzheimer's disease. Cell Calcium 2018; 76:23-35. [PMID: 30248575 DOI: 10.1016/j.ceca.2018.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 02/06/2023]
Abstract
Intracellular accumulation of oligomeric forms of β amyloid (Aβ) are now believed to play a key role in the earliest phase of Alzheimer's disease (AD) as their rise correlates well with the early symptoms of the disease. Extensive evidence points to impaired neuronal Ca2+ homeostasis as a direct consequence of the intracellular Aβ oligomers. However, little is known about the downstream effects of the resulting Ca2+ rise on the many intracellular Ca2+-dependent pathways. Here we use multiscale modeling in conjunction with patch-clamp electrophysiology of single inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and fluorescence imaging of whole-cell Ca2+ response, induced by exogenously applied intracellular Aβ42 oligomers to show that Aβ42 inflicts cytotoxicity by impairing mitochondrial function. Driven by patch-clamp experiments, we first model the kinetics of IP3R, which is then extended to build a model for the whole-cell Ca2+ signals. The whole-cell model is then fitted to fluorescence signals to quantify the overall Ca2+ release from the endoplasmic reticulum by intracellular Aβ42 oligomers through G-protein-mediated stimulation of IP3 production. The estimated IP3 concentration as a function of intracellular Aβ42 content together with the whole-cell model allows us to show that Aβ42 oligomers impair mitochondrial function through pathological Ca2+ uptake and the resulting reduced mitochondrial inner membrane potential, leading to an overall lower ATP and increased production of reactive oxygen species and H2O2. We further show that mitochondrial function can be restored by the addition of Ca2+ buffer EGTA, in accordance with the observed abrogation of Aβ42 cytotoxicity by EGTA in our live cells experiments.
Collapse
Affiliation(s)
- Patrick Toglia
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Angelo Demuro
- Department of Neurobiology and Behavior and Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Don-On Daniel Mak
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ghanim Ullah
- Department of Physics, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
138
|
Alkadhi KA. Neuroprotective Effects of Nicotine on Hippocampal Long-Term Potentiation in Brain Disorders. J Pharmacol Exp Ther 2018; 366:498-508. [PMID: 29914875 DOI: 10.1124/jpet.118.247841] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 05/23/2018] [Indexed: 08/30/2023] Open
Abstract
Long-term potentiation (LTP) is commonly considered the cellular correlate of learning and memory. In learning and memory impairments, LTP is invariably diminished in the hippocampus, the brain region responsible for memory formation. LTP is measured electrophysiologically in various areas of the hippocampus. Two mechanistically distinct phases of LTP have been identified: early phase LTP, related to short-term memory; and late-phase LTP, related to long-term memory. These two forms can be severely reduced in a variety of conditions but can be rescued by treatment with nicotine. This report reviews the literature on the beneficial effect of nicotine on LTP in conditions that compromise learning and memory.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Professor of Pharmacology, Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
139
|
Galvin VC, Arnsten AFT, Wang M. Evolution in Neuromodulation-The Differential Roles of Acetylcholine in Higher Order Association vs. Primary Visual Cortices. Front Neural Circuits 2018; 12:67. [PMID: 30210306 PMCID: PMC6121028 DOI: 10.3389/fncir.2018.00067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 08/06/2018] [Indexed: 11/29/2022] Open
Abstract
This review contrasts the neuromodulatory influences of acetylcholine (ACh) on the relatively conserved primary visual cortex (V1), compared to the newly evolved dorsolateral prefrontal association cortex (dlPFC). ACh is critical both for proper circuit development and organization, and for optimal functioning of mature systems in both cortical regions. ACh acts through both nicotinic and muscarinic receptors, which show very different expression profiles in V1 vs. dlPFC, and differing effects on neuronal firing. Cholinergic effects mediate attentional influences in V1, enhancing representation of incoming sensory stimuli. In dlPFC ACh plays a permissive role for network communication. ACh receptor expression and ACh actions in higher visual areas have an intermediate profile between V1 and dlPFC. This changing role of ACh modulation across association cortices may help to illuminate the particular susceptibility of PFC in cognitive disorders, and provide therapeutic targets to strengthen cognition.
Collapse
Affiliation(s)
- Veronica C. Galvin
- Department of Neuroscience, Yale University, New Haven, CT, United States
| | | | | |
Collapse
|
140
|
Alkadhi KA. Delayed effects of combined stress and Aβ infusion on L-LTP of the dentate gyrus: Prevention by nicotine. Neurosci Lett 2018; 682:10-15. [PMID: 29883681 DOI: 10.1016/j.neulet.2018.05.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/15/2018] [Accepted: 05/25/2018] [Indexed: 11/16/2022]
Abstract
Alzheimer's Disease (AD) is a progressive dementia hallmarked by the presence in the brain of extracellular beta-amyloid (Aβ) plaques and intraneuronal fibrillary tangles. Chronic stress is associated with heightened Aβ buildup and acceleration of development of AD, however, stress alone has no significant effect on synaptic plasticity in the dentate gyrus (DG) area. Previously, we have reported that the combination of stress and AD causes more severe inhibition of synaptic plasticity of hippocampal area CA1 than chronic stress or AD alone, and that chronic nicotine treatment prevents this impairment. To investigate the effect of stress and nicotine on synaptic plasticity in the relatively injury-resistant DG area, the present experiments analyzed the effect of chronic stress and the neuroprotective effect of nicotine on LTP in the DG area of a rat model of AD. Wistar rats were chronically stressed and treated with nicotine (1 mg/kg/twice daily; s.c.) for six weeks. Then, at weeks 5-6, AD model was generated by 14-day i.c.v osmotic pump infusion of Aβ peptides (300 pmol/day) into the brains of these rats. Field potential recordings from the DG area of anesthetized rats, revealed that while chronic stress did not accentuate Aβ-induced impairments of E-LTP, it markedly augmented Aβ effect on L-LTP that was only seen 100 min after multiple high frequency stimulation. This delayed action is likely to be due to impairment of process of de novo protein synthesis required for maintenance phase of L-LTP. Chronic nicotine treatment prevented stress-enhanced suppression of synaptic plasticity.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
141
|
Huang F, Qu A, Yang H, Zhu L, Zhou H, Liu J, Long J, Shi L. Self-Assembly Molecular Chaperone to Concurrently Inhibit the Production and Aggregation of Amyloid β Peptide Associated with Alzheimer's Disease. ACS Macro Lett 2018; 7:983-989. [PMID: 35650950 DOI: 10.1021/acsmacrolett.8b00495] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Amyloid β peptide (Aβ) plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Currently, decreasing Aβ production and preventing Aβ aggregation are thought to be important strategies in anti-AD therapy. However, inhibiting Aβ production or aggregation in isolation is not sufficient to reverse the neurodegenerative process of AD patients in clinical testing. Here, a self-assembly molecular chaperone (SAMC) consisting of γ-secretase inhibitor DAPT and mixed-shell polymeric micelles is devised, serving as a bifunctional suppressor of AD. This two-in-one combinational system can simultaneously inhibit Aβ production and aggregation, which would contribute to enhancing the therapeutic effect by decreasing Aβ levels. Decorating a neuron-specific RVG29 peptide onto the surface, the DAPT-incorporated SAMC can specifically target neuronal cells and, thus, will relieve the strong side effect of DAPT on normal cells. Therefore, this combination strategy holds great potential to open up an avenue for AD treatment.
Collapse
Affiliation(s)
- Fan Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, People’s Republic of China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, People’s Republic of China
| | - Aoting Qu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, People’s Republic of China
| | - Huiru Yang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, People’s Republic of China
| | - Lin Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, People’s Republic of China
| | - Hao Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, People’s Republic of China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, People’s Republic of China
| | - Jiafu Long
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, People’s Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, People’s Republic of China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, People’s Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, People’s Republic of China
| |
Collapse
|
142
|
Sawamura N, Ju Y, Asahi T. Cholinergic receptor, nicotinic, alpha 7 as a target molecule of Arctic mutant amyloid β. Neural Regen Res 2018; 13:1360-1361. [PMID: 30106044 PMCID: PMC6108216 DOI: 10.4103/1673-5374.235238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Naoya Sawamura
- Faculty of Science and Engineering; Research Organization for Nano & Life Innovation, Waseda University, Tokyo, Japan
| | - Ye Ju
- Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | - Toru Asahi
- Faculty of Science and Engineering; Research Organization for Nano & Life Innovation, Waseda University, Tokyo, Japan
| |
Collapse
|
143
|
Brai E, Simon F, Cogoni A, Greenfield SA. Modulatory Effects of a Novel Cyclized Peptide in Reducing the Expression of Markers Linked to Alzheimer's Disease. Front Neurosci 2018; 12:362. [PMID: 29950969 PMCID: PMC6008575 DOI: 10.3389/fnins.2018.00362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/09/2018] [Indexed: 12/17/2022] Open
Abstract
Despite many studies attempt to identify the primary mechanisms underlying neurodegeneration in Alzheimer's disease (AD), the key events still remain elusive. We have previously shown that a peptide cleaved from the acetylcholinesterase (AChE) C-terminus (T14) can play a pivotal role as a signaling molecule in neurodegeneration, via its interaction with the α7 nicotinic acetylcholine receptor. The main goal of this study is to determine whether a cyclized variant (NBP14) of the toxic AChE-derived peptide can antagonize the effects of its linear counterpart, T14, in modulating well-known markers linked to neurodegeneration. We investigate this hypothesis applying NBP14 on ex-vivo rat brain slices containing the basal forebrain. Western blot analysis revealed an inhibitory action of NBP14 on naturally occurring T14 peptide, as well as on endogenous amyloid beta, whereas the expression of the nicotinic receptor and phosphorylated Tau was relatively unaffected. These results further confirm the neurotoxic properties of the AChE-peptide and show for the first time in an ex-vivo preparation the possible neuroprotective activity of NBP14, over a protracted period of hours, indicating that T14 pathway may offer a new prospect for therapeutic intervention in AD pathobiology.
Collapse
Affiliation(s)
- Emanuele Brai
- Culham Science Centre, Neuro-Bio Ltd., Oxfordshire, United Kingdom
| | - Florian Simon
- Culham Science Centre, Neuro-Bio Ltd., Oxfordshire, United Kingdom.,Department of Biotechnology, University of Nîmes, Nîmes, France
| | - Antonella Cogoni
- Culham Science Centre, Neuro-Bio Ltd., Oxfordshire, United Kingdom
| | | |
Collapse
|
144
|
Cellular Receptors of Amyloid β Oligomers (AβOs) in Alzheimer's Disease. Int J Mol Sci 2018; 19:ijms19071884. [PMID: 29954063 PMCID: PMC6073792 DOI: 10.3390/ijms19071884] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
It is estimated that Alzheimer’s disease (AD) affects tens of millions of people, comprising not only suffering patients, but also their relatives and caregivers. AD is one of age-related neurodegenerative diseases (NDs) characterized by progressive synaptic damage and neuronal loss, which result in gradual cognitive impairment leading to dementia. The cause of AD remains still unresolved, despite being studied for more than a century. The hallmark pathological features of this disease are senile plaques within patients’ brain composed of amyloid beta (Aβ) and neurofibrillary tangles (NFTs) of Tau protein. However, the roles of Aβ and Tau in AD pathology are being questioned and other causes of AD are postulated. One of the most interesting theories proposed is the causative role of amyloid β oligomers (AβOs) aggregation in the pathogenesis of AD. Moreover, binding of AβOs to cell membranes is probably mediated by certain proteins on the neuronal cell surface acting as AβO receptors. The aim of our paper is to describe alternative hypotheses of AD etiology, including genetic alterations and the role of misfolded proteins, especially Aβ oligomers, in Alzheimer’s disease. Furthermore, in this review we present various putative cellular AβO receptors related to toxic activity of oligomers.
Collapse
|
145
|
Identification of a common immune regulatory pathway induced by small heat shock proteins, amyloid fibrils, and nicotine. Proc Natl Acad Sci U S A 2018; 115:7081-7086. [PMID: 29915045 DOI: 10.1073/pnas.1804599115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although certain dogma portrays amyloid fibrils as drivers of neurodegenerative disease and neuroinflammation, we have found, paradoxically, that amyloid fibrils and small heat shock proteins (sHsps) are therapeutic in experimental autoimmune encephalomyelitis (EAE). They reduce clinical paralysis and induce immunosuppressive pathways, diminishing inflammation. A key question was the identification of the target for these molecules. When sHsps and amyloid fibrils were chemically cross-linked to immune cells, a limited number of proteins were precipitated, including the α7 nicotinic acetylcholine receptor (α7 NAChR). The α7 NAChR is noteworthy among the over 20 known receptors for amyloid fibrils, because it plays a central role in a well-defined immune-suppressive pathway. Competitive binding between amyloid fibrils and α-bungarotoxin to peritoneal macrophages (MΦs) confirmed the involvement of α7 NAChR. The mechanism of immune suppression was explored, and, similar to nicotine, amyloid fibrils inhibited LPS induction of a common set of inflammatory cytokines while inducing Stat3 signaling and autophagy. Consistent with this, previous studies have established that nicotine, sHsps, and amyloid fibrils all were effective therapeutics in EAE. Interestingly, B lymphocytes were needed for the therapeutic effect. These results suggest that agonists of α7 NAChR might have therapeutic benefit for a variety of inflammatory diseases.
Collapse
|
146
|
Paulo JA, Gygi SP. Isobaric Tag-Based Protein Profiling of a Nicotine-Treated Alpha7 Nicotinic Receptor-Null Human Haploid Cell Line. Proteomics 2018; 18:e1700475. [PMID: 29663646 PMCID: PMC5990481 DOI: 10.1002/pmic.201700475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/04/2018] [Indexed: 12/16/2022]
Abstract
Nicotinic acetylcholine receptors (nAChR), the primary cell surface targets of nicotine, have implications in various neurological disorders. Here we investigate the proteome-wide effects of nicotine on human haploid cell lines (wildtype HAP1 and α7KO-HAP1) to address differences in nicotine-induced protein abundance profiles between these cell lines. We performed an SPS-MS3-based TMT10-plex experiment arranged in a 2-3-2-3 design with two replicates of the untreated samples and three of the treated samples for each cell line. We quantified 8775 proteins across all ten samples, of which several hundred differed significantly in abundance. Comparing α7KO-HAP1 and HAP1wt cell lines to each other revealed significant protein abundance alterations; however, we also measured differences resulting from nicotine treatment in both cell lines. Among proteins with increased abundance levels due to nicotine treatment included those previously identified: APP, APLP2, and ITM2B. The magnitude of these changes was greater in HAP1wt compared to the α7KO-HAP1 cell line, implying a potential role for the α7 nAChR in HAP1 cells. Moreover, the data revealed that membrane proteins and proteins commonly associated with neurons were predominant among those with altered abundance. This study, which is the first TMT-based proteome profiling of HAP1 cells, defines further the effects of nicotine on non-neuronal cellular proteomes.
Collapse
Affiliation(s)
- Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
147
|
Foroutanpay B, Kumar J, Kang S, Danaei N, Westaway D, Sim V, Kar S. The Effects of N-terminal Mutations on β-amyloid Peptide Aggregation and Toxicity. Neuroscience 2018; 379:177-188. [DOI: 10.1016/j.neuroscience.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 03/01/2018] [Accepted: 03/12/2018] [Indexed: 12/31/2022]
|
148
|
Chen T, Wang Y, Zhang T, Zhang B, Chen L, Zhao L, Chen L. Simvastatin Enhances Activity and Trafficking of α7 Nicotinic Acetylcholine Receptor in Hippocampal Neurons Through PKC and CaMKII Signaling Pathways. Front Pharmacol 2018; 9:362. [PMID: 29706890 PMCID: PMC5906710 DOI: 10.3389/fphar.2018.00362] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/28/2018] [Indexed: 11/13/2022] Open
Abstract
Simvastatin (SV) enhances glutamate release and synaptic plasticity in hippocampal CA1 region upon activation of α7 nicotinic acetylcholine receptor (α7nAChR). In this study, we examined the effects of SV on the functional activity of α7nAChR on CA1 pyramidal cells using patch-clamp recording and explored the underlying mechanisms. We found that the treatment of hippocampal slices with SV for 2 h induced a dose-dependent increase in the amplitude of ACh-evoked inward currents (IACh) and the level of α7nAChR protein on the cell membrane without change in the level of α7nAChR phosphorylation. These SV-induced phenotypes were suppressed by addition of farnesol (FOH) that converts farnesyl pyrophosphate, but not geranylgeraniol. Similarly, the farnesyl transferase inhibitor FTI277 was able to increase the amplitude of IACh and enhance the trafficking of α7nAChR. The treatment with SV enhanced phosphorylation of CaMKII and PKC. The SV-enhanced phosphorylation of CaMKII rather than PKC was blocked by FOH, Src inhibitor PP2 or NMDA receptor antagonist MK801 and mimicked by FTI. The SV-enhanced phosphorylation of PKC was sensitive to the IP3R antagonist 2-APB. The SV-increased amplitude of IACh was suppressed by PKC inhibitor GF109203X and Go6983, or CaMKII inhibitor KN93. The SV- and FTI-enhanced trafficking of α7nAChR was sensitive to KN93, but not GF109203X or Go6983. The PKC activator PMA increased α7nAChR activity, but had no effect on trafficking of α7nAChR. Collectively, these results indicate that acute treatment with SV enhances the activity and trafficking of α7nAChR by increasing PKC phosphorylation and reducing farnesyl-pyrophosphate to trigger NMDA receptor-mediated CaMKII activation.
Collapse
Affiliation(s)
- Tingting Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ya Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Tingting Zhang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Baofeng Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Liandong Zhao
- Department of Neurology, Huaian Second People's Hospital, Huaian, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Physiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
149
|
Willén K, Sroka A, Takahashi RH, Gouras GK. Heterogeneous Association of Alzheimer's Disease-Linked Amyloid-β and Amyloid-β Protein Precursor with Synapses. J Alzheimers Dis 2018; 60:511-524. [PMID: 28869466 PMCID: PMC5611798 DOI: 10.3233/jad-170262] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (AD) is increasingly viewed as a disease of synapses. Loss of synapses correlates better with cognitive decline than amyloid plaques and neurofibrillary tangles, the hallmark neuropathological lesions of AD. Soluble forms of amyloid-β (Aβ) have emerged as mediators of synapse dysfunction. Aβ binds to, accumulates, and aggregates in synapses. However, the anatomical and neurotransmitter specificity of Aβ and the amyloid-β protein precursor (AβPP) in AD remain poorly understood. In addition, the relative roles of Aβ and AβPP in the development of AD, at pre- versus post-synaptic compartments and axons versus dendrites, respectively, remain unclear. Here we use immunogold electron microscopy and confocal microscopy to provide evidence for heterogeneity in the localization of Aβ/AβPP. We demonstrate that Aβ binds to a subset of synapses in cultured neurons, with preferential binding to glutamatergic compared to GABAergic neurons. We also highlight the challenge of defining pre- versus post-synaptic localization of this binding by confocal microscopy. Further, endogenous Aβ42 accumulates in both glutamatergic and GABAergic AβPP/PS1 transgenic primary neurons, but at varying levels. Moreover, upon knock-out of presenilin 1 or inhibition of γ-secretase AβPP C-terminal fragments accumulate both pre- and post-synaptically; however earlier pre-synaptically, consistent with a higher rate of AβPP processing in axons. A better understanding of the synaptic and anatomical selectivity of Aβ/AβPP in AD can be important for the development of more effective new therapies for this major disease of aging.
Collapse
Affiliation(s)
- Katarina Willén
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Agnieszka Sroka
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Gunnar K Gouras
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
150
|
Chang CC, Edwald E, Veatch S, Steel DG, Gafni A. Interactions of amyloid-β peptides on lipid bilayer studied by single molecule imaging and tracking. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1616-1624. [PMID: 29580771 DOI: 10.1016/j.bbamem.2018.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/17/2022]
Abstract
The amyloid-β peptides (Aβ40 and Aβ42) feature prominently in the synaptic dysfunction and neuronal loss associated with Alzheimer's disease (AD). This has been proposed to be due either to interactions between Aβ and cell surface receptors affecting cell signaling, or to the formation of calcium-permeable channels in the membrane that disrupt calcium homeostasis. In both mechanisms the cell membrane is the primary cellular structure with which Aβ interacts. Aβ concentrations in human bodily fluids are very low (pM-nM) rendering studies of the size, composition, cellular binding sites and mechanism of action of the oligomers formed in vivo very challenging. Most studies, therefore, have utilized Aβ oligomers prepared at micromolar peptide concentrations, where Aβ forms oligomeric species which possess easily observable cell toxicity. Such toxicity has not been observed when nM concentrations of peptide are used in the experiment highlighting the importance of employing physiologically relevant peptide concentrations for the results to be of biological significance. In this paper single-molecule microscopy was used to monitor Aβ oligomer formation and diffusion on a supported lipid bilayer at nanomolar peptide concentrations. Aβ monomers, the dominant species in solution, tightly associate with the membrane and are highly mobile whereas trimers and higher-order oligomers are largely immobile. Aβ dimers exist in a mixture of mobile and immobile states. Oligomer growth on the membrane is more rapid for Aβ40 than for the more amyloidogenic Aβ42 but is largely inhibited for a 1:1 Aβ40:Aβ42 mixture. The mechanism underlying these Aβ40-Aβ42 interactions may feature in Alzheimer's pathology.
Collapse
Affiliation(s)
- Chun-Chieh Chang
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elin Edwald
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah Veatch
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Duncan G Steel
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Physics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ari Gafni
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|