1
|
Liu S, Chen L, Li J, Sun Y, Xu Y, Li Z, Zhu Z, Li X. Asiaticoside Mitigates Alzheimer's Disease Pathology by Attenuating Inflammation and Enhancing Synaptic Function. Int J Mol Sci 2023; 24:11976. [PMID: 37569347 PMCID: PMC10418370 DOI: 10.3390/ijms241511976] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, hallmarked by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles. Due to the uncertainty of the pathogenesis of AD, strategies aimed at suppressing neuroinflammation and fostering synaptic repair are eagerly sought. Asiaticoside (AS), a natural triterpenoid derivative derived from Centella asiatica, is known for its anti-inflammatory, antioxidant, and wound-healing properties; however, its neuroprotective function in AD remains unclear. Our current study reveals that AS, when administered (40 mg/kg) in vivo, can mitigate cognitive dysfunction and attenuate neuroinflammation by inhibiting the activation of microglia and proinflammatory factors in Aβ1-42-induced AD mice. Further mechanistic investigation suggests that AS may ameliorate cognitive impairment by inhibiting the activation of the p38 MAPK pathway and promoting synaptic repair. Our findings propose that AS could be a promising candidate for AD treatment, offering neuroinflammation inhibition and enhancement of synaptic function.
Collapse
Affiliation(s)
- Sai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Long Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jinran Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yue Xu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Zhaoxing Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Zheying Zhu
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Xinuo Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
2
|
Young KA, Mancera RL. Review: Investigating the aggregation of amyloid beta with surface plasmon resonance: Do different approaches yield different results? Anal Biochem 2022; 654:114828. [PMID: 35931183 DOI: 10.1016/j.ab.2022.114828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/30/2022]
Abstract
Aggregation of amyloid beta into amyloid plaques in the brain is a hallmark characteristic of Alzheimer's disease. Therapeutics aimed at preventing or retarding amyloid formation often rely on detailed characterization of the underlying mechanism and kinetics of protein aggregation. Surface plasmon resonance (SPR) spectroscopy is a robust technique used to determine binding affinity and kinetics of biomolecular interactions. This approach has been used to characterize the mechanism of aggregation of amyloid beta but there are multiple pitfalls that need to be addressed when working with this and other amyloidogenic proteins. The choice of method for analyte preparation and ligand immobilization to a sensor chip can lead to different theoretical and practical implications in terms of the mathematical modelling of binding data, different mechanisms of binding and the presence of different interacting species. This review examines preparation methods for SPR characterisation of the aggregation of amyloid beta and their influence on the findings derived from such studies.
Collapse
Affiliation(s)
- Kimberly A Young
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia
| | - Ricardo L Mancera
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, GPO Box U1987, Perth, WA, 6845, Australia.
| |
Collapse
|
3
|
Novel Rivastigmine Derivatives as Promising Multi-Target Compounds for Potential Treatment of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10071510. [PMID: 35884815 PMCID: PMC9313321 DOI: 10.3390/biomedicines10071510] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 01/03/2023] Open
Abstract
Alzheimer’s disease (AD) is the most serious and prevalent neurodegenerative disorder still without cure. Since its aetiology is diverse, recent research on anti-AD drugs has been focused on multi-target compounds. In this work, seven novel hybrids (RIV–BIM) conjugating the active moiety of the drug rivastigmine (RIV) with 2 isomeric hydroxyphenylbenzimidazole (BIM) units were developed and studied. While RIV assures the inhibition of cholinesterases, BIM provides further appropriate properties, such as inhibition of amyloid β-peptide (Aβ) aggregation, antioxidation and metal chelation. The evaluated biological properties of these hybrids included antioxidant activity; inhibition of acetylcholinesterase (AChE), butyrylcholinesterase (BChE) and Aβ42 aggregation; as well as promotion of cell viability and neuroprotection. All the compounds are better inhibitors of AChE than rivastigmine (IC50 = 32.1 µM), but compounds of series 5 are better inhibitors of BChE (IC50 = 0.9−1.7 µM) than those of series 4. Series 5 also showed good capacity to inhibit self- (42.1−58.7%) and Cu(II)-induced (40.3−60.8%) Aβ aggregation and also to narrow (22.4−42.6%) amyloid fibrils, the relevant compounds being 5b and 5d. Some of these compounds can also prevent the toxicity induced in SH-SY5Y cells by Aβ42 and oxidative stress. Therefore, RIV–BIM hybrids seem to be potential drug candidates for AD with multi-target abilities.
Collapse
|
4
|
Budvytyte R, Ambrulevičius F, Jankaityte E, Valincius G. Electrochemical Assessment of Dielectric Damage to Phospholipid Bilayers by Amyloid β-Oligomers. Bioelectrochemistry 2022; 145:108091. [DOI: 10.1016/j.bioelechem.2022.108091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/02/2022]
|
5
|
Volatile Anesthetic Sevoflurane Precursor 1,1,1,3,3,3-Hexafluoro-2-Propanol (HFIP) Exerts an Anti-Prion Activity in Prion-Infected Culture Cells. Neurochem Res 2021; 46:2056-2065. [PMID: 34043140 PMCID: PMC8254714 DOI: 10.1007/s11064-021-03344-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/20/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022]
Abstract
Prion disease is a neurodegenerative disorder with progressive neurologic symptoms and accelerated cognitive decline. The causative protein of prion disease is the prion protein (PrP), and structural transition of PrP from the normal helix rich form (PrPC) to the abnormal β-sheet rich form (PrPSc) occurs in prion disease. While so far numerous therapeutic agents for prion diseases have been developed, none of them are still useful. A fluorinated alcohol, hexafluoro isopropanol (HFIP), is a precursor to the inhalational anesthetic sevoflurane and its metabolites. HFIP is also known as a robust α-helix inducer and is widely used as a solvent for highly aggregated peptides. Here we show that the α-helix-inducing activity of HFIP caused the conformational transformation of the fibrous structure of PrP into amorphous aggregates in vitro. HFIP added to the ScN2a cell medium, which continuously expresses PrPSc, reduced PrPSc protease resistance after 24-h incubation. It was also clarified that ScN2a cells are more susceptible to HFIP than any of the cells being compared. Based on these findings, HFIP is expected to develop as a therapeutic agent for prion disease.
Collapse
|
6
|
Cojocaru A, Burada E, Bălșeanu AT, Deftu AF, Cătălin B, Popa-Wagner A, Osiac E. Roles of Microglial Ion Channel in Neurodegenerative Diseases. J Clin Med 2021; 10:jcm10061239. [PMID: 33802786 PMCID: PMC8002406 DOI: 10.3390/jcm10061239] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
As the average age and life expectancy increases, the incidence of both acute and chronic central nervous system (CNS) pathologies will increase. Understanding mechanisms underlying neuroinflammation as the common feature of any neurodegenerative pathology, we can exploit the pharmacology of cell specific ion channels to improve the outcome of many CNS diseases. As the main cellular player of neuroinflammation, microglia play a central role in this process. Although microglia are considered non-excitable cells, they express a variety of ion channels under both physiological and pathological conditions that seem to be involved in a plethora of cellular processes. Here, we discuss the impact of modulating microglia voltage-gated, potential transient receptor, chloride and proton channels on microglial proliferation, migration, and phagocytosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Alexandru Cojocaru
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.C.); (E.B.); (A.-T.B.)
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Emilia Burada
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.C.); (E.B.); (A.-T.B.)
| | - Adrian-Tudor Bălșeanu
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.C.); (E.B.); (A.-T.B.)
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandru-Florian Deftu
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), CH-1011 Lausanne, Switzerland;
- Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| | - Bogdan Cătălin
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (A.C.); (E.B.); (A.-T.B.)
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence: (B.C.); (A.P.-W.)
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology, Dementia and Ageing Research, University Hospital Essen, 45147 Essen, Germany
- Correspondence: (B.C.); (A.P.-W.)
| | - Eugen Osiac
- Department of Biophysics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
7
|
Sharma S, Saini A, Nehru B. Neuroprotective effects of carbenoxolone against amyloid-beta 1-42 oligomer-induced neuroinflammation and cognitive decline in rats. Neurotoxicology 2021; 83:89-105. [PMID: 33412218 DOI: 10.1016/j.neuro.2020.12.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/01/2020] [Accepted: 12/28/2020] [Indexed: 01/01/2023]
Abstract
The aggregation of Aβ plays a major role in the progression of Alzheimer's disease (AD) and induces neuroinflammation, neurodegeneration and cognitive decline. Recent studies have shown that the soluble aggregates of Aβ are the major culprits in the development of these aberrations inside the brain. In this study, we investigated the neuroprotective potential of carbenoxolone (Cbx), which has been found to possess anti-inflammatory and nootropic properties. Male SD rats (250-300 g) were divided into the four groups (n = 8 per group): (1) sham control rats injected with vehicles, (2) Aβ 1-42 group rats injected i.c.v. with Aβ 42 oligomers (10 μl/rat), (3) Aβ 1-42+Cbx group rats injected i.c.v. with Aβ 42 oligomers (10 μl/rat) and i.p. with carbenoxolone disodium (20 mg/kg body weight) for six-weeks and (4) Cbx group rats injected i.p. with carbenoxolone disodium (20 mg/kg body weight) for six-weeks. Progressive learning and memory deficits were seen through a battery of behavioral tests and a significant increase in the expressions of GFAP and Iba-1 was observed which resulted in the release of pro-inflammatory cytokines post Aβ oligomer injection. The levels of BDNF, Bcl-2 and pCREB were decreased while Bax, caspase-3, caspase-9 and cytochrome c levels were induced. Also, neurotransmitter levels were altered and neuronal damage was observed through histopathological studies. After Cbx supplementation, the expressions of GFAP, IBA-1, pro-inflammatory cytokines, iNOS, nNOS and nitric oxide levels were normalized. The expression levels of pro-apoptotic markers were decreased and neurotrophin levels were restored. Also, neurotransmitter levels and neuronal profile were improved and progressive improvements in behavioural performance were observed. Our results demonstrated that Cbx might have prevented the Aβ induced neurodegeneration and cognitive decline by inhibiting the neuroinflammation and inducing BDNF/CREB signalling. These findings suggest that Cbx can be explored as a potential therapeutic agent against the progression of AD.
Collapse
Affiliation(s)
- Sheetal Sharma
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh 160014, India.
| | - Avneet Saini
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh 160014, India.
| | - Bimla Nehru
- Department of Biophysics, Basic Medical Sciences Block II, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
8
|
Foley AR, Raskatov JA. Assessing Reproducibility in Amyloid β Research: Impact of Aβ Sources on Experimental Outcomes. Chembiochem 2020; 21:2425-2430. [PMID: 32249510 PMCID: PMC7647053 DOI: 10.1002/cbic.202000125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/04/2020] [Indexed: 12/16/2022]
Abstract
The difficulty of synthesizing and purifying the amyloid β (Aβ) peptide, combined with its high aggregation propensity and low solubility under physiological conditions, leads to a wide variety of experimental results from kinetic assays to biological activity. Thus, it becomes challenging to reproduce outcomes, and this limits our ability to rely on reported results as the foundation for new research. This article examines variability of the Aβ peptide from different sources, comparing purity, and oligomer and fibril formation propensity side by side. The results highlight the importance of performing rigorous controls so that meaningful biophysical, biochemical, and neurobiological results can be obtained to improve our understanding on Aβ.
Collapse
Affiliation(s)
- Alejandro R Foley
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Jevgenij A Raskatov
- Department of Chemistry and Biochemistry, University of California Santa Cruz, 1156 High Street, Santa Cruz, CA, 95064, USA
| |
Collapse
|
9
|
Effect of Aβ Oligomers on Neuronal APP Triggers a Vicious Cycle Leading to the Propagation of Synaptic Plasticity Alterations to Healthy Neurons. J Neurosci 2020; 40:5161-5176. [PMID: 32444385 DOI: 10.1523/jneurosci.2501-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/04/2020] [Accepted: 04/03/2020] [Indexed: 01/04/2023] Open
Abstract
Alterations of excitatory synaptic function are the strongest correlate to the pathologic disturbance of cognitive ability observed in the early stages of Alzheimer's disease (AD). This pathologic feature is driven by amyloid-β oligomers (Aβos) and propagates from neuron to neuron. Here, we investigated the mechanism by which Aβos affect the function of synapses and how these alterations propagate to surrounding healthy neurons. We used complementary techniques ranging from electrophysiological recordings and molecular biology to confocal microscopy in primary cortical cultures, and from acute hippocampal and cortical slices from male wild-type and amyloid precursor protein (APP) knock-out (KO) mice to assess the effects of Aβos on glutamatergic transmission, synaptic plasticity, and dendritic spine structure. We showed that extracellular application of Aβos reduced glutamatergic synaptic transmission and long-term potentiation. These alterations were not observed in APP KO neurons, suggesting that APP expression is required. We demonstrated that Aβos/APP interaction increases the amyloidogenic processing of APP leading to intracellular accumulation of newly produced Aβos. Intracellular Aβos participate in synaptic dysfunctions as shown by pharmacological inhibition of APP processing or by intraneuronal infusion of an antibody raised against Aβos. Furthermore, we provide evidence that following APP processing, extracellular release of Aβos mediates the propagation of the synaptic pathology characterized by a decreased spine density of neighboring healthy neurons in an APP-dependent manner. Together, our data unveil a complementary role for Aβos in AD, while intracellular Aβos alter synaptic function, extracellular Aβos promote a vicious cycle that propagates synaptic pathology from diseased to healthy neurons.SIGNIFICANCE STATEMENT Here we provide the proof that a vicious cycle between extracellular and intracellular pools of Aβ oligomers (Aβos) is required for the spreading of Alzheimer's disease (AD) pathology. We showed that extracellular Aβos propagate excitatory synaptic alterations by promoting amyloid precursor protein (APP) processing. Our results also suggest that subsequent to APP cleavage two pools of Aβos are produced. One pool accumulates inside the cytosol, inducing the loss of synaptic plasticity potential. The other pool is released into the extracellular space and contributes to the propagation of the pathology from diseased to healthy neurons. Pharmacological strategies targeting the proteolytic cleavage of APP disrupt the relationship between extracellular and intracellular Aβ, providing a therapeutic approach for the disease.
Collapse
|
10
|
Ability of selenium species to inhibit metal-induced Aβ aggregation involved in the development of Alzheimer’s disease. Anal Bioanal Chem 2020; 412:6485-6497. [DOI: 10.1007/s00216-020-02644-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/27/2020] [Accepted: 04/03/2020] [Indexed: 12/20/2022]
|
11
|
Sharma S, Saini R, Sharma P, Saini A, Nehru B. Maintenance of Amyloid-beta Homeostasis by Carbenoxolone Post Aβ-42 Oligomer Injection in Rat Brain. Neuroscience 2020; 431:86-102. [DOI: 10.1016/j.neuroscience.2020.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 10/25/2022]
|
12
|
Wei Y, Zhou J, Wu J, Huang J. ERβ promotes Aβ degradation via the modulation of autophagy. Cell Death Dis 2019; 10:565. [PMID: 31332160 PMCID: PMC6646346 DOI: 10.1038/s41419-019-1786-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's Disease (AD) is the most common neurodegenerative disorder in the elderly. Beta-amyloid (Aβ) peptide accumulation is considered as a primary cause of AD pathogenesis, with defective autophagy in patients' brains. Enhanced autophagic activity has been reported to promote Aβ clearance in vitro and in vivo models. Meanwhile, there is growing evidence that estrogen receptor β (ERβ) is a viable therapeutic target that can ameliorate the pathological features associated with AD. Very little is known about the detailed molecular mechanisms underlying the relationship between ERβ, autophagy, and Aβ degradation in AD. This study aims to uncover whether ERβ participates in autophagy and promotes extracellular Aβ1-42 degradation through the autophagy-lysosome system. Here we find that overexpression of ERβ caused autophagic activation as seen by increased microtubule-associated protein 1 light chain 3-II (LC3-II), SQSTM1 (sequestosome 1) degradation, LC3 punctate distribution, autophagosome, and autolysosome accumulation. In addition, we show that ERβ could induce autophagy through direct protein-protein interaction with ATG7 (E1-like enzyme). Furthermore, ERβ-mediated decrease in Aβ1-42 was blocked by the autophagy inhibitor chloroquine (CQ) in SH-SY5Y cells and the HEK293T (AβPPsw) model. Aβ1-42 or CQ induced cytotoxicity was restored by a selective ERβ activator diarylpropionitrile (DPN). Collectively, these data indicate that overexpression of ERβ exerts a neuroprotective effect through interacting with ATG7 protein and further enhances autophagy-lysosomal activity for Aβ1-42 clearance at the cellular level.
Collapse
Affiliation(s)
- Yong Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jiawei Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jun Wu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China.
| |
Collapse
|
13
|
Sharma S, Sharma N, Saini A, Nehru B. Carbenoxolone Reverses the Amyloid Beta 1–42 Oligomer–Induced Oxidative Damage and Anxiety-Related Behavior in Rats. Neurotox Res 2018; 35:654-667. [DOI: 10.1007/s12640-018-9975-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 10/17/2018] [Accepted: 10/22/2018] [Indexed: 12/25/2022]
|
14
|
Thei L, Imm J, Kaisis E, Dallas ML, Kerrigan TL. Microglia in Alzheimer's Disease: A Role for Ion Channels. Front Neurosci 2018; 12:676. [PMID: 30323735 PMCID: PMC6172337 DOI: 10.3389/fnins.2018.00676] [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: 03/30/2018] [Accepted: 09/07/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease is the most common form of dementia, it is estimated to affect over 40 million people worldwide. Classically, the disease has been characterized by the neuropathological hallmarks of aggregated extracellular amyloid-β and intracellular paired helical filaments of hyperphosphorylated tau. A wealth of evidence indicates a pivotal role for the innate immune system, such as microglia, and inflammation in the pathology of Alzheimer's disease. The over production and aggregation of Alzheimer's associated proteins results in chronic inflammation and disrupts microglial clearance of these depositions. Despite being non-excitable, microglia express a diverse array of ion channels which shape their physiological functions. In support of this, there is a growing body of evidence pointing to the involvement of microglial ion channels contributing to neurodegenerative diseases such as Alzheimer's disease. In this review, we discuss the evidence for an array of microglia ion channels and their importance in modulating microglial homeostasis and how this process could be disrupted in Alzheimer's disease. One promising avenue for assessing the role that microglia play in the initiation and progression of Alzheimer's disease is through using induced pluripotent stem cell derived microglia. Here, we examine what is already understood in terms of the molecular underpinnings of inflammation in Alzheimer's disease, and the utility that inducible pluripotent stem cell derived microglia may have to advance this knowledge. We outline the variability that occurs between the use of animal and human models with regards to the importance of microglial ion channels in generating a relevant functional model of brain inflammation. Overcoming these hurdles will be pivotal in order to develop new drug targets and progress our understanding of the pathological mechanisms involved in Alzheimer's disease.
Collapse
Affiliation(s)
- Laura Thei
- Reading School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Jennifer Imm
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Eleni Kaisis
- Reading School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Talitha L Kerrigan
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
15
|
Zhang M, Peyear T, Patmanidis I, Greathouse DV, Marrink SJ, Andersen OS, Ingólfsson HI. Fluorinated Alcohols' Effects on Lipid Bilayer Properties. Biophys J 2018; 115:679-689. [PMID: 30077334 PMCID: PMC6104562 DOI: 10.1016/j.bpj.2018.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/23/2018] [Accepted: 07/02/2018] [Indexed: 12/21/2022] Open
Abstract
Fluorinated alcohols (fluoroalcohols) have physicochemical properties that make them excellent solvents of peptides, proteins, and other compounds. Like other alcohols, fluoroalcohols also alter membrane protein function and lipid bilayer properties and stability. Thus, the questions arise: how potent are fluoroalcohols as lipid-bilayer-perturbing compounds, could small residual amounts that remain after adding compounds dissolved in fluoroalcohols alter lipid bilayer properties sufficiently to affect membranes and membrane protein function, and do they behave like other alcohols? To address these questions, we used a gramicidin-based fluorescence assay to determine the bilayer-modifying potency of selected fluoroalcohols: trifluoroethanol (TFE), HFIP, and perfluoro-tert-butanol (PFTB). These fluoroalcohols alter bilayer properties in the low (PFTB) to high (TFE) mM range. Using the same assay, we determined the bilayer partitioning of the alcohols. When referenced to the aqueous concentrations, the fluoroalcohols are more bilayer perturbing than their nonfluorinated counterparts, with the largest fluoroalcohol, PFTB, being the most potent and the smallest, TFE, the least. When referenced to the mole fractions in the membrane, however, the fluoroalcohols have equal or lesser bilayer-perturbing potency than their nonfluorinated counterparts, with TFE being more bilayer perturbing than PFTB. We compared the fluoroalcohols' molecular level bilayer interactions using atomistic molecular dynamics simulations and showed how, at higher concentrations, they can cause bilayer breakdown using absorbance measurements and 31P nuclear magnetic resonance.
Collapse
Affiliation(s)
- Mike Zhang
- Department Physiology and Biophysics, Weill Cornell Medicine, New York City, New York; The Bronx High School of Science, New York City, New York
| | - Thasin Peyear
- Department Physiology and Biophysics, Weill Cornell Medicine, New York City, New York
| | - Ilias Patmanidis
- Groningen Biomolecular Science and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, the Netherlands
| | - Denise V Greathouse
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas
| | - Siewert J Marrink
- Groningen Biomolecular Science and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, the Netherlands
| | - Olaf S Andersen
- Department Physiology and Biophysics, Weill Cornell Medicine, New York City, New York.
| | - Helgi I Ingólfsson
- Department Physiology and Biophysics, Weill Cornell Medicine, New York City, New York; Groningen Biomolecular Science and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, the Netherlands; Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California.
| |
Collapse
|
16
|
Affiliation(s)
- Ferenc Zsila
- Biomolecular Self-Assembly Group; Institute of Materials and Environmental Chemistry; Research Centre for Natural Sciences; Hungarian Academy of Sciences; 1117 Budapest Hungary
| |
Collapse
|
17
|
Sharma S, Nehru B, Saini A. Inhibition of Alzheimer's amyloid-beta aggregation in-vitro by carbenoxolone: Insight into mechanism of action. Neurochem Int 2017; 108:481-493. [DOI: 10.1016/j.neuint.2017.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/09/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022]
|
18
|
Use-dependent inhibition of glycine-activated chloride current in rat neurons by β-amyloid peptide pretreated with hexafluoroisopropanol. Neuroreport 2017; 28:579-583. [PMID: 28489663 DOI: 10.1097/wnr.0000000000000801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hexafluoroisopropanol (HFIP) is a nonpolar organic solvent that is often used to prepare β-amyloid peptide (Aβ) samples. In this work, we compare the effects of two different species derived from synthetic Aβ1-42 and prepared without HFIP (Aβ) or using HFIP (Aβ/HFIP) on the glycine-activated chloride current (IGly). The experiments were conducted on the pyramidal neurons isolated from CA3 region of rat hippocampus. Transmembrane currents were recorded using a conventional patch-clamp technique in the whole-cell configuration. The IGly was induced by a step application of the agonist for 600 ms through glass capillary. Aβ or Aβ/HFIP was coapplied with glycine. The effects of the two species of the peptide have similar and distinctive features. Both substances caused a reduction in the peak amplitude and an acceleration of desensitization of the IGly. At the same time, the effect of Aβ/HFIP was found to develop and recover more slowly and required several repeated applications for its saturation (use dependence). The effect of Aβ/HFIP was voltage independent and equally pronounced at negative and positive membrane potentials. First, our results confirm that HFIP pretreatment may influence the properties of Aβ. Second, new information on the glycine receptor ability to interact with drugs in use-dependent mode was obtained.
Collapse
|
19
|
Kaufmann TJ, Harrison PM, Richardson MJE, Pinheiro TJT, Wall MJ. Intracellular soluble α-synuclein oligomers reduce pyramidal cell excitability. J Physiol 2016; 594:2751-72. [PMID: 26915902 PMCID: PMC4865569 DOI: 10.1113/jp271968] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/17/2016] [Indexed: 02/04/2023] Open
Abstract
Key points The presynaptic protein α‐synuclein forms aggregates during Parkinson's disease. Accumulating evidence suggests that the small soluble oligomers of α‐synuclein are more toxic than the larger aggregates appearing later in the disease. The link between oligomer toxicity and structure still remains unclear. In the present study, we have produced two structurally‐defined oligomers that have a similar morphology but differ in secondary structure. These oligomers were introduced into neocortical pyramidal cells during whole‐cell recording and, using a combination of experimentation and modelling, electrophysiological parameters were extracted. Both oligomeric species had similar effects on neuronal properties reducing input resistance, time constant and increasing capacitance. The net effect was a marked reduction in neuronal excitability that could impact on network activity.
Abstract The presynaptic protein α‐synuclein (αSyn) aggregates during Parkinson's disease (PD) to form large proteinaceous amyloid plaques, the spread of which throughout the brain clinically defines the severity of the disease. During early stages of aggregation, αSyn forms soluble annular oligomers that show greater toxicity than much larger fibrils. These oligomers produce toxicity via a number of possible mechanisms, including the production of pore‐forming complexes that permeabilize membranes. In the present study, two well‐defined species of soluble αSyn oligomers were produced by different protocols: by polymerization of monomer and by sonication of fibrils. The two oligomeric species produced were morphologically similar, with both having an annular structure and consisting of approximately the same number of monomer subunits, although they differed in their secondary structure. Oligomeric and monomeric αSyn were injected directly into the soma of pyramidal neurons in mouse neocortical brain slices during whole‐cell patch clamp recording. Using a combined experimental and modelling approach, neuronal parameters were extracted to measure, for the first time in the neocortex, specific changes in neuronal electrophysiology. Both species of oligomer had similar effects: (i) a significant reduction in input resistance and the membrane time constant and (ii) an increase in the current required to trigger an action potential with a resultant reduction in the firing rate. Differences in oligomer secondary structure appeared to produce only subtle differences in the activity of the oligomers. Monomeric αSyn had no effect on neuronal parameters, even at high concentrations. The oligomer‐induced fall in neuronal excitability has the potential to impact both network activity and cognitive processing. The presynaptic protein α‐synuclein forms aggregates during Parkinson's disease. Accumulating evidence suggests that the small soluble oligomers of α‐synuclein are more toxic than the larger aggregates appearing later in the disease. The link between oligomer toxicity and structure still remains unclear. In the present study, we have produced two structurally‐defined oligomers that have a similar morphology but differ in secondary structure. These oligomers were introduced into neocortical pyramidal cells during whole‐cell recording and, using a combination of experimentation and modelling, electrophysiological parameters were extracted. Both oligomeric species had similar effects on neuronal properties reducing input resistance, time constant and increasing capacitance. The net effect was a marked reduction in neuronal excitability that could impact on network activity.
Collapse
Affiliation(s)
| | - Paul M Harrison
- Warwick Systems Biology Centre, University of Warwick, Coventry, UK
| | | | | | | |
Collapse
|
20
|
Nuclear magnetic resonance evidence for the dimer formation of beta amyloid peptide 1-42 in 1,1,1,3,3,3-hexafluoro-2-propanol. Anal Biochem 2016; 498:59-67. [PMID: 26772162 DOI: 10.1016/j.ab.2015.12.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/04/2015] [Accepted: 12/29/2015] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease involves accumulation of senile plaques in which filamentous aggregates of amyloid beta (Aβ) peptides are deposited. Recent studies demonstrate that oligomerization pathways of Aβ peptides may be complicated. To understand the mechanisms of Aβ(1-42) oligomer formation in more detail, we have established a method to produce (15)N-labeled Aβ(1-42) suited for nuclear magnetic resonance (NMR) studies. For physicochemical studies, the starting protein material should be solely monomeric and all Aβ aggregates must be removed. Here, we succeeded in fractionating a "precipitation-resistant" fraction of Aβ(1-42) from an "aggregation-prone" fraction by high-performance liquid chromatography (HPLC), even from bacterially overexpressed Aβ(1-42). However, both Aβ(1-42) fractions after 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) treatment formed amyloid fibrils. This indicates that the "aggregation seed" was not completely monomerized during HFIP treatment. In addition, Aβ(1-42) dissolved in HFIP was found to display a monomer-dimer equilibrium, as shown by two-dimensional (1)H-(15)N NMR. We demonstrated that the initial concentration of Aβ during the HFIP pretreatment altered the kinetic profiles of Aβ fibril formation in a thioflavin T fluorescence assay. The findings described here should ensure reproducible results when studying the Aβ(1-42) peptide.
Collapse
|
21
|
Malishev R, Nandi S, Kolusheva S, Levi-Kalisman Y, Klärner FG, Schrader T, Bitan G, Jelinek R. Toxicity inhibitors protect lipid membranes from disruption by Aβ42. ACS Chem Neurosci 2015; 6:1860-9. [PMID: 26317327 DOI: 10.1021/acschemneuro.5b00200] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although the precise molecular factors linking amyloid β-protein (Aβ) to Alzheimer's disease (AD) have not been deciphered, interaction of Aβ with cellular membranes has an important role in the disease. However, most therapeutic strategies targeting Aβ have focused on interfering with Aβ self-assembly rather than with its membrane interactions. Here, we studied the impact of three toxicity inhibitors on membrane interactions of Aβ42, the longer form of Aβ, which is associated most strongly with AD. The inhibitors included the four-residue C-terminal fragment Aβ(39-42), the polyphenol (-)-epigallocatechin-3-gallate (EGCG), and the lysine-specific molecular tweezer, CLR01, all of which previously were shown to disrupt different steps in Aβ42 self-assembly. Biophysical experiments revealed that incubation of Aβ42 with each of the three modulators affected membrane interactions in a distinct manner. Interestingly, EGCG and CLR01 were found to have significant interaction with membranes themselves. However, membrane bilayer disruption was reduced when the compounds were preincubated with Aβ42, suggesting that binding of the assembly modulators to the peptide attenuated their membrane interactions. Importantly, our study reveals that even though the three tested compounds affect Aβ42 assembly differently, membrane interactions were significantly inhibited upon incubation of each compound with Aβ42, suggesting that preventing the interaction of Aβ42 with the membrane contributes substantially to inhibition of its toxicity by each compound. The data suggest that interference with membrane interactions is an important factor for Aβ42 toxicity inhibitors and should be taken into account in potential therapeutic strategies, in addition to disruption or remodeling of amyloid assembly.
Collapse
Affiliation(s)
- Ravit Malishev
- Department
of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sukhendu Nandi
- Department
of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Sofiya Kolusheva
- Ilse
Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Yael Levi-Kalisman
- Department
of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Frank-Gerrit Klärner
- Institute
of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Thomas Schrader
- Institute
of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Gal Bitan
- Department
of Neurology, David Geffen School of Medicine, Brain Research Institute,
and Molecular Biology Institute, University of California at Los Angeles, Los
Angeles, California 90095, United States
| | - Raz Jelinek
- Department
of Chemistry, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
- Ilse
Katz Institute for Nanotechnology, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
22
|
Rangaraju S, Gearing M, Jin LW, Levey A. Potassium channel Kv1.3 is highly expressed by microglia in human Alzheimer's disease. J Alzheimers Dis 2015; 44:797-808. [PMID: 25362031 DOI: 10.3233/jad-141704] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent genetic studies suggest a central role for innate immunity in Alzheimer's disease (AD) pathogenesis, wherein microglia orchestrate neuroinflammation. Kv1.3, a voltage-gated potassium channel of therapeutic relevance in autoimmunity, is upregulated by activated microglia and mediates amyloid-mediated microglial priming and reactive oxygen species production in vitro. We hypothesized that Kv1.3 channel expression is increased in human AD brain tissue. In a blinded postmortem immunohistochemical semi-quantitative analysis performed on ten AD patients and ten non-disease controls, we observed a significantly higher Kv1.3 staining intensity (p = 0.03) and Kv1.3-positive cell density (p = 0.03) in the frontal cortex of AD brains, compared to controls. This paralleled an increased number of Iba1-positive microglia in AD brains. Kv1.3-positive cells had microglial morphology and were associated with amyloid-β plaques. In immunofluorescence studies, Kv1.3 channels co-localized primarily with Iba1 but not with astrocyte marker GFAP, confirming that elevated Kv1.3 expression is limited to microglia. Higher Kv1.3 expression in AD brains was also confirmed by western blot analysis. Our findings support that Kv1.3 channels are biologically relevant and microglia-specific targets in human AD.
Collapse
Affiliation(s)
| | - Marla Gearing
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, Alzheimer's Disease Center, University of California Davis, CA, USA
| | - Allan Levey
- Department of Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
23
|
Porzoor A, Caine JM, Macreadie IG. Pretreatment of chemically-synthesized Aβ42 affects its biological activity in yeast. Prion 2015; 8:404-10. [PMID: 25495906 DOI: 10.4161/19336896.2014.992275] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The tendency of amyloid β (Aβ42) peptide to misfold and aggregate into insoluble amyloid fibrils in Alzheimer's disease (AD) has been well documented. Accumulation of Aβ42 fibrils has been correlated with abnormal apoptosis and unscheduled cell division which can also trigger the death of neuronal cells, while oligomers can also exhibit similar activities. While investigations using chemically-synthesized Aβ42 peptide have become common practice, there appear to be differences in outcomes from different preparations. In order to resolve this inconsistency, we report 2 separate methods of preparing chemically-synthesized Aβ42 and we examined their effects in yeast. Hexafluoroisopropanol pretreatment caused toxicity while, ammonium hydroxide treated Aβ42 induced cell proliferation in both C. glabrata and S. cerevisiae. The hexafluoroisopropanol prepared Aβ42 had greater tendency to form amyloid on yeast cells as determined by thioflavin T staining followed by flow cytometry and microscopy. Both quiescent and non-quiescent cells were analyzed by these methods of peptide preparation. Non-quiescent cells were susceptible to the toxicity of Aβ42 compared with quiescent cells (p < 0.005). These data explain the discrepancy in the previous publications about the effects of chemically-synthesized Aβ42 on yeast cells. The effect of Aβ42 on yeast cells was independent of the size of the peptide aggregates. However, the Aβ42 pretreatment determined whether the molecular conformation of peptide resulted in proliferation or toxicity in yeast based assays.
Collapse
Affiliation(s)
- Afsaneh Porzoor
- a School of Applied Sciences; Biosciences ; RMIT University ; Bundoora , Victoria , Australia
| | | | | |
Collapse
|
24
|
Zhu D, Bungart BL, Yang X, Zhumadilov Z, Lee JCM, Askarova S. Role of membrane biophysics in Alzheimer's-related cell pathways. Front Neurosci 2015; 9:186. [PMID: 26074758 PMCID: PMC4444756 DOI: 10.3389/fnins.2015.00186] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/11/2015] [Indexed: 01/04/2023] Open
Abstract
Cellular membrane alterations are commonly observed in many diseases, including Alzheimer's disease (AD). Membrane biophysical properties, such as membrane molecular order, membrane fluidity, organization of lipid rafts, and adhesion between membrane and cytoskeleton, play an important role in various cellular activities and functions. While membrane biophysics impacts a broad range of cellular pathways, this review addresses the role of membrane biophysics in amyloid-β peptide aggregation, Aβ-induced oxidative pathways, amyloid precursor protein processing, and cerebral endothelial functions in AD. Understanding the mechanism(s) underlying the effects of cell membrane properties on cellular processes should shed light on the development of new preventive and therapeutic strategies for this devastating disease.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Chemical, Biological and Bioengineering, North Carolina A&T State UniversityGreensboro, NC, USA
| | - Brittani L. Bungart
- Indiana University School of Medicine Medical Scientist Training Program, Indiana University School of MedicineIndianapolis, IN, USA
| | - Xiaoguang Yang
- Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of GothenburgGothenburg, Sweden
- The Hope Center for Neurological Disorders and Department of Neurology, Washington University School of MedicineSt. Louis, MO, USA
| | - Zhaxybay Zhumadilov
- Department of Bioengineering and Regenerative Medicine, Center for Life Sciences, Nazarbayev UniversityAstana, Kazakhstan
| | - James C-M. Lee
- Department of Bioengineering, University of Illinois at ChicagoChicago, IL, USA
| | - Sholpan Askarova
- Department of Bioengineering and Regenerative Medicine, Center for Life Sciences, Nazarbayev UniversityAstana, Kazakhstan
| |
Collapse
|
25
|
Hung SY, Huang WP, Liou HC, Fu WM. LC3 overexpression reduces Aβ neurotoxicity through increasing α7nAchR expression and autophagic activity in neurons and mice. Neuropharmacology 2015; 93:243-51. [PMID: 25686800 DOI: 10.1016/j.neuropharm.2015.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 01/26/2015] [Accepted: 02/01/2015] [Indexed: 12/19/2022]
Abstract
Autophagy is an intracellular degradation pathway with dynamic interactions for eliminating damaged organelles and protein aggregates by lysosomal digestion. The EGFP-conjugated microtubule-associated protein 1 light chain 3 (EGFP-LC3) serves to monitor autophagic process. Extracellular β-amyloid peptide accumulation is reported as a major cause in Alzheimer's disease (AD) pathogenesis; large numbers of autophagic vacuoles accumulate in patients' brains. We previously demonstrated that extracellular Aβ (eAβ) induces strong autophagic response and α7nAChR acts as a carrier to bind with eAβ; which further inhibits Aβ-induced neurotoxicity via autophagic degradation. In the present study, we overexpressed LC3 in both neuroblastoma cells (SH-SY5Y/pEGFP-LC3) and mice (TgEGFP-LC3) to assess the effect of LC3 overexpression on Aβ neurotoxicity. SH-SY5Y/pEGFP-LC3 cells and primary cortical neuron cultures derived from E17 (embryonic day 17) TgEGFP-LC3 mice showed not only better resistance against Aβ neurotoxicity but also higher α7nAChR expression and autophagic activity than control. Administration of α-bungarotoxin (α-BTX) to block α7nAChR antagonized the neuroprotective action of SH-SY5Y/pECGF-LC3 cells, suggesting that eAβ binding with α7nAChR is an important step in Aβ detoxification. LC3 overexpression thus exerts neuroprotection through increasing α7nAChR expression for eAβ binding and further enhancing autophagic activity for Aβ clearance in vitro and in vivo.
Collapse
Affiliation(s)
- Shih-Ya Hung
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Wei-Pang Huang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Houng-Chi Liou
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Wen-Mei Fu
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei 10051, Taiwan.
| |
Collapse
|
26
|
Morkuniene R, Cizas P, Jankeviciute S, Petrolis R, Arandarcikaite O, Krisciukaitis A, Borutaite V. Small Aβ1-42 oligomer-induced membrane depolarization of neuronal and microglial cells: role of N-methyl-D-aspartate receptors. J Neurosci Res 2014; 93:475-86. [PMID: 25400096 DOI: 10.1002/jnr.23510] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 10/01/2014] [Accepted: 10/02/2014] [Indexed: 11/08/2022]
Abstract
Although it is well documented that soluble beta amyloid (Aβ) oligomers are critical factors in the pathogenesis of Alzheimer's disease (AD) by causing synaptic dysfunction and neuronal death, the primary mechanisms by which Aβ oligomers trigger neurodegeneration are not entirely understood. We sought to investigate whether toxic small Aβ(1-42) oligomers induce changes in plasma membrane potential of cultured neurons and glial cells in rat cerebellar granule cell cultures leading to neuronal death and whether these effects are sensitive to the N-methyl-D-aspartate receptor (NMDA-R) antagonist MK801. We found that small Aβ(1-42) oligomers induced rapid, protracted membrane depolarization of both neurons and microglia, whereas there was no change in membrane potential of astrocytes. MK801 did not modulate Aβ-induced neuronal depolarization. In contrast, Aβ1(-42) oligomer-induced decrease in plasma membrane potential of microglia was prevented by MK801. Small Aβ(1-42) oligomers significantly elevated extracellular glutamate and caused neuronal necrosis, and both were prevented by MK801. Also, small Aβ(1-42) oligomers decreased resistance of isolated brain mitochondria to calcium-induced opening of mitochondrial permeability transition pore. In conclusion, the results suggest that the primary effect of toxic small Aβ oligomers on neurons is rapid, NMDA-R-independent plasma membrane depolarization, which leads to neuronal death. Aβ oligomers-induced depolarization of microglial cells is NMDA-R dependent.
Collapse
Affiliation(s)
- Ramune Morkuniene
- Institute of Neurosciences, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | | | | | | | |
Collapse
|
27
|
Yoshiya T, Maruno T, Uemura T, Kubo S, Kiso Y, Sohma Y, Yoshizawa-Kumagaye K, Kobayashi Y, Nishiuchi Y. Non-pretreated O-acyl isopeptide of amyloid β peptide 1-42 is monomeric with a random coil structure but starts to aggregate in a concentration-dependent manner. Bioorg Med Chem Lett 2014; 24:3861-4. [PMID: 25017031 DOI: 10.1016/j.bmcl.2014.06.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/17/2014] [Accepted: 06/19/2014] [Indexed: 11/25/2022]
Abstract
An isopeptide of amyloid β peptide 1-42 (isoAβ42) was considered as a non-aggregative precursor molecule for the highly aggregative Aβ42. It has been applied to biological studies after several pretreatments. Here we report that isoAβ42 is monomeric with a random coil structure at 40 μM without any pretreatment. But we also found that isoAβ42 retains a slight aggregative nature, which is significantly weaker than that of the native Aβ42.
Collapse
Affiliation(s)
- Taku Yoshiya
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan.
| | - Takahiro Maruno
- Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Tsuyoshi Uemura
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan
| | - Shigeru Kubo
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan
| | - Yoshiaki Kiso
- Laboratory of Peptide Science, Nagahama Institute of Bio-Science and Technology, Shiga 526-0829, Japan
| | - Youhei Sohma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | | | - Yuji Kobayashi
- Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Yuji Nishiuchi
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| |
Collapse
|
28
|
Wilson NP, Gates B, Castellanos M. Modeling the short time-scale dynamics of β-amyloid–neuron interactions. J Theor Biol 2013; 331:28-37. [DOI: 10.1016/j.jtbi.2013.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 01/08/2013] [Accepted: 02/18/2013] [Indexed: 12/31/2022]
|