1
|
Zhao H, Fan M, Zhang J, Gao Y, Chen L, Huang L. Amyloid beta-induced mitochondrial dysfunction and endothelial permeability in cerebral microvascular endothelial cells: The protective role of dexmedetomidine. Brain Res Bull 2025; 220:111137. [PMID: 39577505 DOI: 10.1016/j.brainresbull.2024.111137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication in patients who undergo anesthesia in different types of surgeries. Emerging evidence implicates elevated beta-amyloid (Aβ) in the pathogenesis of POCD. Meanwhile, Dexmedetomidine (DEX) has recently shown promise in reducing POCD incidence. This study aimed to elucidate the role of Aβ in inducing endothelial permeability in cerebral microvascular endothelial cells and the underlying mechanisms and testing the effects of DEX. We demonstrated that Aβ1-42, the prevalent Aβ form related to POCD, is cytotoxic to HBMECs, increasing transendothelial permeability and inducing mitochondrial dysfunction, as evidenced by elevated mitochondrial reactive oxygen species (ROS) and decreased ATP production and mitochondrial membrane potential. Furthermore, Aβ1-42 was shown to inhibit Sirt3, exacerbating mitochondrial dysfunction. Conversely, DEX was found to prevent Aβ1-42-induced mitochondrial dysfunction and permeability increases and preserved tight junction proteins in HBMECs.These findings suggest that DEX, as a Sirt3 activator, may offer a pharmacological strategy to mitigate Aβ1-42-related cerebral microvascular endothelial cell dysfunction and preserve cognitive function post-surgery.
Collapse
Affiliation(s)
- Haifeng Zhao
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050061, PR China; Department of Anesthesiology, Shijiazhuang Obstetrics and Gynecology Hospital, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, PR China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Jin Zhang
- Department of Anesthesiology, Shijiazhuang Obstetrics and Gynecology Hospital, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, PR China
| | - Yi Gao
- Department of Anesthesiology, Shijiazhuang Obstetrics and Gynecology Hospital, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, PR China
| | - Liang Chen
- Department of Anesthesiology, Shijiazhuang Obstetrics and Gynecology Hospital, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, PR China
| | - Lining Huang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050061, PR China; Hebei Key Laboratory of Neurodegenerative Disease Mechanism, PR China; Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, PR China.
| |
Collapse
|
2
|
Zhao F, Fan L, Yang J, Yang M, Zhang C, Wang F, Wang Y. Heterologous expression of BACE1 and its interaction with Codonopsis pilosula polysaccharides and Lobetyolin. Int J Biol Macromol 2024; 277:133440. [PMID: 38944075 DOI: 10.1016/j.ijbiomac.2024.133440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
BACE1, a crucial enzyme in the amyloid-β deposition theory of Alzheimer's disease (AD), is targeted by Codonopsis pilosula, a traditional tonic believed to impede AD onset. However, the specific active compounds responsible for its effects remain elusive. Our prior network pharmacology research identified C. pilosula polysaccharides (CPPS) and Lobetyolin may serve as potential inhibitors of AD by suppressing amyloidogenesis. Here, we recombinantly expressed BACE1 under varied conditions and assessed its activity using Fluorescence Resonance Energy Transfer technology. Through spectroscopy, molecular docking, and dynamics, we elucidated the interactions of CPPS, Lobetyolin, and BACE1. Optimal BACE1 expression occurred at 22 °C with 0.4 mM IPTG for 6 h, yielding a 72 kDa protein. Enzyme kinetics displayed a maximum rate of 4096 μmol/min and a Michaelis constant of 16 mg/mL for BACE1. Spectroscopic analysis revealed differing binding affinities of the compounds at various temperatures, peaking at 293 K. Lobetyolin exhibited superior binding to BACE1 compared to CPPS, driven by hydrophobic and electrostatic forces. Molecular docking and dynamics highlighted hydrophobic amino acids' role in BACE1 interactions with Lobetyolin and CPPS, with binding energy < -1.2 kcal/mol signifying strong affinities. Notably, Lobetyolin and CPPS showed higher BACE1 affinity than APP, with the Lobetyolin-BACE1 complex being the most stable.
Collapse
Affiliation(s)
- Feitao Zhao
- School of life science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Lili Fan
- School of life science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Jumei Yang
- Lanzhou University Second Hospital, Lanzhou 730000, China
| | - Mingjun Yang
- School of life science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Chun Zhang
- Lanzhou University of Technology Hospital, Lanzhou 730000, China
| | - Fang Wang
- School of life science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | - Yonggang Wang
- School of life science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| |
Collapse
|
3
|
Zondagh LS, Malan SF, Joubert J. Edaravone N-benzyl pyridinium derivatives: BACE-1 inhibition, kinetics and in silico binding pose determination. Eur J Pharm Sci 2024; 201:106869. [PMID: 39102997 DOI: 10.1016/j.ejps.2024.106869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/08/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
BACE-1 plays a pivotal role in the production of β-amyloid (Aβ) peptides, implicated in Alzheimer's Disease (AD) pathology. We previously described edaravone N-benzyl pyridinium derivatives (EBPDs) that exhibited multifunctional activity against multiple AD targets. In this study we explored the EBPDs BACE-1 inhibitory activity to potentially enhance the compounds therapeutic profile. The EBPDs exhibited moderate BACE-1 inhibitory activity (IC50 = 44.10 µM - 123.70 µM) and obtained IC50 values between 2.0 and 5.8-fold greater than resveratrol, a known BACE-1 inhibitor (IC50 = 253.20 µM), in this assay. Compound 3 was the most potent inhibitor with an IC50 of 44.10 µM and a Ki of 19.96 µM and a mixed-type mode of inhibition that favored binding in a competitive manner. Molecular docking identified crucial interactions with BACE-1 active site residues, supported by 100 ns MD simulations. The study highlighted the EBPDs therapeutic potential as BACE-1 inhibitors and multifunctional anti-AD therapeutic agents.
Collapse
Affiliation(s)
- L S Zondagh
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Cape Town, Western Cape 7535, South Africa
| | - S F Malan
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Cape Town, Western Cape 7535, South Africa
| | - J Joubert
- Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Cape Town, Western Cape 7535, South Africa.
| |
Collapse
|
4
|
Sehra N, Parmar R, Maurya IK, Kumar V, Tikoo K, Jain R. Synthesis and mechanistic study of ultrashort peptides that inhibits Alzheimer's Aβ-aggregation-induced neurotoxicity. Bioorg Chem 2024; 144:107159. [PMID: 38309001 DOI: 10.1016/j.bioorg.2024.107159] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/02/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Misfolding/aggregation of β-amyloid peptide lead to the formation of toxic oligomers or accumulation of amyloid plaques, which is a seminal step in the progression of Alzheimer's disease (AD). Despite continuous efforts in the development of therapeutic agents, the cure for AD remains a major challenge. Owing to specific binding affinity of structure-based peptides, we report the synthesis of new peptide-based inhibitors derived from the C-terminal sequences, Aβ38-40 and Aβ40-42. Preliminary screening using MTT cell viability assay and corroborative results from ThT fluorescence assay revealed a tripeptide showing significantly effective inhibition towards Aβ1-42 aggregation and induced toxicity. Peptide 3 exhibited excellent cell viability of 94.3 % at 2 μM and of 100 % at 4 μM and 10 μM. CD study showed that peptide 3 restrict the conformation transition of Aβ1-42 peptide towards cross-β-sheet structure and electron microscopy validated the absence of Aβ aggregates as indicated by the altered morphology of Aβ1-42 in the presence of peptide 3. The HRMS-ESI, DLS and ANS studies were performed to gain mechanistic insights into the effect of inhibitor against Aβ aggregation. This Aβ-derived ultrashort motif provides impetus for the development of peptide-based anti-AD agents.
Collapse
Affiliation(s)
- Naina Sehra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Rajesh Parmar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Indresh K Maurya
- Center of Infectious Disease, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Vinod Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
5
|
Firoozi A, Alizadeh A, Zarifkar A, Esmaeilpour T, Namavar MR, Alavi O, Dehghani F. Comparison of the efficacy of human umbilical cord mesenchymal stem cells conditioned medium and platelet-rich plasma on the hippocampus of STZ-induced rat model of Alzheimer's disease: A behavioral and stereological study. IBRO Neurosci Rep 2023; 15:209-217. [PMID: 37780033 PMCID: PMC10539893 DOI: 10.1016/j.ibneur.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/13/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Alzheimer's disease (AD) is accompanied by progressive cognitive disorders and memory loss. This study aims to determine the combined effects of conditioned medium of human umbilical cord mesenchymal stem cells (CM) and platelet-rich plasma (PRP) on AD model rats. Methods Forty-eight male Sprague Dawley rats were classified into 6 groups: Control, Sham, AD, and three treatment groups. AD was induced by streptozotocin(STZ; 3 mg/kg, intracerebroventricular (ICV)) and the treatment groups received injections of CM [(200 µl, intraperitoneally (i.p.), and/or PRP (100 µl, intravenously(i.v)] for 8 days. Behavioral tests (Morris water maze and novel objective recognition) were used to assess learning ability and memory. At the end of the behavioral tests, the rats were sacrificed and their brain was entirely removed, sectioned, and stained with cresyl violet. The hippocampus volume and number of neurons were evaluated by stereological techniques. Results In the AD group, the discrimination ratio, time spent in the target zone, volume of Cornu Ammonis1 (CA1) and Dentate Gyrus (DG), and the number of pyramidal and granular cells decreased significantly compared to the Sham group. The mentioned parameters increased in the CM and CM+PRP groups compared to the AD group (p < 0.01). PRP did not have any noticeable effect on the examined parameters. Conclusions CM may be beneficial in the treatment of AD as it led to better improvement in STZ-induced learning and memory impairments as well as the structure of the hippocampus.
Collapse
Affiliation(s)
- Amin Firoozi
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry & Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aliakbar Alizadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Neuroscience Research Center and Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tahereh Esmaeilpour
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry & Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Alavi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Dehghani
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry & Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Ratan Y, Rajput A, Maleysm S, Pareek A, Jain V, Pareek A, Kaur R, Singh G. An Insight into Cellular and Molecular Mechanisms Underlying the Pathogenesis of Neurodegeneration in Alzheimer's Disease. Biomedicines 2023; 11:biomedicines11051398. [PMID: 37239068 DOI: 10.3390/biomedicines11051398] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is the most prominent neurodegenerative disorder in the aging population. It is characterized by cognitive decline, gradual neurodegeneration, and the development of amyloid-β (Aβ)-plaques and neurofibrillary tangles, which constitute hyperphosphorylated tau. The early stages of neurodegeneration in AD include the loss of neurons, followed by synaptic impairment. Since the discovery of AD, substantial factual research has surfaced that outlines the disease's causes, molecular mechanisms, and prospective therapeutics, but a successful cure for the disease has not yet been discovered. This may be attributed to the complicated pathogenesis of AD, the absence of a well-defined molecular mechanism, and the constrained diagnostic resources and treatment options. To address the aforementioned challenges, extensive disease modeling is essential to fully comprehend the underlying mechanisms of AD, making it easier to design and develop effective treatment strategies. Emerging evidence over the past few decades supports the critical role of Aβ and tau in AD pathogenesis and the participation of glial cells in different molecular and cellular pathways. This review extensively discusses the current understanding concerning Aβ- and tau-associated molecular mechanisms and glial dysfunction in AD. Moreover, the critical risk factors associated with AD including genetics, aging, environmental variables, lifestyle habits, medical conditions, viral/bacterial infections, and psychiatric factors have been summarized. The present study will entice researchers to more thoroughly comprehend and explore the current status of the molecular mechanism of AD, which may assist in AD drug development in the forthcoming era.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sushmita Maleysm
- Department of Bioscience & Biotechnology, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Ranjeet Kaur
- Adesh Institute of Dental Sciences and Research, Bathinda 151101, Punjab, India
| | - Gurjit Singh
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
7
|
Mockett BG, Ryan MM. The therapeutic potential of the neuroactive peptides of soluble amyloid precursor protein-alpha in Alzheimer's disease and related neurological disorders. Semin Cell Dev Biol 2023; 139:93-101. [PMID: 35654665 DOI: 10.1016/j.semcdb.2022.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/31/2022]
Abstract
Soluble amyloid precursor protein-alpha (sAPPα) is a multi-functional brain-derived protein that has neuroprotective, neurogenic and neurotropic properties. Moreover, it is known to facilitate synaptic function and promote neural repair. These properties suggest sAPPα may be useful as a therapeutic agent for the treatment of neurological diseases characterized by synaptic failure and neuronal loss, such as occurs in Alzheimer's disease, and for neural repair following traumatic brain injury and stroke. However, sAPPα's relatively large size and the difficulty of ongoing delivery of therapeutics to the brain mean this is not currently practicable. Importantly, however, sAPPα is composed of several neuroactive domains that each possess properties that collectively are remarkably similar to those of sAPPα itself. Here, we review the molecular structure of sAPPα and identify the domains that contribute to its overall functionality. Four peptide motifs present as possible targets for therapeutic development. We review their physiochemical and neuroactive properties, both within sAPPα and as isolated peptides, and discuss their potential for future development as multipurpose therapeutic agents for the treatment of Alzheimer's disease and other disorders of neuronal function. Further, we discuss the role of heparin binding sites, found within sAPPα's structure and overlapping with the neuroactive domains, as sites for interactions with effector proteins and synaptic receptors. The potential role of the neuroactive peptides known as Cationic Arginine-Rich Peptides (CARPs) as neuroprotective motifs is also reviewed. Mechanisms of peptide delivery to the brain are briefly discussed. Finally, we summarise the potential benefits and pitfalls of using the isolated peptides, either individually or in combination, for the treatment of neurological diseases.
Collapse
Affiliation(s)
- Bruce G Mockett
- Department of Psychology, University of Otago, PO Box 56, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Margaret M Ryan
- Department of Anatomy, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, New Zealand; Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
8
|
Iliyasu MO, Musa SA, Oladele SB, Iliya AI. Amyloid-beta aggregation implicates multiple pathways in Alzheimer's disease: Understanding the mechanisms. Front Neurosci 2023; 17:1081938. [PMID: 37113145 PMCID: PMC10128090 DOI: 10.3389/fnins.2023.1081938] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative condition characterized by tau pathology and accumulations of neurofibrillary tangles (NFTs) along with amyloid-beta (Aβ). It has been associated with neuronal damage, synaptic dysfunction, and cognitive deficits. The current review explained the molecular mechanisms behind the implications of Aβ aggregation in AD via multiple events. Beta (β) and gamma (γ) secretases hydrolyzed amyloid precursor protein (APP) to produce Aβ, which then clumps together to form Aβ fibrils. The fibrils increase oxidative stress, inflammatory cascade, and caspase activation to cause hyperphosphorylation of tau protein into neurofibrillary tangles (NFTs), which ultimately lead to neuronal damage. Acetylcholine (Ach) degradation is accelerated by upstream regulation of the acetylcholinesterase (AChE) enzyme, which leads to a deficiency in neurotransmitters and cognitive impairment. There are presently no efficient or disease-modifying medications for AD. It is necessary to advance AD research to suggest novel compounds for treatment and prevention. Prospectively, it might be reasonable to conduct clinical trials with unclean medicines that have a range of effects, including anti-amyloid and anti-tau, neurotransmitter modulation, anti-neuroinflammatory, neuroprotective, and cognitive enhancement.
Collapse
Affiliation(s)
- Musa O. Iliyasu
- Department of Anatomy, Kogi State University, Anyigba, Nigeria
- *Correspondence: Musa O. Iliyasu, ;
| | - Sunday A. Musa
- Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | - Sunday B. Oladele
- Department of Veterinary Pathology, Ahmadu Bello University, Zaria, Nigeria
| | | |
Collapse
|
9
|
Jeong H, Shin H, Hong S, Kim Y. Physiological Roles of Monomeric Amyloid-β and Implications for Alzheimer's Disease Therapeutics. Exp Neurobiol 2022; 31:65-88. [PMID: 35673997 PMCID: PMC9194638 DOI: 10.5607/en22004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) progressively inflicts impairment of synaptic functions with notable deposition of amyloid-β (Aβ) as senile plaques within the extracellular space of the brain. Accordingly, therapeutic directions for AD have focused on clearing Aβ plaques or preventing amyloidogenesis based on the amyloid cascade hypothesis. However, the emerging evidence suggests that Aβ serves biological roles, which include suppressing microbial infections, regulating synaptic plasticity, promoting recovery after brain injury, sealing leaks in the blood-brain barrier, and possibly inhibiting the proliferation of cancer cells. More importantly, these functions were found in in vitro and in vivo investigations in a hormetic manner, that is to be neuroprotective at low concentrations and pathological at high concentrations. We herein summarize the physiological roles of monomeric Aβ and current Aβ-directed therapies in clinical trials. Based on the evidence, we propose that novel therapeutics targeting Aβ should selectively target Aβ in neurotoxic forms such as oligomers while retaining monomeric Aβ in order to preserve the physiological functions of Aβ monomers.
Collapse
Affiliation(s)
- Hyomin Jeong
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Heewon Shin
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Seungpyo Hong
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - YoungSoo Kim
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
10
|
Khonacha SE, Mirbehbahani SH, Rahdar M, Davoudi S, Borjkhani M, Khodaghli F, Motamedi F, Janahmadia M. Kisspeptin-13 prevented the electrophysiological alterations induced by Amyloid-Beta pathology in rat: Possible involvement of stromal interaction molecules and pCREB. Brain Res Bull 2022; 184:13-23. [PMID: 35272006 DOI: 10.1016/j.brainresbull.2022.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological disease that slowly causing memory impairments with no effective treatment. We have recently reported that kisspeptin-13 (KP-13) ameliorates Aβ toxicity-induced memory deficit in rats. Here, the possible cellular impact of kisspeptin receptor activation in a rat model of the early stage AD was assessed using whole-cell patch-clamp recording from CA1 pyramidal neurons and molecular approaches. Compared to neurons from the control group, cells from the Aβ-treated group displayed spontaneous and evoked hyperexcitability with lower spike frequency adaptation. These cells had also a lower sag ratio in response to hyperpolarizing prepulse current delivered before a depolarizing current injection. Neurons from the Aβ-treated group exhibited short spike onset latency, lower rheobase and short utilization time compared with those in the control group. Furthermore, phase plot analysis of action potential showed that Aβ treatment affected the action potential features. These electrophysiological changes induced by Aβ were associated with increased expression of stromal interaction molecules (STIMs), particularly (STIM2) and decreased pCREB/CREB ratio. Treatment with KP-13 following Aβ injection into the entorhinal cortex, however, prevented the excitatory effect of Aβ on spontaneous and evoked neuronal activity, increased the latency of onset, enhanced the sag ratio, increased the rheobase and utilization time, and prevented the changes induced Aβ on spike parameters. In addition, the KP-13 application after Aβ treatment reduced the expression of STIMs and increased the pCREB/CREB ratio compared to those receiving Aβ treatment alone. In summary, these results provide evidence that activation of kisspeptin receptor may be effective against pathology of Aβ.
Collapse
Affiliation(s)
- Shima Ebrahimi Khonacha
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mona Rahdar
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shima Davoudi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Borjkhani
- Department of Electrical Engineering, Urmia University of Technology, Urmia, Iran
| | - Fariba Khodaghli
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadia
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Ma C, Hong F, Yang S. Amyloidosis in Alzheimer's Disease: Pathogeny, Etiology, and Related Therapeutic Directions. Molecules 2022; 27:1210. [PMID: 35209007 PMCID: PMC8876037 DOI: 10.3390/molecules27041210] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/25/2022] [Accepted: 02/01/2022] [Indexed: 11/23/2022] Open
Abstract
The amyloid hypothesis of Alzheimer's disease has long been the predominant theory, suggesting that Alzheimer's disease is caused by the accumulation of amyloid beta protein (Aβ) in the brain, leading to neuronal toxicity in the central nervous system (CNS). Because of breakthroughs in molecular medicine, the amyloid pathway is thought to be central to the pathophysiology of Alzheimer's disease (AD). Currently, it is believed that altered biochemistry of the Aβ cycle remains a central biological feature of AD and is a promising target for treatment. This review provides an overview of the process of amyloid formation, explaining the transition from amyloid precursor protein to amyloid beta protein. Moreover, we also reveal the relationship between autophagy, cerebral blood flow, ACHE, expression of LRP1, and amyloidosis. In addition, we discuss the detailed pathogenesis of amyloidosis, including oxidative damage, tau protein, NFTs, and neuronal damage. Finally, we list some ways to treat AD in terms of decreasing the accumulation of Aβ in the brain.
Collapse
Affiliation(s)
- Chen Ma
- Experimental Center of Pathogen Biology, Nanchang University, Nanchang 330006, China;
- Queen Marry College, School of Medicine, Nanchang University, Nanchang 330036, China
| | - Fenfang Hong
- Experimental Center of Pathogen Biology, Nanchang University, Nanchang 330006, China;
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Nanchang 344099, China
| |
Collapse
|
12
|
Jin S, Wang X, Xiang X, Wu Y, Hu J, Li Y, Lin Dong Y, Tan Y, Wu X. Inhibition of GPR17 with cangrelor improves cognitive impairment and synaptic deficits induced by Aβ 1-42 through Nrf2/HO-1 and NF-κB signaling pathway in mice. Int Immunopharmacol 2021; 101:108335. [PMID: 34781121 DOI: 10.1016/j.intimp.2021.108335] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/23/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022]
Abstract
The accumulation of amyloid beta (Aβ) in the brain is thought to be associated with cognitive deficits in Alzheimer's disease (AD). However, current methods to combat Aβ neurotoxicity are still lacking. G protein-coupled receptor 17 (GPR17) has become a target for treating inflammation in brain diseases, but it is unclear whether it has a role in AD. Here, we investigated the effects of cangrelor, a GPR17 antagonist, on neurotoxicity and memory impairment induced by intracerebroventricular (i.c.v.) injection of Aβ1-42 in mice. The behavior results showed that cangrelor (2.0 or 4.0 μg/mouse, i.c.v.) treatment reversed the deficits in memory and learning ability induced by Aβ1-42 in mice. Importantly, we demonstrated for the first time that GPR17 expression in the hippocampus and frontal cortex is increased in response to Aβ1-42 exposures. We also found that cangrelor treatment reduced the activity of β-secretase 1 (BACE1) and the levels of soluble Aβ1-42 in the hippocampus and frontal cortex. Meanwhile, cangrelor treatment suppressed oxidative stress induced by Aβ1-42, as proved by reduced production of malondialdehyde (MDA), and increased glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), and promoted the expression of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1). Furthermore, cangrelor also suppressed Aβ1-42-induced neuroinflammation, characterized by suppressed activation of microglia, decreased the levels of pro-inflammatory cytokines, and nuclear translocation of NF-κB p65, as well as ameliorated synaptic deficits by promoting the upregulation of synaptic proteins, and increasing the number of Golgi-Cox stained dendritic spines. These results suggest that cangrelor may reverse Aβ1-42-induced cognition deficits via inhibiting oxidative stress, neuroinflammation, and synaptic dysfunction mediated by Nrf2/HO-1 and NF-κB signaling.
Collapse
Affiliation(s)
- ShiYu Jin
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xin Wang
- West Anhui Health Vocational College, Luan 237000, China
| | - XiaoTong Xiang
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - YuMei Wu
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Jie Hu
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - YueYue Li
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Yue Lin Dong
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - YueQiang Tan
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China
| | - Xian Wu
- School of Pharmacy, Anhui Medical University, The Key Laboratory of Major Autoimmune Diseases of Anhui Province, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
13
|
Shiratori T, Goto S, Sakaguchi T, Kasai T, Otsuka Y, Higashi K, Makino K, Takahashi H, Komatsu K. Singular value decomposition analysis of the secondary structure features contributing to the circular dichroism spectra of model proteins. Biochem Biophys Rep 2021; 28:101153. [PMID: 34712848 PMCID: PMC8528683 DOI: 10.1016/j.bbrep.2021.101153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Amyloid fibril formation occurs in restricted environment, such as the interface between intercellular fluids and bio-membranes. Conformational interconversion from α-helix to β-structure does not progress in fluids; however, it can occur after sedimentary aggregation during amyloid fibril formation induced by heat treatment of hen egg white lysozyme (HEWL). Secondary structures of various proteins and denatured proteins titrated with 2,2,2-trifluoroethanol (TFE) were examined using their CD spectra. Gaussian peak/trough and singular value decompositions (SVD) showed that the spectral pattern of the α-helix comprised a sharp trough at wavelength 207 nm and a broad trough at 220 nm. Conversely, we distinguished two patterns for β-sheet-a spread barrel type, corresponding to ConA, and a tightly weaved type, corresponding to the soybean trypsin inhibitor. Herein, we confirmed that the spectral/conformational interconversion of the heat-treated HEWL was not observed in the dissolved fluid.
Collapse
Affiliation(s)
- Tomoki Shiratori
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Satoru Goto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Tomoyo Sakaguchi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Takahiro Kasai
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Yuta Otsuka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kyohei Higashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kosho Makino
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Hideyo Takahashi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazushi Komatsu
- Department of Mathematics, Faculty of Science, Kochi University, 2-5-1 Akebonocho, Kochi, 780-8520, Japan
| |
Collapse
|
14
|
Living with the enemy: from protein-misfolding pathologies we know, to those we want to know. Ageing Res Rev 2021; 70:101391. [PMID: 34119687 DOI: 10.1016/j.arr.2021.101391] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/19/2021] [Accepted: 06/09/2021] [Indexed: 12/12/2022]
Abstract
Conformational diseases are caused by the aggregation of misfolded proteins. The risk for such pathologies develops years before clinical symptoms appear, and is higher in people with alpha-1 antitrypsin (AAT) polymorphisms. Thousands of people with alpha-1 antitrypsin deficiency (AATD) are underdiagnosed. Enemy-aggregating proteins may reside in these underdiagnosed AATD patients for many years before a pathology for AATD fully develops. In this perspective review, we hypothesize that the AAT protein could exert a new and previously unconsidered biological effect as an endogenous metal ion chelator that plays a significant role in essential metal ion homeostasis. In this respect, AAT polymorphism may cause an imbalance of metal ions, which could be correlated with the aggregation of amylin, tau, amyloid beta, and alpha synuclein proteins in type 2 diabetes mellitus (T2DM), Alzheimer's and Parkinson's diseases, respectively.
Collapse
|
15
|
Shah H, Patel A, Parikh V, Nagani A, Bhimani B, Shah U, Bambharoliya T. The β-Secretase Enzyme BACE1: A Biochemical Enigma for Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2021; 19:184-194. [PMID: 32452328 DOI: 10.2174/1871527319666200526144141] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/18/2020] [Accepted: 04/29/2020] [Indexed: 01/08/2023]
Abstract
Beta site amyloid precursor protein cleaving enzyme 1 (BACE1) is a rational target in Alzheimer's Disease (AD) drug development due to its role in amyloidogenic cleavage of Amyloid Precursor Protein (APP) in generating Amyloid β (Aβ). This β-secretase cleaves not only Amyloid Precursor Protein (APP) and its homologues, but also small series of substrates including neuregulin and β subunit of voltage-gated sodium channel that play a very important role in the development and normal function of the brain. Moreover, BACE1 is modulated at the post-translational level by several factors that are associated with both physiological and pathological functions. Since the discovery of BACE1 over a decade ago, medicinal chemistry and pharmacokinetics of BACE1 small molecule inhibitors have proven challenging for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Hirak Shah
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat 391760, India
| | - Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| | - Vruti Parikh
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat 391760, India
| | - Afzal Nagani
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat 391760, India
| | - Bhargav Bhimani
- Piramal Discovery Solution, Pharmaceutical Special Economic Zone, Ahmedabad 382213, India
| | - Umang Shah
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat 388421, India
| | - Tushar Bambharoliya
- Pharmaceutical Polymer Technology, North Carolina State University, North Carolina, NC, United States
| |
Collapse
|
16
|
Mzezewa SC, Omoruyi SI, Zondagh LS, Malan SF, Ekpo OE, Joubert J. Design, synthesis, and evaluation of 3,7-substituted coumarin derivatives as multifunctional Alzheimer's disease agents. J Enzyme Inhib Med Chem 2021; 36:1607-1621. [PMID: 34281458 PMCID: PMC8291583 DOI: 10.1080/14756366.2021.1913137] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Multitarget directed ligands (MTDLs) are emerging as promising treatment options for Alzheimer's disease (AD). Coumarin derivatives serve as a good starting point for designing MTDLs due to their inherent inhibition of monoamine oxidase (MAO) and cholinesterase enzymes, which are complicit in AD's complex pathophysiology. A preliminary series of 3,7-substituted coumarin derivatives were synthesised and evaluated for enzyme inhibitory activity, cytotoxicity as well as neuroprotective ability. The results indicated that the compounds are weak cholinesterase inhibitors with five compounds demonstrating relatively potent inhibition and selectivity towards MAO-B with IC50 values between 0.014 and 0.498 hx00B5;µM. Significant neuroprotective effects towards MPP+-compromised SH-SY5Y neuroblastoma cells were also observed, with no inherent cytotoxicity at 10 µM for all compounds. The overall results demonstrated that substitution of the phenylethyloxy moiety at the 7-position imparted superior general activity to the derivatives, with the propargylamine substitution at the 3-position, in particular, displaying the best MAO-B selectivity and neuroprotection.
Collapse
Affiliation(s)
- Sheunopa C Mzezewa
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Sylvester I Omoruyi
- Department of Medical Biosciences, University of the Western Cape, Bellville, South Africa
| | - Luke S Zondagh
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Sarel F Malan
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| | - Okobi E Ekpo
- Department of Medical Biosciences, University of the Western Cape, Bellville, South Africa
| | - Jacques Joubert
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
17
|
Liu Q, Xi Y, Wang Q, Liu J, Li P, Meng X, Liu K, Chen W, Liu X, Liu Z. Mannan oligosaccharide attenuates cognitive and behavioral disorders in the 5xFAD Alzheimer's disease mouse model via regulating the gut microbiota-brain axis. Brain Behav Immun 2021; 95:330-343. [PMID: 33839232 DOI: 10.1016/j.bbi.2021.04.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/18/2021] [Accepted: 04/07/2021] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive deficits and psychiatric symptoms. The gut microbiota-brain axis plays a pivotal role during AD development, which could target nutritional intervention. The prebiotic mannan oligosaccharide (MOS) has been reported to reshape the gut microbiome and enhanced the formation of the neuroprotective metabolites short-chain fatty acids (SCFAs). Here, we found that an 8-week treatment of MOS (0.12%, w/v in the drinking water) significantly improved cognitive function and spatial memory, accompanied by attenuated the anxiety- and obsessive-like behaviors in the 5xFAD transgenic AD mice model. MOS substantially reduced the Aβ accumulation in the cortex, hippocampus, and amygdala of the brain. Importantly, MOS treatment significantly balanced the brain redox status and suppressed the neuroinflammatory responses. Moreover, MOS also alleviated the HPA-axis disorders by decreasing the levels of hormones corticosterone (CORT) and corticotropin-releasing hormone (CRH) and upregulated the norepinephrine (NE) expressions. Notably, the gut barrier integrity damage and the LPS leak were prevented by the MOS treatment. MOS re-constructed the gut microbiota composition, including increasing the relative abundance of Lactobacillus and reducing the relative abundance of Helicobacter. MOS enhanced the butyrate formation and related microbes levels. The correlation analysis indicated that the reshaped gut microbiome and enhanced butyrate formation are highly associated with behavioral alteration and brain oxidative status. SCFAs supplementation experiment also attenuated the behavioral disorders and Aβ accumulation in the AD mice brain, accompanied by balanced HPA-axis and redox status. In conclusion, the present study indicated that MOS significantly attenuates the cognitive and mental deficits in the 5xFAD mice, which could be partly explained by the reshaped microbiome and enhanced SCFAs formation in the gut. MOS, as a prebiotics, can be translated into a novel microbiota-targeted approach for managing metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Qing Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Yujia Xi
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Qianxu Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinhui Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Peiran Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xue Meng
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Kai Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Weixuan Chen
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China; Department of Food Science, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
18
|
Brunori M. From Kuru to Alzheimer: A personal outlook. Protein Sci 2021; 30:1776-1792. [PMID: 34118168 DOI: 10.1002/pro.4145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 01/02/2023]
Abstract
Seventy years ago, we learned from Chris Anfinsen that the stereochemical code necessary to fold a protein is embedded into its amino acid sequence. In water, protein morphogenesis is a spontaneous reversible process leading from an ensemble of disordered structures to the ordered functionally competent protein; conforming to Aristotle's definition of substance, the synolon of matter and form. The overall process of folding is generally consistent with a two state transition between the native and the denatured protein: not only the denatured state is an ensemble of several structures, but also the native protein populates distinct functionally relevant conformational (sub)states. This two-state view should be revised, given that any globular protein can populate a peculiar third state called amyloid, characterized by an overall architecture that at variance with the native state, is by-and-large independent of the primary structure. In a nut shell, we should accept that beside the folded and unfolded states, any protein can populate a third state called amyloid which gained center stage being the hallmark of incurable neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases as well as others. These fatal diseases are characterized by clear-cut clinical differences, yet display some commonalities such as the presence in the brain of amyloid deposits constituted by one misfolded protein specific for each disease. Some aspects of this complex problem are summarized here as an excursus from the prion's fibrils observed in the brain of aborigines who died of Kuru to the amyloid detectable in the cortex of Alzheimer's patients.
Collapse
Affiliation(s)
- Maurizio Brunori
- Accademia Nazionale dei Lincei and Dipartimento di Scienze Biochimiche "A. Rossi Fanelli,", Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
19
|
Libard S, Walter J, Alafuzoff I. In vivo Characterization of Biochemical Variants of Amyloid-β in Subjects with Idiopathic Normal Pressure Hydrocephalus and Alzheimer's Disease Neuropathological Change. J Alzheimers Dis 2021; 80:1003-1012. [PMID: 33612546 PMCID: PMC8150506 DOI: 10.3233/jad-201469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Stepwise occurrence of biochemically modified amyloid-β (Aβ) in the brain of subjects with Alzheimer’s disease (AD) has been suggested to be of significance for cognitive impairment. Our previous reports have shown that Aβ is observed in 63% of all subjects with idiopathic normal pressure hydrocephalus (iNPH) suggesting that the majority of iNPH subjects with Aβ are indeed also suffering from AD. Objective: We assessed the occurrence of biochemically modified Aβ variants, in vivo, in subjects with iNPH and in a cohort of postmortem brain samples from patients with dementia. Methods: We assessed Aβ proteins in 127 diagnostic brain biopsies obtained from subjects with iNPH and in a cohort of subjects with dementia by means of immunohistochemistry. Results: The pyroglutamylated Aβ (pyAβ) precedes the aggregation of phosphorylated Aβ (pAβ) during the AD neuropathological change progression; moreover, these modified variants of Aβ correlate with hyperphosphorylated tau in the frontal cortical area of human brain. Our results confirm the existence of the suggested biochemical stages of Aβ aggregation that might be of significance for neurodegeneration leading to cognitive impairment. Conclusion: The observation that both pyAβ and pAβ are seen in vivo in iNPH subjects is intriguing. It has been reported that most of the iNPH subjects with Aβ in the brain biopsy indeed develop AD with time. Based on our current and previous results, it is clinically merited to obtain a diagnostic biopsy from a subject with iNPH. When Aβ is observed in the biopsy, the biochemical characterization is of interest.
Collapse
Affiliation(s)
- Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Irina Alafuzoff
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
20
|
Liu C, Luo X. Potential molecular and graphene oxide chelators to dissolve amyloid-β plaques in Alzheimer's disease: a density functional theory study. J Mater Chem B 2021; 9:2736-2746. [PMID: 33688880 DOI: 10.1039/d0tb02985h] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The onset of Alzheimer's disease (AD) is caused by amyloid-β (Aβ) aggregation. Elevated levels of metals, specifically copper, zinc, iron, and aluminum, accumulate in senile Aβ; plaque deposits, disrupting normal brain homeostasis and cognitive functions. In this investigation, we studied the potential of several molecular and graphene oxide chelators to be used for future AD research and chelation therapy. To understand the interactions between selected metals (Cu, Zn, Fe, and Al), the Aβ peptide, and various potential metal chelating compounds, we implemented the density functional theory (DFT) method to calculate the binding energies of each metal-molecule complex. The binding energy of each metal-chelator complex was compared with that of the metal-Aβ compound to determine the chelation potential of the selected chelator. The potential chelating agents studied were 8-hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazone (INNHQ), 8-hydroxyquinoline-2-carboxaldehyde 2-furoyl hydrazone (HQFUH), quercetin, and graphene oxide (GO). Our calculated binding energies revealed that the HQFUH molecule holds direct ability to chelate copper, zinc, iron, and aluminum. In addition, the GO complex with a 12.5% oxygen concentration demonstrates aluminum chelation ability. Our results demonstrate that HQFUH and GO can be used in future AD drug development research and therapy to target toxic metal-Aβ interactions and reduce Aβ aggregation.
Collapse
Affiliation(s)
- Christina Liu
- National Graphene Research and Development Center, Springfield, Virginia 22151, USA.
| | | |
Collapse
|
21
|
Kim D, Lee JH, Kim HY, Shin J, Kim K, Lee S, Park J, Kim J, Kim Y. Fluorescent indolizine derivative YI-13 detects amyloid-β monomers, dimers, and plaques in the brain of 5XFAD Alzheimer transgenic mouse model. PLoS One 2020; 15:e0243041. [PMID: 33362250 PMCID: PMC7757811 DOI: 10.1371/journal.pone.0243041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/15/2020] [Indexed: 01/02/2023] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized by the aberrant production and accumulation of amyloid-β (Aβ) peptides in the brain. Accumulated Aβ in soluble oligomer and insoluble plaque forms are considered to be a pathological culprit and biomarker of the disorder. Here, we report a fluorescent universal Aβ-indicator YI-13, 5-(4-fluorobenzoyl)-7,8-dihydropyrrolo[1,2-b]isoquinolin-9(6H)-one, which detects Aβ monomers, dimers, and plaques. We synthesized a library of 26 fluorescence chemicals with the indolizine core and screen them through a series of in vitro tests utilizing Aβ as a target and YI-13 was selected as the final imaging candidate. YI-13 was found to stain and visualize insoluble Aβ plaques in the brain tissue, of a transgenic mouse model with five familial AD mutations (5XFAD), by a histochemical approach and to label soluble Aβ oligomers within brain lysates of the mouse model under a fluorescence plate reader. Among oligomers aggregated from monomers and synthetic dimers from chemically conjugated monomers, YI-13 preferred the dimeric Aβ.
Collapse
Affiliation(s)
- DaWon Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Jeong Hwa Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Jisu Shin
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Kyeonghwan Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Sejin Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | | | - JinIkyon Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
- * E-mail: (JK); (YK)
| | - YoungSoo Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
- * E-mail: (JK); (YK)
| |
Collapse
|
22
|
Lee EY, Srinivasan Y, de Anda J, Nicastro LK, Tükel Ç, Wong GCL. Functional Reciprocity of Amyloids and Antimicrobial Peptides: Rethinking the Role of Supramolecular Assembly in Host Defense, Immune Activation, and Inflammation. Front Immunol 2020; 11:1629. [PMID: 32849553 PMCID: PMC7412598 DOI: 10.3389/fimmu.2020.01629] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
Pathological self-assembly is a concept that is classically associated with amyloids, such as amyloid-β (Aβ) in Alzheimer's disease and α-synuclein in Parkinson's disease. In prokaryotic organisms, amyloids are assembled extracellularly in a similar fashion to human amyloids. Pathogenicity of amyloids is attributed to their ability to transform into several distinct structural states that reflect their downstream biological consequences. While the oligomeric forms of amyloids are thought to be responsible for their cytotoxicity via membrane permeation, their fibrillar conformations are known to interact with the innate immune system to induce inflammation. Furthermore, both eukaryotic and prokaryotic amyloids can self-assemble into molecular chaperones to bind nucleic acids, enabling amplification of Toll-like receptor (TLR) signaling. Recent work has shown that antimicrobial peptides (AMPs) follow a strikingly similar paradigm. Previously, AMPs were thought of as peptides with the primary function of permeating microbial membranes. Consistent with this, many AMPs are facially amphiphilic and can facilitate membrane remodeling processes such as pore formation and fusion. We show that various AMPs and chemokines can also chaperone and organize immune ligands into amyloid-like ordered supramolecular structures that are geometrically optimized for binding to TLRs, thereby amplifying immune signaling. The ability of amphiphilic AMPs to self-assemble cooperatively into superhelical protofibrils that form structural scaffolds for the ordered presentation of immune ligands like DNA and dsRNA is central to inflammation. It is interesting to explore the notion that the assembly of AMP protofibrils may be analogous to that of amyloid aggregates. Coming full circle, recent work has suggested that Aβ and other amyloids also have AMP-like antimicrobial functions. The emerging perspective is one in which assembly affords a more finely calibrated system of recognition and response: the detection of single immune ligands, immune ligands bound to AMPs, and immune ligands spatially organized to varying degrees by AMPs, result in different immunologic outcomes. In this framework, not all ordered structures generated during multi-stepped AMP (or amyloid) assembly are pathological in origin. Supramolecular structures formed during this process serve as signatures to the innate immune system to orchestrate immune amplification in a proportional, situation-dependent manner.
Collapse
Affiliation(s)
- Ernest Y Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yashes Srinivasan
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jaime de Anda
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lauren K Nicastro
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Çagla Tükel
- Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, United States.,California Nano Systems Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
23
|
Enhancing α-secretase Processing for Alzheimer's Disease-A View on SFRP1. Brain Sci 2020; 10:brainsci10020122. [PMID: 32098349 PMCID: PMC7071437 DOI: 10.3390/brainsci10020122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022] Open
Abstract
Amyloid β (Aβ) peptides generated via sequential β- and γ-secretase processing of the amyloid precursor protein (APP) are major etiopathological agents of Alzheimer's disease (AD). However, an initial APP cleavage by an α-secretase, such as the a disintegrin and metalloproteinase domain-containing protein ADAM10, precludes β-secretase cleavage and leads to APP processing that does not produce Aβ. The latter appears to underlie the disease symptom-attenuating effects of a multitude of experimental therapeutics in AD animal models. Recent work has indicated that an endogenous inhibitor of ADAM10, secreted-frizzled-related protein 1 (SFRP1), is elevated in human AD brains and associated with amyloid plaques in mouse AD models. Importantly, genetic or functional attenuation of SFRP1 lowered Aβ accumulation and improved AD-related histopathological and neurological traits. Given SFRP1's well-known activity in attenuating Wnt signaling, which is also commonly impaired in AD, SFRP1 appears to be a promising therapeutic target for AD. This idea, however, needs to be addressed with care because of cancer enhancement potentials resulting from a systemic loss of SFRP1 activity, as well as an upregulation of ADAM10 activity. In this focused review, I shall discuss α-secretase-effected APP processing in AD with a focus on SFRP1, and explore the contrasting perspectives arising from the recent findings.
Collapse
|
24
|
Hameed S, Fuh JL, Senanarong V, Ebenezer EGM, Looi I, Dominguez JC, Park KW, Karanam AK, Simon O. Role of Fluid Biomarkers and PET Imaging in Early Diagnosis and its Clinical Implication in the Management of Alzheimer's Disease. J Alzheimers Dis Rep 2020; 4:21-37. [PMID: 32206755 PMCID: PMC7081089 DOI: 10.3233/adr-190143] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical diagnosis of Alzheimer's disease (AD) is based on symptoms; however, the challenge is to diagnose AD at the preclinical stage with the application of biomarkers and initiate early treatment (still not widely available). Currently, cerebrospinal fluid (CSF) amyloid-β 42 (Aβ42) and tau are used in the clinical diagnosis of AD; nevertheless, blood biomarkers (Aβ42 and tau) are less predictive. Amyloid-positron emission tomography (PET) imaging is an advancement in technology that uses approved radioactive diagnostic agents (florbetapir, flutemetamol, or florbetaben) to estimate Aβ neuritic plaque density in adults with cognitive impairment evaluated for AD and other causes of cognitive decline. There is no cure for AD to date-the disease progression cannot be stopped or reversed; approved pharmacological agents (donepezil, galantamine, and rivastigmine; memantine) provide symptomatic treatment. However, the disease-modifying therapies are promising; aducanumab and CAD106 are in phase III trials for the early stages of AD. In conclusion, core CSF biomarkers reflect pathophysiology of AD in the early and late stages; the application of approved radiotracers have potential in amyloid-PET brain imaging to detect early AD.
Collapse
Affiliation(s)
- Shahul Hameed
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
- Duke NUS Medical School, Singapore
| | - Jong-Ling Fuh
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan
| | - Vorapun Senanarong
- Division of Neurology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Irene Looi
- Clinical Research Centre, Hospital Seberang Jaya, Penang, Malaysia
- Department of Medicine, Hospital Seberang Jaya, Penang, Malaysia
| | | | - Kyung Won Park
- Department of Neurology and Cognitive Disorders and Dementia Center, Institute of Convergence Bio-Health, Dong-A University College of Medicine, Busan, Republic of Korea
| | | | - Oliver Simon
- Novartis (Singapore) Pte. Ltd., Singapore, Singapore
| |
Collapse
|
25
|
Rashid MH, Zahid MF, Zain S, Kabir A, Hassan SU. The Neuroprotective Effects of Exercise on Cognitive Decline: A Preventive Approach to Alzheimer Disease. Cureus 2020; 12:e6958. [PMID: 32190507 PMCID: PMC7067577 DOI: 10.7759/cureus.6958] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive disorder that causes brain cells to slowly degenerate and die. This leads to a continuous decline in thinking, behavioral and social skills that disrupts a person's ability to function independently. AD is the most common cause of dementia globally. Neuroinflammation caused by intracellular neurofibrillary tangles and extracellular amyloid deposits leads to atrophy of brain cells especially the hippocampus, which is associated with memory formation. This atrophy leads to dementia and cognitive decline. Among the many preventive factors being studied, exercise is thought to play a vital role in not only preventing the pre-clinical stage of AD but also slowing the clinical progression of AD. It is also deployed as a treatment option for late-stage AD along with pharmacological treatment options. Various studies and clinical trials in both human and animal models are of the opinion that exercise slows the onset and progression of cognitive decline in AD patients. Some studies suggest that this effect is due to a decrease in neurofibrillary tangles and amyloid deposits in brain parenchyma. Others suggest that exercise causes an increase in angiogenesis, neurogenesis, and synaptogenesis mainly due to an increase in blood flow, brain-derived neurotrophic factor (BDNF), insulin-like growth factor 1 (IGF-1), hormones, and second messengers.
Collapse
Affiliation(s)
| | | | - Sarmad Zain
- Internal Medicine, Nishtar Hospital, Nishtar Medical University, Multan, PAK
| | - Ahmad Kabir
- Pathology, Bakhtawar Amin Medical and Dental College, Multan, PAK
| | - Sibt Ul Hassan
- Internal Medicine, Bakhtawar Amin Memorial Hospital, Multan, PAK
| |
Collapse
|
26
|
Active-site environment of Cu bound amyloid β and amylin peptides. J Biol Inorg Chem 2019; 24:1245-1259. [DOI: 10.1007/s00775-019-01724-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/05/2019] [Indexed: 01/09/2023]
|
27
|
Pal I, Nath AK, Roy M, Seal M, Ghosh C, Dey A, Dey SG. Formation of compound I in heme bound Aβ-peptides relevant to Alzheimer's disease. Chem Sci 2019; 10:8405-8410. [PMID: 31803419 PMCID: PMC6844219 DOI: 10.1039/c9sc01679a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/22/2019] [Indexed: 12/02/2022] Open
Abstract
Proteolysis of Amyloid Precursor Protein, APP, results in the formation of amyloid β (Aβ) peptides, which have been associated with Alzheimer's disease (AD). Recently the failure of therapeutic agents that prohibit Aβ aggregation and sequester Cu/Zn in providing symptomatic relief to AD patients has questioned the amyloid and metal ion hypothesis. Alternatively, abnormal heme homeostasis and reduced levels of neurotransmitters in the brain are hallmark features of AD. Heme can bind Aβ peptides forming a peroxidase type active site which can oxidatively degrade neurotransmitters like serotonin. To date the reactive species responsible for this activity has not been identified. Using rapid kinetics and freeze quenching, we show that heme bound Aβ forms a highly reactive intermediate, compound I. Thus, compound I provides a basis for elucidating the oxidative degradation of neurotransmitters like serotonin, resulting in abnormal neurotransmission, a key pathological feature of AD. Site directed mutants indicate that the Arg5 and Tyr10 residues, unique to human Aβ, affect the rates of formation and decay of compound I providing insight into their roles in the oxidative degradation of neurotransmitters. Tyr10 can potentially play a natural protective role against the highly reactive oxidant, compound I, in AD.
Collapse
Affiliation(s)
- Ishita Pal
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Arnab Kumar Nath
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Madhuparna Roy
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Manas Seal
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Chandradeep Ghosh
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Abhishek Dey
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| | - Somdatta Ghosh Dey
- Indian Association for the Cultivation of Science , 2A & 2B, Raja S. C. Mullick Road, Jadavpur , Kolkata 700032 , India .
| |
Collapse
|
28
|
Serum amyloid A1 is involved in amyloid plaque aggregation and memory decline in amyloid beta abundant condition. Transgenic Res 2019; 28:499-508. [PMID: 31407125 DOI: 10.1007/s11248-019-00166-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 08/02/2019] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by cognitive impairment, progressive neurodegeneration, and amyloid-β (Aβ) lesion. In the neuronal death and disease progression, inflammation is known to play an important role. Our previous study on acute-phase protein serum amyloid A1 (SAA1) overexpressed mice showed that the liver-derived SAA1 accumulated in the brain by crossing the brain blood barrier (BBB) and trigger the depressive-like behavior on mouse. Since SAA1 involved in immune responses in other diseases, we focused on the possibility that SAA1 may exacerbate the neuronal inflammation related to Alzheimer's disease. A APP/SAA overexpressed double transgenic mouse was generated using amyloid precursor protein overexpressed (APP)-c105 mice and SAA1 overexpressed mice to examine the function of SAA1 in Aβ abundant condition. Comparisons between APP and APP/SAA1 transgenic mice showed that SAA1 exacerbated amyloid aggregation and glial activation; which lead to the memory decline. Behavior tests also supported this result. Overall, overexpression of SAA1 intensified the neuronal inflammation in amyloid abundant condition and causes the greater memory decline compared to APP mice, which only expresses Aβ 1-42.
Collapse
|
29
|
Sanagavarapu K, Nüske E, Nasir I, Meisl G, Immink JN, Sormanni P, Vendruscolo M, Knowles TPJ, Malmendal A, Cabaleiro-Lago C, Linse S. A method of predicting the in vitro fibril formation propensity of Aβ40 mutants based on their inclusion body levels in E. coli. Sci Rep 2019; 9:3680. [PMID: 30842594 PMCID: PMC6403284 DOI: 10.1038/s41598-019-39216-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Overexpression of recombinant proteins in bacteria may lead to their aggregation and deposition in inclusion bodies. Since the conformational properties of proteins in inclusion bodies exhibit many of the characteristics typical of amyloid fibrils. Based on these findings, we hypothesize that the rate at which proteins form amyloid fibrils may be predicted from their propensity to form inclusion bodies. To establish a method based on this concept, we first measured by SDS-PAGE and confocal microscopy the level of inclusion bodies in E. coli cells overexpressing the 40-residue amyloid-beta peptide, Aβ40, wild-type and 24 charge mutants. We then compared these results with a number of existing computational aggregation propensity predictors as well as the rates of aggregation measured in vitro for selected mutants. Our results show a strong correlation between the level of inclusion body formation and aggregation propensity, thus demonstrating the power of this approach and its value in identifying factors modulating aggregation kinetics.
Collapse
Affiliation(s)
- Kalyani Sanagavarapu
- Lund University, Biochemistry and Structural Biology, Chemical Center, Lund, Sweden.
| | | | - Irem Nasir
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, 10550 N, Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Georg Meisl
- University of Cambridge, Chemistry Department, Lensfield Road, Cambridge, UK
| | - Jasper N Immink
- Lund University, Physical Chemistry, Chemical Center, Lund, Sweden
| | - Pietro Sormanni
- University of Cambridge, Chemistry Department, Lensfield Road, Cambridge, UK
| | - Michele Vendruscolo
- University of Cambridge, Chemistry Department, Lensfield Road, Cambridge, UK
| | - Tuomas P J Knowles
- University of Cambridge, Chemistry Department, Lensfield Road, Cambridge, UK.,Cavendish Laboratory, Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, UK
| | - Anders Malmendal
- Lund University, Biochemistry and Structural Biology, Chemical Center, Lund, Sweden
| | - Celia Cabaleiro-Lago
- Lund University, Biochemistry and Structural Biology, Chemical Center, Lund, Sweden.,Faculty of natural sciences, Kristianstad University, Kristianstad, Sweden
| | - Sara Linse
- Lund University, Biochemistry and Structural Biology, Chemical Center, Lund, Sweden.
| |
Collapse
|
30
|
Abstract
Alzheimer's disease (AD), the most common cause of age-dependent dementia, is one of the most significant healthcare problems worldwide. Aggravating this situation, drugs that are currently US Food and Drug Administration (FDA)-approved for AD treatment do not prevent or delay disease progression. Therefore, developing effective therapies for AD patients is of critical urgency. Human genetic and clinical studies over the past three decades have indicated that abnormal generation or accumulation of amyloid-β (Aβ) peptides is a likely culprit in AD pathogenesis. Aβ is generated from amyloid precursor protein (APP) via proteolytic cleavage by β-site APP cleaving enzyme 1 (BACE1) (memapsin 2, β-secretase, Asp 2 protease) and γ-secretase. Mice deficient in BACE1 show abrogated production of Aβ. Therefore, pharmacological inhibition of BACE1 is being intensively pursued as a therapeutic approach to treat AD patients. Recent setbacks in clinical trials with BACE1 inhibitors have highlighted the critical importance of understanding how to properly inhibit BACE1 to treat AD patients. This review summarizes the recent studies on the role of BACE1 in synaptic functions as well as our views on BACE1 inhibition as an effective AD treatment.
Collapse
Affiliation(s)
- Brati Das
- Department of Neuroscience, Room E4032, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Riqiang Yan
- Department of Neuroscience, Room E4032, UConn Health, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| |
Collapse
|
31
|
Derf A, Mudududdla R, Bharate SB, Chaudhuri B. Inhibitors of Aβ42-induced endoplasmic reticular unfolded protein response (UPR ER), in yeast, also rescue yeast cells from Aβ42-mediated apoptosis. Eur J Pharm Sci 2019; 128:118-127. [PMID: 30502452 DOI: 10.1016/j.ejps.2018.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/10/2018] [Accepted: 11/25/2018] [Indexed: 11/19/2022]
Abstract
Aggregated Aβ peptides which cause amyloid deposits, a characteristic of Alzheimer's disease (AD), activate a stress response in the endoplasmic reticulum (ER), known as the unfolded protein response, UPRER. Nascent UPRER induction helps in reducing ER stress by eliminating accumulated misfolded/aggregated secretory proteins. However, prolonged UPRER induction may trigger apoptosis. Here we show that, when expressed in yeast with an NH2-terminal secretory signal sequence (ss), the 42-amino acid human Aβ42 (h_Aβ42), but not the mouse/ratAβ42 (m_Aβ42) which reportedly does not misfold/aggregate, induces UPRER as monitored via an eGFP reporter. We also show that expression of ss-h_Aβ42, not ss-m_Aβ42, blocks yeast cell growth, with cells expressing ss-h_Aβ42 manifesting distinctive features of apoptosis such as loss of mitochondrial membrane potential, increase in ROS levels and DNA fragmentation. Screening for suppressors of ss-h_Aβ42-activated UPRER-eGFP induction, in a computationally-designed 29-compound methoxy-stilbene library, revealed three compounds that reduce >95% of UPRER-eGFP induction at 5 μM concentration, with EC50 values of 40-50 nM. Surprisingly, the compounds also rescue yeast cells from ss-h_Aβ42-mediated apoptosis, with EC50-s of 50-60 nM. These results provide direct evidence, probably for the first time, that there is a direct correlation between deactivation of UPRER and attenuation of apoptosis.
Collapse
Affiliation(s)
- Asma Derf
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| | - Ramesh Mudududdla
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Sandip B Bharate
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific & Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Bhabatosh Chaudhuri
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK.
| |
Collapse
|
32
|
Libard S, Laurell K, Cesarini KG, Alafuzoff I. Progression of Alzheimer's Disease-Related Pathology and Cell Counts in a Patient with Idiopathic Normal Pressure Hydrocephalus. J Alzheimers Dis 2019; 61:1451-1462. [PMID: 29376849 DOI: 10.3233/jad-170446] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We had an opportunity to assess the change observed in the brain regarding Alzheimer's disease (AD)-related alterations, cell count, and inflammation that took place during a period of 21 months in a subject with a definite diagnosis of AD and idiopathic Normal Pressure Hydrocephalus (iNPH). Four neuronal markers, i.e., synaptophysin, microtubule associated protein 2, non-phosphorylated neurofilament H (SMI32), and embryonic lethal abnormal visual system proteins 3/4 HuC/HuD (HuC/HuD); three microglial markers CD68, Human Leucocytic Antigen DR, ionized calcium-binding adaptor molecule 1, glial fibrillary acidic protein (GFAP); and AD-related markers, hyperphosphorylated τ (HPτ) and amyloid-β (Aβ, Aβ40, Aβ42) were assessed. Morphometrically assessed immunoreactivity of all neuronal and all microglial markers and Aβ42 decreased parallel with an increase in the HPτ in the frontal cortex. The expression of GFAP was stable with time. The first sample was obtained during the therapeutic shunting procedure for iNPH, and the second sample was obtained postmortem. Negligible reactive changes were observed surrounding the shunt channel. In conclusion, in the late stage of AD with time, a neuronal loss, increase in the HPτ, and decrease in Aβ42 and microglia was observed, whereas the expression of GFAP was rather stable. The observations described here suggest that when a brain biopsy has been obtained from an adult subject with iNPH, the assessment of postmortem brain is of major significance.
Collapse
Affiliation(s)
- Sylwia Libard
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden.,Department of Pathology, Uppsala University Hospital, Sweden
| | - Katarina Laurell
- Department of Pharmacology and Clinical Neuroscience, Östersund, Umeå University, Sweden
| | | | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden.,Department of Pathology, Uppsala University Hospital, Sweden
| |
Collapse
|
33
|
Bachmann MF, Jennings GT, Vogel M. A vaccine against Alzheimer`s disease: anything left but faith? Expert Opin Biol Ther 2018; 19:73-78. [DOI: 10.1080/14712598.2019.1554646] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Martin F. Bachmann
- RIA Immunology, Department of BioMedical Research, University of Bern, Bern,
Switzerland
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, University of Oxford,
Oxford, UK
| | - Gary T Jennings
- Hypopet AG, Zürich,
Switzerland
- Saiba GmbH, Pfäffikon,
Switzerland
| | - Monique Vogel
- RIA Immunology, Department of BioMedical Research, University of Bern, Bern,
Switzerland
| |
Collapse
|
34
|
Lee SY, Chiu YJ, Yang SM, Chen CM, Huang CC, Lee-Chen GJ, Lin W, Chang KH. Novel synthetic chalcone-coumarin hybrid for Aβ aggregation reduction, antioxidation, and neuroprotection. CNS Neurosci Ther 2018; 24:1286-1298. [PMID: 30596401 DOI: 10.1111/cns.13058] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/11/2018] [Accepted: 08/11/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Aggregation of misfolded amyloid β (Aβ) in senile plaques causes oxidative stress and neuronal death in Alzheimer's disease (AD). Compounds possessing antiaggregation and antioxidant properties are promising candidate compounds for AD treatment. METHODS We examined the potential of synthetic derivatives of licochalcone A and coumarin for inhibiting Aβ aggregation, scavenging reactive oxygen species (ROS), and providing neuroprotection by using biochemical assays and Tet-On Aβ-GFP 293/SH-SY5Y cell models for AD. RESULTS Among test compounds, LM-031, a novel chalcone-coumarin hybrid, inhibited Aβ aggregation and scavenged free oxygen radicals. LM-031 markedly reduced Aβ misfolding and ROS as well as promoted neurite outgrowth and inhibited acetylcholinesterase in Tet-On Aβ-GFP 293/SH-SY5Y cells. Mechanistic studies showed upregulation of the HSPB1 chaperone, NRF2/NQO1/GCLC pathway, and CREB/BDNF/BCL2 pathway. Decreased neurite outgrowth upon the induction of Aβ-GFP was rescued by LM-031, which was counteracted by knockdown of HSPB1, NRF2, or CREB. CONCLUSION Taken together, these findings demonstrate that LM-031 exhibited antiaggregation, antioxidant, and neuroprotective effects against Aβ toxicity by enhancing HSPB1 and the NRF2-related antioxidant pathway as well as by activating the CREB-dependent survival and antiapoptosis pathway. These results imply that LM-031 may be a new therapeutic compound for AD.
Collapse
Affiliation(s)
- Shin-Ying Lee
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Shu-Mei Yang
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chin-Chang Huang
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Wenwei Lin
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital-Linkou Medical Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
35
|
Arendt T, Stieler JT, Holzer M. Tau and tauopathies. Brain Res Bull 2016; 126:238-292. [PMID: 27615390 DOI: 10.1016/j.brainresbull.2016.08.018] [Citation(s) in RCA: 422] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022]
|
36
|
Brunori M, Gianni S. Molecular medicine - To be or not to be. Biophys Chem 2016; 214-215:33-46. [PMID: 27214761 DOI: 10.1016/j.bpc.2016.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 12/17/2022]
Abstract
Molecular medicine is founded on the synergy between Chemistry, Physics, Biology and Medicine, with the ambitious goal of tackling diseases from a molecular perspective. This Review aims at retracing a personal outlook of the birth and development of molecular medicine, as well as at highlighting some of the most urgent challenges linked to aging and represented by incurable neurodegenerative diseases caused by protein misfolding. Furthermore, we emphasize the emerging role of the retromer dysfunctions and improper protein sorting in Alzheimer's disease and other important neurological disordered.
Collapse
Affiliation(s)
- Maurizio Brunori
- Istituto Pasteur - Fondazione Cenci Bolognetti and Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, 00185 Rome, Italy.
| | - Stefano Gianni
- Istituto Pasteur - Fondazione Cenci Bolognetti and Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Università di Roma, 00185 Rome, Italy.
| |
Collapse
|
37
|
Zamolodchikov D, Renné T, Strickland S. The Alzheimer's disease peptide β-amyloid promotes thrombin generation through activation of coagulation factor XII. J Thromb Haemost 2016; 14:995-1007. [PMID: 26613657 PMCID: PMC4870142 DOI: 10.1111/jth.13209] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 11/12/2015] [Indexed: 11/30/2022]
Abstract
UNLABELLED Essentials How the Alzheimer's disease (AD) peptide β-amyloid (Aβ) disrupts neuronal function in the disease is unclear. Factor (F) XII initiates blood clotting via FXI, and thrombosis has been implicated in AD. Aβ triggers FXII-dependent FXI and thrombin activation, evidence of which is seen in AD plasma. Aβ-triggered clotting could contribute to neuronal dysfunction in AD and be a novel therapeutic target. SUMMARY Background β-Amyloid (Aβ) is a key pathologic element in Alzheimer's disease (AD), but the mechanisms by which it disrupts neuronal function in vivo are not completely understood. AD is characterized by a prothrombotic state, which could contribute to neuronal dysfunction by affecting cerebral blood flow and inducing inflammation. The plasma protein factor XII triggers clot formation via the intrinsic coagulation cascade, and has been implicated in thrombosis. Objectives To investigate the potential for Aβ to contribute to a prothrombotic state. Methods and results We show that Aβ activates FXII, resulting in FXI activation and thrombin generation in human plasma, thereby establishing Aβ as a possible driver of prothrombotic states. We provide evidence for this process in AD by demonstrating decreased levels of FXI and its inhibitor C1 esterase inhibitor in AD patient plasma, suggesting chronic activation, inhibition and clearance of FXI in AD. Activation of the intrinsic coagulation pathway in AD is further supported by elevated fibrin levels in AD patient plasma. Conclusions The ability of Aβ to promote coagulation via the FXII-driven contact system identifies new mechanisms by which it could contribute to neuronal dysfunction and suggests potential new therapeutic targets in AD.
Collapse
Affiliation(s)
- Daria Zamolodchikov
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Renné
- Division of Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm S-171 76, Sweden
- Center of Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
38
|
Majd S, Power JH, Grantham HJM. Neuronal response in Alzheimer's and Parkinson's disease: the effect of toxic proteins on intracellular pathways. BMC Neurosci 2015; 16:69. [PMID: 26499115 PMCID: PMC4619058 DOI: 10.1186/s12868-015-0211-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 10/13/2015] [Indexed: 01/09/2023] Open
Abstract
Accumulation of protein aggregates is the leading cause of cellular dysfunction in neurodegenerative disorders. Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease, Prion disease and motor disorders such as amyotrophic lateral sclerosis, present with a similar pattern of progressive neuronal death, nervous system deterioration and cognitive impairment. The common characteristic is an unusual misfolding of proteins which is believed to cause protein deposition and trigger degenerative signals in the neurons. A similar clinical presentation seen in many neurodegenerative disorders suggests the possibility of shared neuronal responses in different disorders. Despite the difference in core elements of deposits in each neurodegenerative disorder, the cascade of neuronal reactions such as activation of glycogen synthase kinase-3 beta, mitogen-activated protein kinases, cell cycle re-entry and oxidative stress leading to a progressive neurodegeneration are surprisingly similar. This review focuses on protein toxicity in two neurodegenerative diseases, AD and PD. We reviewed the activated mechanisms of neurotoxicity in response to misfolded beta-amyloid and α-synuclein, two major toxic proteins in AD and PD, leading to neuronal apoptosis. The interaction between the proteins in producing an overlapping pathological pattern will be also discussed.
Collapse
Affiliation(s)
- Shohreh Majd
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - John H Power
- Department of Human Physiology, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| | - Hugh J M Grantham
- Centre for Neuroscience and Paramedic Unit, School of Medicine, Flinders University of South Australia, Adelaide, SA, 5042, Australia.
| |
Collapse
|
39
|
Qiao J, Wang J, Wang H, Zhang Y, Zhu S, Adilijiang A, Guo H, Zhang R, Guo W, Luo G, Qiu Y, Xu H, Kong J, Huang Q, Li XM. Regulation of astrocyte pathology by fluoxetine prevents the deterioration of Alzheimer phenotypes in an APP/PS1 mouse model. Glia 2015; 64:240-54. [PMID: 26446044 DOI: 10.1002/glia.22926] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 09/10/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Jinping Qiao
- Mental Health Center, Shantou University; Shantou Guangdong People's Republic of China
- Clinical Laboratory; The First Affiliated Hospital of Anhui Medical University; Hefei Anhui People's Republic of China
- Department of Anatomy and Cell Science; University of Manitoba; Winnipeg Manitoba Canada
| | - Junhui Wang
- Mental Health Center, Shantou University; Shantou Guangdong People's Republic of China
- Department of Psychiatry; University of Alberta; Edmonton Alberta Canada
| | - Hongxing Wang
- Beijing Anding Hospital, Capital Medical University; Beijing People's Republic of China
| | - Yanbo Zhang
- Department of Psychiatry; College of Medicine, University of Saskatchewan; Saskatoon Saskatchewan Canada
| | - Shenghua Zhu
- Department of Anatomy and Cell Science; University of Manitoba; Winnipeg Manitoba Canada
| | | | - Huining Guo
- Department of Psychiatry; University of Alberta; Edmonton Alberta Canada
| | - Ruiguo Zhang
- Department of Psychiatry; Xijing Hospital, the Fourth Military Medical University; Xi'an Shanxi People's Republic of China
| | - Wei Guo
- Beijing Anding Hospital, Capital Medical University; Beijing People's Republic of China
| | - Gang Luo
- Department of Neurology; First Affiliated Hospital of Henan University; Henan People's Republic of China
| | - Yiqing Qiu
- Department of Neurosurgery; Shanghai Changhai Hospital, Secondary Military Medical University; Shanghai People's Republic of China
| | - Haiyun Xu
- Mental Health Center, Shantou University; Shantou Guangdong People's Republic of China
| | - Jiming Kong
- Department of Anatomy and Cell Science; University of Manitoba; Winnipeg Manitoba Canada
| | - Qingjun Huang
- Mental Health Center, Shantou University; Shantou Guangdong People's Republic of China
| | - Xin-Min Li
- Department of Psychiatry; University of Alberta; Edmonton Alberta Canada
| |
Collapse
|
40
|
D'Ambrosi N, Rossi L. Copper at synapse: Release, binding and modulation of neurotransmission. Neurochem Int 2015; 90:36-45. [PMID: 26187063 DOI: 10.1016/j.neuint.2015.07.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 10/23/2022]
Abstract
Over the last decade, a piece of the research studying copper role in biological systems was devoted to unravelling a still elusive, but extremely intriguing, aspect that is the involvement of copper in synaptic function. These studies were prompted to provide a rationale to the finding that copper is released in the synaptic cleft upon depolarization. The copper pump ATP7A, which mutations are responsible for diseases with a prominent neurodegenerative component, seems to play a pivotal role in the release of copper at synapses. Furthermore, it was found that, when in the synaptic cleft, copper can control, directly or indirectly, the activity of the neurotransmitter receptors (NMDA, AMPA, GABA, P2X receptors), thus affecting excitability. In turn, neurotransmission can affect copper trafficking and delivery in neuronal cells. Furthermore, it was reported that copper can also modulate synaptic vesicles trafficking and the interaction between proteins of the secretory pathways. Interestingly, proteins with a still unclear role in neuronal system though associated with the pathogenesis of neurodegenerative diseases (the amyloid precursor protein, APP, the prion protein, PrP, α-synuclein, α-syn) show copper-binding domains. They may act as copper buffer at synapses and participate in the interplay between copper and the neurotransmitters receptors. Given that copper dysmetabolism occurs in several diseases affecting central and peripheral nervous system, the findings on the contribution of copper in synaptic transmission, beside its more consolidate role as a neuronal enzymes cofactor, may open new insights for therapy interventions.
Collapse
Affiliation(s)
- Nadia D'Ambrosi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
41
|
Correlations between mini-mental state examination score, cerebrospinal fluid biomarkers, and pathology observed in brain biopsies of patients with normal-pressure hydrocephalus. J Neuropathol Exp Neurol 2015; 74:470-9. [PMID: 25868149 DOI: 10.1097/nen.0000000000000191] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Alzheimer disease (AD)-related pathology was assessed in cortical biopsy samples of 111 patients with idiopathic normal-pressure hydrocephalus. Alzheimer disease hallmark lesions-β-amyloid (Aβ) and hyperphosphorylated tau (HPtau)-were observed in 47% of subjects, a percentage consistent with that for whole-brain assessment reported postmortem in unselected cohorts. Higher-immunostained area fraction of AD pathology corresponded with lower preoperative mini-mental state examination scores. Concomitant Aβ and HPtau pathology, reminiscent of that observed in patients with AD, was observed in 22% of study subjects. There was a significant correlation between Aβ-immunostained area fraction in tissue and Aβ42 (42-amino-acid form of Aβ) in cerebrospinal fluid (CSF). Levels of Aβ42 were significantly lower in CSF in subjects with concomitant Aβ and HPtau pathology compared with subjects lacking pathology. Moreover, a significant correlation between HPtau-immunostained area fraction and HPtau in CSF was noted. Both HPtau and total tau were significantly higher in CSF in subjects with concomitant Aβ and HPtau pathology compared with subjects lacking pathology. The 42-amino-acid form of Aβ (Aβ42) and HPtau in CSF were the most significant predictors of the presence of AD pathology in cortical biopsies. Long-term follow-up studies are warranted to assess whether all patients with idiopathic normal-pressure hydrocephalus with AD pathology progress to AD and to determine the pathologic substrate of idiopathic normal-pressure hydrocephalus.
Collapse
|
42
|
Zhu S, Shi R, Li V, Wang J, Zhang R, Tempier A, He J, Kong J, Wang JF, Li XM. Quetiapine attenuates glial activation and proinflammatory cytokines in APP/PS1 transgenic mice via inhibition of nuclear factor-κB pathway. Int J Neuropsychopharmacol 2015; 18:pyu022. [PMID: 25618401 PMCID: PMC4360237 DOI: 10.1093/ijnp/pyu022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In Alzheimer's disease, growing evidence has shown that uncontrolled glial activation and neuroinflammation may contribute independently to neurodegeneration. Antiinflammatory strategies might provide benefits for this devastating disease. The aims of the present study are to address the issue of whether glial activation and proinflammatory cytokine increases could be modulated by quetiapine in vivo and in vitro and to explore the underlying mechanism. METHODS Four-month-old amyloid precursor protein (APP) and presenilin 1 (PS1) transgenic and nontransgenic mice were treated with quetiapine (5mg/kg/d) in drinking water for 8 months. Animal behaviors, total Aβ levels, and glial activation were evaluated by behavioral tests, enzyme-linked immunosorbent assay, immunohistochemistry, and Western blot accordingly. Inflammatory cytokines and the nuclear factor kappa B pathway were analyzed in vivo and in vitro. RESULTS Quetiapine improves behavioral performance, marginally affects total Aβ40 and Aβ42 levels, attenuates glial activation, and reduces proinflammatory cytokines in APP/PS1 mice. Quetiapine suppresses Aβ1-42-induced activation of primary microglia by decresing proinflammatory cytokines. Quetiapine inhibits the activation of nuclear factor kappa B p65 pathway in both transgenic mice and primary microglia stimulated by Aβ1-42. CONCLUSIONS The antiinflammatory effects of quetiapine in Alzheimer's disease may be involved in the nuclear factor kappa B pathway. Quetiapine may be an efficacious and promising treatment for Alzheimer's disease targeting on neuroinflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Xin-Min Li
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada (Drs Zhu and J-F. Wang); Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada (Drs Shi and Kong); Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada (Dr Li); Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University, Xi'an, China (Dr Zhang); Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada (Drs J. Wang, Tempier, and X-M. Li); First Affiliated Hospital, Henan University, Kaifeng, Henan, China (Dr He).
| |
Collapse
|
43
|
Early onset Alzheimer's disease and oxidative stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:375968. [PMID: 24669286 PMCID: PMC3942075 DOI: 10.1155/2014/375968] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/18/2013] [Indexed: 01/30/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly adults. It is estimated that 10% of the world's population aged more than 60-65 years could currently be affected by AD, and that in the next 20 years, there could be more than 30 million people affected by this pathology. One of the great challenges in this regard is that AD is not just a scientific problem; it is associated with major psychosocial and ethical dilemmas and has a negative impact on national economies. The neurodegenerative process that occurs in AD involves a specific nervous cell dysfunction, which leads to neuronal death. Mutations in APP, PS1, and PS2 genes are causes for early onset AD. Several animal models have demonstrated that alterations in these proteins are able to induce oxidative damage, which in turn favors the development of AD. This paper provides a review of many, although not all, of the mutations present in patients with familial Alzheimer's disease and the association between some of these mutations with both oxidative damage and the development of the pathology.
Collapse
|
44
|
Leong SL, Young TR, Barnham KJ, Wedd AG, Hinds MG, Xiao Z, Cappai R. Quantification of copper binding to amyloid precursor protein domain 2 and its Caenorhabditis elegans ortholog. Implications for biological function. Metallomics 2014; 6:105-16. [DOI: 10.1039/c3mt00258f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Vannuvel K, Renard P, Raes M, Arnould T. Functional and morphological impact of ER stress on mitochondria. J Cell Physiol 2013; 228:1802-18. [PMID: 23629871 DOI: 10.1002/jcp.24360] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 03/04/2013] [Indexed: 12/15/2022]
Abstract
Over the past years, knowledge and evidence about the existence of crosstalks between cellular organelles and their potential effects on survival or cell death have been constantly growing. More recently, evidence accumulated showing an intimate relationship between endoplasmic reticulum (ER) and mitochondria. These close contacts not only establish extensive physical links allowing exchange of lipids and calcium but they can also coordinate pathways involved in cell life and death. It is now obvious that ER dysfunction/stress and unfolded protein response (UPR) as well as mitochondria play major roles in apoptosis. However, while the effects of major ER stress on cell death have been largely studied and reviewed, it becomes more and more evident that cells might regularly deal with sublethal ER stress, a condition that does not necessarily lead to cell death but might affect the function/activity of other organelles such as mitochondria. In this review, we will particularly focus on these new, interesting and intriguing metabolic and morphological events that occur during the early adaptative phase of the ER stress, before the onset of cell death, and that remain largely unknown. Relevance and implication of these mitochondrial changes in response to ER stress conditions for human diseases such as type II diabetes and Alzheimer's disease will also be considered.
Collapse
Affiliation(s)
- Kayleen Vannuvel
- Laboratory of Biochemistry and Cellular Biology, URBC-NARILIS, University of Namur, Namur, Belgium
| | | | | | | |
Collapse
|
46
|
Corrigan F, Thornton E, Roisman LC, Leonard AV, Vink R, Blumbergs PC, van den Heuvel C, Cappai R. The neuroprotective activity of the amyloid precursor protein against traumatic brain injury is mediated via the heparin binding site in residues 96-110. J Neurochem 2013; 128:196-204. [DOI: 10.1111/jnc.12391] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/12/2013] [Accepted: 07/16/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Frances Corrigan
- Discipline of Anatomy and Pathology; School of Medical Sciences; University of Adelaide; Adelaide SA Australia
| | - Emma Thornton
- Discipline of Anatomy and Pathology; School of Medical Sciences; University of Adelaide; Adelaide SA Australia
| | - Laila C. Roisman
- Department of Pathology and Bio21 Molecular Science and BioTechnology Institute; The University of Melbourne; Victoria Australia
| | - Anna V. Leonard
- Discipline of Anatomy and Pathology; School of Medical Sciences; University of Adelaide; Adelaide SA Australia
| | - Robert Vink
- Discipline of Anatomy and Pathology; School of Medical Sciences; University of Adelaide; Adelaide SA Australia
| | - Peter C. Blumbergs
- Centre for Neurological Diseases; Hanson Institute; Adelaide SA Australia
| | - Corinna van den Heuvel
- Discipline of Anatomy and Pathology; School of Medical Sciences; University of Adelaide; Adelaide SA Australia
| | - Roberto Cappai
- Department of Pathology and Bio21 Molecular Science and BioTechnology Institute; The University of Melbourne; Victoria Australia
| |
Collapse
|
47
|
Amyloidosis in Alzheimer's Disease: The Toxicity of Amyloid Beta (A β ), Mechanisms of Its Accumulation and Implications of Medicinal Plants for Therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:413808. [PMID: 23762130 PMCID: PMC3671299 DOI: 10.1155/2013/413808] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 04/10/2013] [Accepted: 04/22/2013] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to memory deficits and death. While the number of individuals with AD is rising each year due to the longer life expectancy worldwide, current therapy can only somewhat relieve the symptoms of AD. There is no proven medication to cure or prevent the disease, possibly due to a lack of knowledge regarding the molecular mechanisms underlying disease pathogenesis. Most previous studies have accepted the “amyloid hypothesis,” in which the neuropathogenesis of AD is believed to be triggered by the accumulation of the toxic amyloid beta (Aβ) protein in the central nervous system (CNS). Lately, knowledge that may be critical to unraveling the hidden pathogenic pathway of AD has been revealed. This review concentrates on the toxicity of Aβ and the mechanism of accumulation of this toxic protein in the brain of individuals with AD and also summarizes recent advances in the study of these accumulation mechanisms together with the role of herbal medicines that could facilitate the development of more effective therapeutic and preventive strategies.
Collapse
|
48
|
Protective effect of cyanidin 3-O-glucoside on beta-amyloid peptide-induced cognitive impairment in rats. Neurosci Lett 2012; 534:285-8. [PMID: 23274703 DOI: 10.1016/j.neulet.2012.12.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 12/06/2012] [Accepted: 12/08/2012] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that results in cognitive impairment. It has been proposed that deposits of beta-amyloid (Aβ) form the cores of the plaque and, subsequently, induce the activation of GSK-3β and the hyperphosphorylation of tau, resulting in cognitive impairment. Oxidative stress has been proposed to be an important factor in the pathogenesis of AD. Cyanidin 3-O-glucoside (Cy3G) is a neuroprotective antioxidant. However, the effects of Cy3G on cognition are unclear. In this paper, we show that Cy3G is protective against the Aβ-induced impairment of learning and memory, but has no effect on normal learning and memory. Moreover, we found that Gy3G attenuated the Aβ-induced tau hyperphosphorylation and GSK-3β hyperactivation observed in AD. Taken together, these results demonstrated that Cy3G can rescue the cognitive impairments that are induced by Aβ via the modulation of GSK-3β/tau, suggesting a potential therapeutic role of Cy3G in AD.
Collapse
|
49
|
Abstract
A patient with the distinctive pathologic findings of macrophagic myofasciitis (MMF) is presented. This case is similar to patients with MMF first seen in France in 1993. The French group ruled out a histiocytic disorder, and later (1999) reported that this disorder represented an unusual focal reaction to intramuscular injections of vaccines containing aluminum. More recently (2001) symptomatic demyelinating disease was reported in 8 of the 92 patients with MMF who were recognized in France. The present case has thickening of the aortic and mitral valves, a finding not reported by the French group. Whereas the French group ruled out a histiocytic etiology for MMF in their patients, the cardiac findings, along with the innumerable small patchy abnormalities in the supratentorial white matter identified on magnetic resonance imaging (MRI), likely implicate a systemic histiocytic basis for this patient's unique disorder. The importance of an exhaustive workup of the patient with multiple sclerosis with diffuse myalgias is reemphasized.
Collapse
|
50
|
Lovestone S. Fleshing out the amyloid cascade hypothesis: the molecular biology of Alzheimer's disease. DIALOGUES IN CLINICAL NEUROSCIENCE 2012. [PMID: 22033981 PMCID: PMC3181594 DOI: 10.31887/dcns.2000.2.2/slovestone] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alzheimer's disease (AD) is a disorder of two pathologies- plaques and tangles. The former have as a key constituent amyloid protein and the latter the microtubule-associaied protein tau. Genetics has demonstrated that changes in either protein are sufficient to cause dementia. The amyloid cascade hypothesis proposes that plaque-related changes precede tangle-related changes and positions amyloid as central to the degeneration of AD. All the evidence suggests this is correct, including evidence that presenil ins alter the processing of the amyloid precursor protein and evidence that disrupting the normal properties of tau underlies the related froniotemporal dementias. The amyloid cascade hypothesis has provided the basis for nearly a decade of intensive basic science - the skeleton of that hypothesis can now be fleshed out, and confidence is growing that this will result in useful disease-modifying therapies in the future.
Collapse
Affiliation(s)
- S Lovestone
- Author affiliations: Institute of Psychiatry, De Crespigny Park, London, UK
| |
Collapse
|