1151
|
Machado ML, Arantes LP, da Silveira TL, Zamberlan DC, Cordeiro LM, Obetine FBB, da Silva AF, da Cruz IBM, Soares FAA, Oliveira RDP. Ilex paraguariensis extract provides increased resistance against oxidative stress and protection against Amyloid beta-induced toxicity compared to caffeine in Caenorhabditis elegans. Nutr Neurosci 2019; 24:697-709. [PMID: 31595831 DOI: 10.1080/1028415x.2019.1671694] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ilex paraguariensis is a plant from South America, used to prepare a tea-like beverage rich in caffeine and polyphenols with antioxidant proprieties. Caffeine consumption is associated with a lower risk of age-associated neuropathologies, besides several extracts that have antioxidant proprieties are known to be neuroprotective, and oxidative stress strongly correlates with Aβ-toxicity. This study aims to investigate the neuroprotective effects of the Ilex paraguariensis hydroalcoholic extract (IPHE) and to evaluate if caffeine agent present in IPHE exerts neuroprotective effects in an amyloid beta-peptide (Aβ)-induced toxicity in Caenorhabditis elegans. The wild-type and CL2006 worms were treated with IPHE (2 and 4 mg/mL) or caffeine (200 and 400 μM) since larval stage 1 (L1) until they achieved the required age for each assay. IPHE and caffeine increased the lifespan and appeared to act directly by reactive oxygen species (ROS) scavenger in both wild-type and CL2006 worms, also conferred resistance against oxidative stress in wild-type animals. Furthermore, both treatments delayed Aβ-induced paralysis and decreased AChE activity in CL2006. The protective effect of IPHE against Aβ-induced paralysis was found to be dependent on heat shock factor hsf-1 and FOXO-family transcription factor daf-16, which are respectively involved in aging-related processes and chaperone synthesis, while that of caffeine was dependent only on daf-16. Mechanistically, IPHE and caffeine decreased the levels of Aβ mRNA in the CL2006 worms; however, only IPHE induced expression of the heat shock chaperonin hsp-16.2, involved in protein homeostasis. The results were overall better when treated with IPHE than with caffeine.
Collapse
Affiliation(s)
- Marina Lopes Machado
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Leticia Priscilla Arantes
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Tássia Limana da Silveira
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Daniele Coradini Zamberlan
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Larissa Marafiga Cordeiro
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Fabiane Baptista Bicca Obetine
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Aline Franzen da Silva
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | | | - Felix Alexandre Antunes Soares
- Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Riva de Paula Oliveira
- Departamento de Biologia Celular e Genética, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
1152
|
Ahmad SS, Kamal MA. Current Updates on the Regulation of Beta-Secretase Movement as a Potential Restorative Focus for Management of Alzheimer's Disease. Protein Pept Lett 2019; 26:579-587. [DOI: 10.2174/0929866526666190405125334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 11/22/2022]
Abstract
The most recent decade was described by a developing awareness about the
seriousness of dementia in the field of age-related people. Among the dementias, Alzheimer's
assumes a plentiful role as a result of its amazingly high rate and casualty. A few
pharmacological procedures have been attempted yet at the same time now, Alzheimer continues
being an untreatable malady. The collection of Aβ in the brain is an early poisonous occasion in
the pathogenesis of Alzheimer's disease, which is the most widely recognized type of dementia
correlated with plaques and tangles within the brain. However, the mechanism of the
intraneuronal direction of BACE1 is poorly understood. AD is caused by mutations in one of the
genes that encoding APP, presenilins 1 and 2. Most of the mutations in these genes increase
Aβ42 production. Numerous receptors are associated with initiating Aβ transport and clearance.
Among them, RAGE is an influx transport receptor that binds soluble Aβ and mediates
pathophysiological cellular responses. RAGE additionally intervenes the vehicle of plasma Aβ
over the blood-brain barrier. LRP-1 functions as a clearance receptor for Aβ at the blood-brain
barrier. The regulation of beta-secretase movement is being explored as a potential restorative
focus for treating AD.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
1153
|
Nizami S, Hall‐Roberts H, Warrier S, Cowley SA, Di Daniel E. Microglial inflammation and phagocytosis in Alzheimer's disease: Potential therapeutic targets. Br J Pharmacol 2019; 176:3515-3532. [PMID: 30740661 PMCID: PMC6715590 DOI: 10.1111/bph.14618] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 12/20/2022] Open
Abstract
One of the largest unmet medical needs is a disease-modifying treatment for Alzheimer's disease (AD). Recently, the role of microglia in disease, particularly AD, has gained great interest, following the identification of several disease risk-associated genes that are highly expressed in microglia. Microglia play a critical homeostatic role in the brain, with neuroinflammatory and phagocytic mechanisms being of particular importance. Here, we review the role of NLRP3, the complement system, and the triggering receptor expressed in myeloid cells 2 (TREM2) in modulating microglial functions. We have reviewed the targets, their molecular pathways and the therapeutic interventions aimed at modulating these targets, in the hope of discovering a novel therapeutic approach for the treatment of AD. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Sohaib Nizami
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Hazel Hall‐Roberts
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Sharat Warrier
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Sally A. Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Elena Di Daniel
- Alzheimer's Research UK Oxford Drug Discovery Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
1154
|
Posado-Fernández A, Afonso CF, Dória G, Flores O, Cabrita EJ. Epitope Mapping by NMR of a Novel Anti-Aβ Antibody (STAB-MAb). Sci Rep 2019; 9:12241. [PMID: 31439854 PMCID: PMC6706428 DOI: 10.1038/s41598-019-47626-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 07/19/2019] [Indexed: 11/17/2022] Open
Abstract
Alzheimer´s Disease (AD) is one of the most common neurodegenerative disorders worldwide. Excess of β-amyloid (Aβ), a peptide with a high propensity to misfold and self-aggregate, is believed to be the major contributor to the observed neuronal degeneration and cognitive decline in AD. Here, we characterize the epitope of a novel anti-Aβ monoclonal antibody, the STAB-MAb, which has previously demonstrated picomolar affinities for both monomers (KD = 80 pM) and fibrils (KD = 130 pM) of Aβ(1–42) and has shown therapeutic efficacy in preclinical mouse models of AD. Our findings reveal a widespread epitope that embraces several key Aβ residues that have been previously described as important in the Aβ fibrillation process. Of note, STAB-MAb exhibits a stronger affinity for the N-terminus of Aβ and stabilizes an α-helix conformation in the central to N-terminal region of the peptide, in addition to disrupting a characteristic salt-bridge of a hairpin structure present in fibrils. The NMR derived epitope supports the observed results from ThT-monitored fluorescence and electron microscopy experiments, in which STAB-MAb was shown to inhibit the formation of aggregates and promote disruption of pre-formed fibrils. In combination with the published in vitro and in vivo assays, our study highlights STAB-MAb as a rare and versatile antibody with analytical, diagnostic and therapeutic efficacy.
Collapse
Affiliation(s)
- Adrián Posado-Fernández
- UCIBIO, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2825-516, Caparica, Portugal.,STAB VIDA Lda., Madan Parque, Rua dos Inventores, 2825-182, Caparica, Portugal
| | - Cláudia F Afonso
- UCIBIO, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2825-516, Caparica, Portugal.,Instituto de Medicina Molecular (iMM), Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Gonçalo Dória
- STAB VIDA Lda., Madan Parque, Rua dos Inventores, 2825-182, Caparica, Portugal
| | - Orfeu Flores
- STAB VIDA Lda., Madan Parque, Rua dos Inventores, 2825-182, Caparica, Portugal
| | - Eurico J Cabrita
- UCIBIO, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2825-516, Caparica, Portugal.
| |
Collapse
|
1155
|
Gutti G, Kakarla R, Kumar D, Beohar M, Ganeshpurkar A, Kumar A, Krishnamurthy S, Singh SK. Discovery of novel series of 2-substituted benzo[d]oxazol-5-amine derivatives as multi-target directed ligands for the treatment of Alzheimer's disease. Eur J Med Chem 2019; 182:111613. [PMID: 31437780 DOI: 10.1016/j.ejmech.2019.111613] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is associated with multifactorial neuropathological conditions, which include cholinergic deficit, amyloid-beta plaques formation, loss of neuronal plasticity and neuronal death. Treating such multifactorial conditions with a single target directed approach is considered to be inadequate. Accordingly, multi-target directed ligand (MTDL) strategy has been evolved as an auspicious approach for the treatment of AD. In light of that, a library of 2-substituted benzo[d]oxazol-5-amine derivatives (29-39; 86-107) was designed using the scaffold hopping guided MTDLs strategy, synthesized and evaluated through various in-vitro and in-vivo biological studies. The optimal compound 92 exhibited potent inhibitory activities against AChE (IC50 = 0.052 ± 0.010 μM), BuChE (IC50 = 1.085 ± 0.035 μM), and significant amyloid-beta aggregation (20 μM) inhibition. The compound possessed better blood-brain barrier permeability (Pe = 10.80 ± 0.055 × 10-6 cm s-1) in PAMPA assay and neuro protective properties (40 μM) on SH-SY5Y neuroblastoma cell lines. Furthermore, in-vivo behavioural studies were performed on Y-maze test (scopolamine-induced amnesia model) and Morris water maze test (Aβ1-42 induced ICV rat model). The compound 92, at a dose of 10 mg/kg oral administration, demonstrated a substantial improvement of the cognitive and special memory impairment. In summary, both in-vitro and in-vivo investigations evidenced that compound 92 was a potential lead for the discovery of safe and effective disease-modifying agents for AD.
Collapse
Affiliation(s)
- Gopichand Gutti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ramakrishna Kakarla
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Devendra Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Mahima Beohar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sushil Kumar Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
1156
|
Guest FL. Early Detection and Treatment of Patients with Alzheimer's Disease: Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:295-317. [PMID: 30747429 DOI: 10.1007/978-3-030-05542-4_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease affects approximately 6% of people over the age of 65 years. It is characterized as chronic degeneration of cortical neurons, with loss of memory, cognition and executive functions. As the disease progresses, it is accompanied by accumulation of amyloid plaques and neurofibrillary tangles in key areas of the brain, leading to a loss of neurogenesis and synaptic plasticity in the hippocampus, along with changes in the levels of essential neurotransmitters such as acetylcholine and glutamate. Individuals with concomitant diseases such as depression, diabetes and cardiovascular disorders have a higher risk of developing Alzheimer's disease, and those who have a healthier diet and partake in regular exercise and intellectual stimulation have a lower risk of developing the disorder. This chapter describes the advances made in early diagnosis of Alzheimer's disease as this could help to improve outcomes for the patients by facilitating earlier treatment.
Collapse
Affiliation(s)
- Francesca L Guest
- Taunton and Somerset NHS Trust, Musgrove Park Hospital, Taunton, Somerset, UK.
| |
Collapse
|
1157
|
Wang WT, Tailor BA, Cohen DS, Huang X. Alzheimer's Pathogenesis, Metal-Mediated Redox Stress, and Potential Nanotheranostics. EC PHARMACOLOGY AND TOXICOLOGY 2019; 7:547-558. [PMID: 31565701 PMCID: PMC6764777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) characterized by insoluble amyloid-β (Aβ) deposits, neurofibrillary tangles (NFTs), and neuronal demise. The influence of environmental and genetic factors on AD progression remains elusive, however evidence suggests biometal dyshomeostasis elicits neuronal death, neuroinflammation, and accumulated oxidative damages in AD brain. As such, three pathways have been identified that result from abnormal biometal accumulation and increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) in AD brain parenchyma: (1) the damage caused by direct oxidation of cellular components such as DNA and proteins; (2) the oligomerization of Aβ and NFTs, and (3) the promotion of apoptosis through NF-κB signaling pathway. Finally, given recent developments in nanotechnology, we have briefly reviewed potential nanotheranostic agents as potential AD theranostics.
Collapse
Affiliation(s)
- Willam T Wang
- Department of Diagnostic Radiology, Singapore General Hospital, Singapore
| | - Breeya A Tailor
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - David S Cohen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
1158
|
Sun D, Qiao Y, Jiang X, Li P, Kuai Z, Gong X, Liu D, Fu Q, Sun L, Li H, Ding J, Shi Y, Kong W, Shan Y. Multiple Antigenic Peptide System Coupled with Amyloid Beta Protein Epitopes As An Immunization Approach to Treat Alzheimer's Disease. ACS Chem Neurosci 2019; 10:2794-2800. [PMID: 31042358 DOI: 10.1021/acschemneuro.9b00020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Latest studies suggest that Alzheimer's disease (AD) is one of the "four big killers" that threaten the health of the elderly. AD affects about 46 million people across the world, and there is a critical need for research on new therapies for treating AD. The purpose of the present study was to develop and evaluate immunogens to elicit antibodies against the formation of amyloid beta protein (Aβ), a pathological hallmark of AD, as a therapeutic strategy in AD. In this study, serial potential immunogenic epitopes were screened according to the Aβ sequence. The screened linear epitopes on the Aβ C-terminal fragment were coupled with either the carrier protein keyhole limpet hemocyanin (KLH) or the synthesized 4-branch peptide (MAP4). MAP4 immunogens could effectively elicit immunogenicity against Aβ1-42 monomer and fiber in Balb/C mice. Furthermore, MAP4 sera could also effectively inhibit the formation of the Aβ1-42 fiber. Interestingly, one of the MAP4 sera was able to depolymerize the Aβ1-42 fibers that had aggregated. The monoclonal antibody, 1H7, was shown to inhibit the formation of Aβ1-42 fiber. MAP4 carrier may provide benefits over current immunization strategies, as it does not induce inflammation. In conclusion, the MAP4-Aβ conjugates offer a promising approach for the development of a safe and effective AD vaccine.
Collapse
Affiliation(s)
- Diya Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xiaoyu Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Pengju Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Ziyu Kuai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xin Gong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Dongni Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qiang Fu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, China
| | - Liyan Sun
- Dalian University Affiliated Xinhua Hospital, Dalian, Liaoning 116021, China
| | - He Li
- Affiliated Dalian Friendship Hospital of Dalian Medical University, Dalian, Liaoning 116100, China
| | - Jun Ding
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
1159
|
Zhang T, Loschwitz J, Strodel B, Nagel-Steger L, Willbold D. Interference with Amyloid-β Nucleation by Transient Ligand Interaction. Molecules 2019; 24:E2129. [PMID: 31195746 PMCID: PMC6600523 DOI: 10.3390/molecules24112129] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/02/2019] [Accepted: 06/04/2019] [Indexed: 12/20/2022] Open
Abstract
Amyloid-β peptide (Aβ) is an intrinsically disordered protein (IDP) associated with Alzheimer's disease. The structural flexibility and aggregation propensity of Aβ pose major challenges for elucidating the interaction between Aβ monomers and ligands. All-D-peptides consisting solely of D-enantiomeric amino acid residues are interesting drug candidates that combine high binding specificity with high metabolic stability. Here we characterized the interaction between the 12-residue all-D-peptide D3 and Aβ42 monomers, and how the interaction influences Aβ42 aggregation. We demonstrate for the first time that D3 binds to Aβ42 monomers with submicromolar affinities. These two highly unstructured molecules are able to form complexes with 1:1 and other stoichiometries. Further, D3 at substoichiometric concentrations effectively slows down the β-sheet formation and Aβ42 fibrillation by modulating the nucleation process. The study provides new insights into the molecular mechanism of how D3 affects Aβ assemblies and contributes to our knowledge on the interaction between two IDPs.
Collapse
Affiliation(s)
- Tao Zhang
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Jennifer Loschwitz
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany.
- Institute of Theoretical and Computational Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Birgit Strodel
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany.
- Institute of Theoretical and Computational Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Luitgard Nagel-Steger
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| | - Dieter Willbold
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany.
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
1160
|
Giridharan VV, Masud F, Petronilho F, Dal-Pizzol F, Barichello T. Infection-Induced Systemic Inflammation Is a Potential Driver of Alzheimer's Disease Progression. Front Aging Neurosci 2019; 11:122. [PMID: 31191296 PMCID: PMC6546917 DOI: 10.3389/fnagi.2019.00122] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Vijayasree V Giridharan
- Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Faisal Masud
- Department of Anesthesiology, Houston Methodist Hospital, Houston, TX, United States
| | - Fabricia Petronilho
- Health Sciences Unit, Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, University of South Santa Catarina, Tubarão, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, Brazil
| | - Tatiana Barichello
- Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, Brazil.,Graduate Program in Health Sciences, Laboratory of Neurosciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| |
Collapse
|
1161
|
Yuksel M, Tacal O. Trafficking and proteolytic processing of amyloid precursor protein and secretases in Alzheimer's disease development: An up-to-date review. Eur J Pharmacol 2019; 856:172415. [PMID: 31132354 DOI: 10.1016/j.ejphar.2019.172415] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD), which is predicted to affect 1 in 85 persons worldwide by 2050, results in progressive loss of neuronal functions, leading to impairments in memory and cognitive abilities. As being one of the major neuropathological hallmarks of AD, senile plaques mainly consist of amyloid-β (Aβ) peptides, which are derived from amyloid precursor protein (APP) via the sequential cleavage by β- and γ-secretases. Although the overproduction and accumulation of Aβ peptides are at the center of AD research, the new discoveries point out to the complexity of the disease development. In this respect, it is crucial to understand the processing and the trafficking of APP, the enzymes involved in its processing, the cleavage products and their therapeutic potentials. This review summarizes the salient features of APP processing focusing on APP, the canonical secretases as well as the novel secretases and the cleavage products with an update of the recent developments. We also discussed the intracellular trafficking of APP and secretases in addition to their potential in AD therapy.
Collapse
Affiliation(s)
- Melike Yuksel
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| | - Ozden Tacal
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
1162
|
Corradi V, Sejdiu BI, Mesa-Galloso H, Abdizadeh H, Noskov SY, Marrink SJ, Tieleman DP. Emerging Diversity in Lipid-Protein Interactions. Chem Rev 2019; 119:5775-5848. [PMID: 30758191 PMCID: PMC6509647 DOI: 10.1021/acs.chemrev.8b00451] [Citation(s) in RCA: 312] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Indexed: 02/07/2023]
Abstract
Membrane lipids interact with proteins in a variety of ways, ranging from providing a stable membrane environment for proteins to being embedded in to detailed roles in complicated and well-regulated protein functions. Experimental and computational advances are converging in a rapidly expanding research area of lipid-protein interactions. Experimentally, the database of high-resolution membrane protein structures is growing, as are capabilities to identify the complex lipid composition of different membranes, to probe the challenging time and length scales of lipid-protein interactions, and to link lipid-protein interactions to protein function in a variety of proteins. Computationally, more accurate membrane models and more powerful computers now enable a detailed look at lipid-protein interactions and increasing overlap with experimental observations for validation and joint interpretation of simulation and experiment. Here we review papers that use computational approaches to study detailed lipid-protein interactions, together with brief experimental and physiological contexts, aiming at comprehensive coverage of simulation papers in the last five years. Overall, a complex picture of lipid-protein interactions emerges, through a range of mechanisms including modulation of the physical properties of the lipid environment, detailed chemical interactions between lipids and proteins, and key functional roles of very specific lipids binding to well-defined binding sites on proteins. Computationally, despite important limitations, molecular dynamics simulations with current computer power and theoretical models are now in an excellent position to answer detailed questions about lipid-protein interactions.
Collapse
Affiliation(s)
- Valentina Corradi
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Besian I. Sejdiu
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Haydee Mesa-Galloso
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Haleh Abdizadeh
- Groningen
Biomolecular Sciences and Biotechnology Institute and Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Sergei Yu. Noskov
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Siewert J. Marrink
- Groningen
Biomolecular Sciences and Biotechnology Institute and Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - D. Peter Tieleman
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
1163
|
Salimi A, Li H, Shi H, Lee JY. Intrinsic origin of amyloid aggregation: Behavior of histidine (εεε) and (δδδ) tautomer homodimers of Aβ (1–40). Biochim Biophys Acta Gen Subj 2019; 1863:795-801. [DOI: 10.1016/j.bbagen.2019.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 02/08/2023]
|
1164
|
Cao K, Xiang J, Dong YT, Xu Y, Li Y, Song H, Zeng XX, Ran LY, Hong W, Guan ZZ. Exposure to fluoride aggravates the impairment in learning and memory and neuropathological lesions in mice carrying the APP/PS1 double-transgenic mutation. Alzheimers Res Ther 2019; 11:35. [PMID: 31010414 PMCID: PMC6477877 DOI: 10.1186/s13195-019-0490-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is responsible for 60-70% of all cases of dementia. On the other hand, the tap water consumed by hundreds of millions of people has been fluoridated to prevent tooth decay. However, little is known about the influence of fluoride on the expression of APP and subsequent changes in learning and memory and neuropathological injury. Our aim here was to determine whether exposure to fluoride aggravates the neuropathological lesions in mice carrying the amyloid precursor protein (APP)/presenilin1 (PS1) double mutation. METHODS These transgenic or wide-type (WT) mice received 0.3 ml of a solution of fluoride (0.1 or 1 mg/ml, prepared with NaF) by intragastric administration once each day for 12 weeks. The learning and memory of these animals were assessed with the Morris water maze test. Senile plaques, ionized calcium binding adaptor molecule 1 (Iba-1), and complement component 3 (C3) expression were semi-quantified by immunohistochemical staining; the level of Aβ42 was detected by Aβ42 enzyme-linked immunosorbent assays (ELISAs); the levels of synaptic proteins and enzymes that cleave APP determined by Western blotting; and the malondialdehyde (MDA) content and activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) measured by biochemical procedures. RESULTS The untreated APP mice exhibited a decline in learning and memory after 12 weeks of fluoride treatment, whereas treatment of these some animals with low or high levels of fluoride led to such declines after only 4 or 8 weeks, respectively. Exposure of APP mice to fluoride elevated the number of senile plaques and level of Aβ42, Iba-1, and BACE1, while reducing the level of ADAM10 in their brains. The lower levels of synaptic proteins and enhanced oxidative stress detected in the hippocampus of APP mice were aggravated to fluoride. CONCLUSIONS These findings indicate that exposure to fluoride, even at lower concentration, can aggravate the deficit in learning and memory and neuropathological lesions of the mice that express the high level of APP.
Collapse
Affiliation(s)
- Kun Cao
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Jie Xiang
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Yang-Ting Dong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Yi Xu
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Yi Li
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Hui Song
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Xiao-Xiao Zeng
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Long-Yan Ran
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
| | - Wei Hong
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| | - Zhi-Zhong Guan
- Department of Pathology at the Affiliated Hospital of Guizhou Medical University, Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Endemic and Ethnic Diseases of the Ministry of Education of P. R. China (Guizhou Medical University), Guiyang, 550004 Guizhou People’s Republic of China
- Key Laboratory of Medical Molecular Biology, Guiyang, 550004 Guizhou People’s Republic of China
| |
Collapse
|
1165
|
Toffa DH, Magnerou MA, Kassab A, Hassane Djibo F, Sow AD. Can magnesium reduce central neurodegeneration in Alzheimer's disease? Basic evidences and research needs. Neurochem Int 2019; 126:195-202. [PMID: 30905744 DOI: 10.1016/j.neuint.2019.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/15/2019] [Accepted: 03/18/2019] [Indexed: 12/26/2022]
Abstract
Magnesium (Mg) is a crucial divalent cation with more than 300 cellular functions. This ion shows therapeutic properties in several neurological diseases. Although there are numerous basic evidences showing that Mg can inhibit pathological processes involved in neuroglial degeneration, this low-cost option is not well-considered in clinical research and practice for now. Nevertheless, none of the expensive drugs currently recommended by the classic guidelines (in addition to physiological rehabilitation) had shown exceptional effectiveness. Herein, focusing on Alzheimer's disease (AD), we analyze the therapeutic pathways that support the use of Mg for neurogenesis and neuroprotection. According to experimental findings reviewed, Mg shows interesting abilities to facilitate toxin clearance, reduce neuroinflammation, inhibit the pathologic processing of amyloid protein precursor (APP) as well as the abnormal tau protein phosphorylation, and to reverse the deregulation of N-methyl-D-aspartate receptors. Currently, some crucial details of the mechanisms involved in these proved effects remain elusive and clinical background is poor. Therefore, further studies are required to enable a better overview on pharmacodynamic targets of Mg and thus, to find optimal pharmacologic strategies for clinical use of this ion.
Collapse
Affiliation(s)
- Dènahin Hinnoutondji Toffa
- Epilepsy Lab, CRCHUM, Université de Montréal, Montreal, Canada; Neurology Division, CHUM, Université de Montréal, Montreal, Canada.
| | | | - Ali Kassab
- Epilepsy Lab, CRCHUM, Université de Montréal, Montreal, Canada
| | | | | |
Collapse
|
1166
|
Espargaró A, Pont C, Gamez P, Muñoz-Torrero D, Sabate R. Amyloid Pan-inhibitors: One Family of Compounds To Cope with All Conformational Diseases. ACS Chem Neurosci 2019; 10:1311-1317. [PMID: 30380841 DOI: 10.1021/acschemneuro.8b00398] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyloids are ubiquitous protein aggregates sharing common internal structural features; they are present in all organisms, from prokaryotes to eukaryotes, where they play physiological or pathological roles. Importantly, amyloids, which are generated by aggregation of a range of distinct proteins, could be a key factor in a number of major human disorders, the so-called conformational diseases. Because all amyloids exhibit similar cross-β motifs, one may envisage that molecules capable of blocking the formation of β-sheet structures could abolish aggregation of all amyloid proteins, albeit with different efficacies. Herein, two different β-sheet blockers were tested against a selection of amyloidogenic proteins, encompassing all the major types of amyloid-based disorders. Analysis of their blocking efficiency, using a simple but contrasted cell-based screening procedure, unequivocally confirms that they indeed behave as aggregation pan-inhibitors. The significant inhibitory effects observed for these compounds against all tested amyloidogenic proteins could spur a broader biological evaluation of other known and new amyloid aggregation inhibitors to further determine the potential use of this class of compounds for the universal treatment of conformational diseases.
Collapse
Affiliation(s)
- Alba Espargaró
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, E-08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, E-08028 Barcelona, Spain
| | - Caterina Pont
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, E-08028 Barcelona, Spain
| | - Patrick Gamez
- Department of Organic and Inorganic Chemistry, Faculty of Chemistry, University of Barcelona, E-08028 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), E-08010 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, E-08028 Barcelona, Spain
| | - Diego Muñoz-Torrero
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, E-08028 Barcelona, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, E-08028 Barcelona, Spain
| | - Raimon Sabate
- Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, E-08028 Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, E-08028 Barcelona, Spain
| |
Collapse
|
1167
|
Banerjee V, Oren O, Dagan B, Taube R, Engel S, Papo N. An Engineered Variant of the B1 Domain of Protein G Suppresses the Aggregation and Toxicity of Intra- and Extracellular Aβ42. ACS Chem Neurosci 2019; 10:1488-1496. [PMID: 30428260 DOI: 10.1021/acschemneuro.8b00491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Intra- and extraneuronal deposition of amyloid β (Aβ) peptides have been linked to Alzheimer's disease (AD). While both intra- and extraneuronal Aβ deposits affect neuronal cell viability, the molecular mechanism by which these Aβ structures, especially when intraneuronal, do so is still not entirely understood. This makes the development of inhibitors challenging. To prevent the formation of toxic Aβ structural assemblies so as to prevent neuronal cell death associated with AD, we used a combination of computational and combinatorial-directed evolution approaches to develop a variant of the HTB1 protein (HTB1M2). HTB1M2 inhibits in vitro self-assembly of Aβ42 peptide and shifts the Aβ42 aggregation pathway to the formation of oligomers that are nontoxic to neuroblastoma SH-SY5Y cells overexpressing or treated with Aβ42 peptide. This makes HTB1M2 a potential therapeutic lead in the development of AD-targeted drugs and a tool for elucidating conformational changes in the Aβ42 peptide.
Collapse
Affiliation(s)
- Victor Banerjee
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| | - Ofek Oren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Bar Dagan
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| | - Ran Taube
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Stanislav Engel
- The National Institute for Biotechnology in the Negev, P.O. Box 653, Beer Sheva 84105, Israel
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| | - Niv Papo
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer Sheva 84105, Israel
- The National Institute for Biotechnology in the Negev, P.O. Box 653, Beer Sheva 84105, Israel
| |
Collapse
|
1168
|
Maia MA, Sousa E. BACE-1 and γ-Secretase as Therapeutic Targets for Alzheimer's Disease. Pharmaceuticals (Basel) 2019; 12:ph12010041. [PMID: 30893882 PMCID: PMC6469197 DOI: 10.3390/ph12010041] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a growing global health concern with a massive impact on affected individuals and society. Despite the considerable advances achieved in the understanding of AD pathogenesis, researchers have not been successful in fully identifying the mechanisms involved in disease progression. The amyloid hypothesis, currently the prevalent theory for AD, defends the deposition of β-amyloid protein (Aβ) aggregates as the trigger of a series of events leading to neuronal dysfunction and dementia. Hence, several research and development (R&D) programs have been led by the pharmaceutical industry in an effort to discover effective and safety anti-amyloid agents as disease modifying agents for AD. Among 19 drug candidates identified in the AD pipeline, nine have their mechanism of action centered in the activity of β or γ-secretase proteases, covering almost 50% of the identified agents. These drug candidates must fulfill the general rigid prerequisites for a drug aimed for central nervous system (CNS) penetration and selectivity toward different aspartyl proteases. This review presents the classes of γ-secretase and beta-site APP cleaving enzyme 1 (BACE-1) inhibitors under development, highlighting their structure-activity relationship, among other physical-chemistry aspects important for the successful development of new anti-AD pharmacological agents.
Collapse
Affiliation(s)
- Miguel A Maia
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), Terminal de Cruzeiros do Porto de Leixões, Av. General Norton de Matos s/n, 4450-208 Matosinhos, Portugal.
| |
Collapse
|
1169
|
Dynamic effect of beta-amyloid 42 on cell mechanics. J Biomech 2019; 86:79-88. [DOI: 10.1016/j.jbiomech.2019.01.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/10/2019] [Accepted: 01/24/2019] [Indexed: 01/06/2023]
|
1170
|
Weber A, Mak SH, Berenbaum F, Sellam J, Zheng YP, Han Y, Wen C. Association between osteoarthritis and increased risk of dementia: A systemic review and meta-analysis. Medicine (Baltimore) 2019; 98:e14355. [PMID: 30855434 PMCID: PMC6417538 DOI: 10.1097/md.0000000000014355] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To investigate the possible association between osteoarthritis (OA) and the risk of dementia. METHODS Cohort, case-control, and cross-sectional studies were obtained from wide literature search up to 20 April 2018 from following electronic databases: PubMed, Embase, Cochrane, using the MeSH terms: "osteoarthritis" AND "dementia". The literature search was then expanded to congress abstracts. After screening and selection of relevant studies by two investigators, data was extracted. Estimates were then calculated using a random-effect size model. Sensitivity-analysis was conducted for gender and age adjusted studies and pooled for studies with STROBE quality assessment score ≥75%. Publication bias was assessed by Funnel plot. Analyses were performed using Data Analysis and Statistical Software Version 14.2. RESULTS Nearly 1549 publication references were initially retrieved. Twenty-six publications were checked with full-text. Six observational studies with 388,252 individuals were included. OA was associated with a significantly increased risk for dementia (OR = 1.20; 95% confidence interval (CI), 1.03-1.39, I = 95.6%, P < .05). After pooling the studies with adjustment of age and gender, the risk increased (OR 1.36; 95% CI, 1.22-1.51, I = 75.6%, P < .0001). After pooling the study with a STROBE Quality score ≥75% the risk for dementia was slightly increased (OR 1.33; 95% CI, 1.17-1.5, I = 93.5%, p < 0.0001). CONCLUSIONS There is an association between osteoarthritis and the risk of dementia. This meta-analysis does not provide causality. Further prospective cohort studies are needed to clarify, if knee-, hip-, or hand-OA are independent risk factors for Alzheimer's disease and vascular dementia.
Collapse
Affiliation(s)
- Adrian Weber
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Shing hung Mak
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PR China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Francis Berenbaum
- Department of Rheumatology, Sorbonne University, Saint-Antoine Hospital, Paris, France
| | - Jérémie Sellam
- Department of Rheumatology, Sorbonne University, Saint-Antoine Hospital, Paris, France
| | - Yong-Ping Zheng
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Yifan Han
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PR China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Chunyi Wen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| |
Collapse
|
1171
|
Kowalski K, Mulak A. Brain-Gut-Microbiota Axis in Alzheimer's Disease. J Neurogastroenterol Motil 2019; 25:48-60. [PMID: 30646475 PMCID: PMC6326209 DOI: 10.5056/jnm18087] [Citation(s) in RCA: 516] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/21/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022] Open
Abstract
Disturbances along the brain-gut-microbiota axis may significantly contribute to the pathogenesis of neurodegenerative disorders. Alzheimer's disease (AD) is the most frequent cause of dementia characterized by a progressive decline in cognitive function associated with the formation of amyloid beta (Aβ) plaques and neurofibrillary tangles. Alterations in the gut microbiota composition induce increased permeability of the gut barrier and immune activation leading to systemic inflammation, which in turn may impair the blood-brain barrier and promote neuroinflammation, neural injury, and ultimately neurodegeneration. Recently, Aβ has also been recognized as an antimicrobial peptide participating in the innate immune response. However, in the dysregulated state, Aβ may reveal harmful properties. Importantly, bacterial amyloids through molecular mimicry may elicit cross-seeding of misfolding and induce microglial priming. The Aβ seeding and propagation may occur at different levels of the brain-gut-microbiota axis. The potential mechanisms of amyloid spreading include neuron-to-neuron or distal neuron spreading, direct blood-brain barrier crossing or via other cells as astrocytes, fibroblasts, microglia, and immune system cells. A growing body of experimental and clinical data confirms a key role of gut dysbiosis and gut microbiota-host interactions in neurodegeneration. The convergence of gut-derived inflammatory response together with aging and poor diet in the elderly contribute to the pathogenesis of AD. Modification of the gut microbiota composition by food-based therapy or by probiotic supplementation may create new preventive and therapeutic options in AD.
Collapse
Affiliation(s)
- Karol Kowalski
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Poland
| | - Agata Mulak
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Poland
| |
Collapse
|
1172
|
Diagnostic and prognostic biomarkers for HAND. J Neurovirol 2019; 25:686-701. [PMID: 30607890 DOI: 10.1007/s13365-018-0705-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023]
Abstract
In 2007, the nosology for HIV-1-associated neurocognitive disorders (HAND) was updated to a primarily neurocognitive disorder. However, currently available diagnostic tools lack the sensitivity and specificity needed for an accurate diagnosis for HAND. Scientists and clinicians, therefore, have been on a quest for an innovative biomarker to diagnose (i.e., diagnostic biomarker) and/or predict (i.e., prognostic biomarker) the progression of HAND in the post-combination antiretroviral therapy (cART) era. The present review examined the utility and challenges of four proposed biomarkers, including neurofilament light (NFL) chain concentration, amyloid (i.e., sAPPα, sAPPβ, amyloid β) and tau proteins (i.e., total tau, phosphorylated tau), resting-state functional magnetic resonance imaging (fMRI), and prepulse inhibition (PPI). Although significant genotypic differences have been observed in NFL chain concentration, sAPPα, sAPPβ, amyloid β, total tau, phosphorylated tau, and resting-state fMRI, inconsistencies and/or assessment limitations (e.g., invasive procedures, lack of disease specificity, cost) challenge their utility as a diagnostic and/or prognostic biomarker for milder forms of neurocognitive impairment (NCI) in the post-cART era. However, critical evaluation of the literature supports the utility of PPI as a powerful diagnostic biomarker with high accuracy (i.e., 86.7-97.1%), sensitivity (i.e., 89.3-100%), and specificity (i.e., 79.5-94.1%). Additionally, the inclusion of multiple CSF and/or plasma markers, rather than a single protein, may provide a more sensitive diagnostic biomarker for HAND; however, a pressing need for additional research remains. Most notably, PPI may serve as a prognostic biomarker for milder forms of NCI, evidenced by its ability to predict later NCI in higher-order cognitive domains with regression coefficients (i.e., r) greater than 0.8. Thus, PPI heralds an opportunity for the development of a brief, noninvasive diagnostic and promising prognostic biomarker for milder forms of NCI in the post-cART era.
Collapse
|
1173
|
Coronel R, Palmer C, Bernabeu-Zornoza A, Monteagudo M, Rosca A, Zambrano A, Liste I. Physiological effects of amyloid precursor protein and its derivatives on neural stem cell biology and signaling pathways involved. Neural Regen Res 2019; 14:1661-1671. [PMID: 31169172 PMCID: PMC6585543 DOI: 10.4103/1673-5374.257511] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The pathological implication of amyloid precursor protein (APP) in Alzheimer's disease has been widely documented due to its involvement in the generation of amyloid-β peptide. However, the physiological functions of APP are still poorly understood. APP is considered a multimodal protein due to its role in a wide variety of processes, both in the embryo and in the adult brain. Specifically, APP seems to play a key role in the proliferation, differentiation and maturation of neural stem cells. In addition, APP can be processed through two canonical processing pathways, generating different functionally active fragments: soluble APP-α, soluble APP-β, amyloid-β peptide and the APP intracellular C-terminal domain. These fragments also appear to modulate various functions in neural stem cells, including the processes of proliferation, neurogenesis, gliogenesis or cell death. However, the molecular mechanisms involved in these effects are still unclear. In this review, we summarize the physiological functions of APP and its main proteolytic derivatives in neural stem cells, as well as the possible signaling pathways that could be implicated in these effects. The knowledge of these functions and signaling pathways involved in the onset or during the development of Alzheimer's disease is essential to advance the understanding of the pathogenesis of Alzheimer's disease, and in the search for potential therapeutic targets.
Collapse
Affiliation(s)
- Raquel Coronel
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Charlotte Palmer
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Adela Bernabeu-Zornoza
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - María Monteagudo
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Andreea Rosca
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Alberto Zambrano
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| |
Collapse
|
1174
|
Kim JH, Lee S, Cho EJ. Acer okamotoanumand isoquercitrin improve cognitive functionviaattenuation of oxidative stress in high fat diet- and amyloid beta-induced mice. Food Funct 2019; 10:6803-6814. [DOI: 10.1039/c9fo01694e] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
High fat diet (HFD) and accumulation of amyloid beta (Aβ) are known as a risk factor of Alzheimer's disease.Acer okamotoanumand isoquercitrin improved cognition function against both HFD and Aβ accumulation by inhibiting oxidative stress.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Food Science and Nutrition & Kimchi Research Institute
- Pusan National University
- Busan 46241
- Republic of Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology
- Chung-Ang University
- Anseong 17546
- Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute
- Pusan National University
- Busan 46241
- Republic of Korea
| |
Collapse
|
1175
|
Tan C, Dong Y, Wang J, Yang X. Vanadyl acetylacetonate attenuates Aβ pathogenesis in APP/PS1 transgenic mice depending on the intervention stage. NEW J CHEM 2019. [DOI: 10.1039/c9nj00820a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
VAC treatment caused different Grp75 responses before and after Aβ plaque formation.
Collapse
Affiliation(s)
- Chang Tan
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Yaqiong Dong
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Jing Wang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| | - Xiaoda Yang
- The State Key Laboratories of Natural and Biomimetic Drugs and Department of Chemical Biology
- School of Pharmaceutical Science
- Peking University Health Science Center
- Beijing 100191
- China
| |
Collapse
|
1176
|
Zanon VS, Lima JA, Cuya T, Lima FRS, da Fonseca ACC, Gomez JG, Ribeiro RR, França TCC, Vargas MD. In-vitro evaluation studies of 7-chloro-4-aminoquinoline Schiff bases and their copper complexes as cholinesterase inhibitors. J Inorg Biochem 2018; 191:183-193. [PMID: 30530179 DOI: 10.1016/j.jinorgbio.2018.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is one of the most common age-related neurodegenerative disorders. Aggregation of amyloid-β peptide into extracellular plaques with incorporation of metal ions, such as Cu2+, and reduction of the neurotransmitter acetylcholine levels are among the factors associated to the AD brain. Hence, a series of 7-chloro-4-aminoquinoline Schiff bases (HLa-e) were synthesized and their cytotoxicity and anti-cholinesterase activity, assessed for Alzheimer's disease. The intrinsic relationship between Cu2+ and the amyloidogenic plaques encouraged us to investigate the chelating ability of HLa-e. Dimeric tetracationic compounds, [Cu2(NHLa-e)4]Cl4, containing quinoline protonated ligands were isolated from the reactions with CuCl2·2H2O and fully characterized in the solid state, including an X ray diffraction study, whereas EPR data showed that the complexes exist as monomers in DMSO solution. The inhibitory activity of all compounds was evaluated by Ellman's spectrophotometric method in acetylcholinesterase (AChE) from Electrophorus electricus and butyrylcholinesterase (BChE) from equine serum. HLa-e and [Cu(NHLd)2]Cl2 were selective for AChE (IC50 = 4.61-9.31 μM) and were not neurotoxic in primary brain cultures. Docking and molecular dynamics studies of HLa-e inside AChE were performed and the results suggested that these compounds are able to bind inside AChE similarly to other AChE inhibitors, such as donepezil. Studies of the affinity of HLd for Cu2+ in DMSO/HEPES at pH 6.6 and pH 7.4 in μM concentrations showed formation of analogous 1:2 Cu2+/ligand complexes, which may suggest that in the AD-affected brain HLd may scavenge Cu2+ and the complex, also inhibit AChE.
Collapse
Affiliation(s)
- Vanessa S Zanon
- Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-141 Niterói, RJ, Brazil
| | - Josélia A Lima
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio de Janeiro, 21941-909 Rio de Janeiro, RJ, Brazil; Laboratório de Modelagem Aplicada a Defesa Química e Biológica (LMDQB), Instituto Militar de Engenharia, 22290-270 Rio de Janeiro, RJ, Brazil
| | - Teobaldo Cuya
- Faculdade de Tecnologia, Departamento de Matemática, Física e Computação, Universidade do Estado do Rio de Janeiro, 27537-000 Resende, RJ, Brazil
| | - Flavia R S Lima
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Anna C C da Fonseca
- Laboratório de Biologia das Células Gliais, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Javier G Gomez
- Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-141 Niterói, RJ, Brazil
| | - Ronny R Ribeiro
- Departamento de Química, Universidade Federal do Paraná, CP 19081, 81531-990 Curitiba, PR, Brazil
| | - Tanos C C França
- Laboratório de Modelagem Aplicada a Defesa Química e Biológica (LMDQB), Instituto Militar de Engenharia, 22290-270 Rio de Janeiro, RJ, Brazil
| | - Maria D Vargas
- Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-141 Niterói, RJ, Brazil.
| |
Collapse
|
1177
|
Alzheimer's Aβ
1‐40
peptide degradation by thermolysin: evidence of inhibition by a C‐terminal Aβ product. FEBS Lett 2018; 593:128-137. [DOI: 10.1002/1873-3468.13285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/18/2018] [Accepted: 10/30/2018] [Indexed: 01/23/2023]
|
1178
|
Dapson RW. Amyloid from a histochemical perspective. A review of the structure, properties and types of amyloid, and a proposed staining mechanism for Congo red staining. Biotech Histochem 2018; 93:543-556. [PMID: 30403893 DOI: 10.1080/10520295.2018.1528385] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Amyloid is a diverse group of unrelated peptides or proteins that have positive functionality or are associated with various pathologies. Despite vast differences, all amyloids share several features that together uniquely define the group. 1) All amyloids possess a characteristic cross-ß pattern with X-ray diffraction typical of ß-sheet secondary protein structures. 2) All amyloids are birefringent and dichroic under polarizing microscopy after staining with Congo red, which indicates a crystalline-like (ordered) structure. 3) All amyloids cause a spectral shift in the peak wavelength of Congo red with conventional light microscopy due to perturbation of π electrons of the dye. 4) All amyloids show heightened intensity of fluorescence with Congo red, which suggests an unusual degree of packing of the dye onto the substrate. The ß portion of amyloid molecules, the only logical substrate for specific Congo red staining under histochemical conditions, consists of a stack of ß-sheets laminated by hydrophilic and hydrophobic interactions between adjacent pairs. Only the first and last ß-sheets are accessible to dyes. Each sheet is composed of numerous identical peptides running across the width of the sheet and arranged in parallel with side chains in register over the length of the fibril. Two sets of grooves are bordered by side chains. X grooves run perpendicular to the long axis of the fibril; these grooves are short (the width of the sheet) and number in the hundreds or thousands. Y grooves are parallel with the long axis. Each groove runs the entire length of the fibril, but there are very few of them. While Congo red is capable of ionic bonding with proteins via two sulfonic acid groups, physical constraints on the staining solution preclude ionic interactions. Hydrogen bonding between dye amine groups and peptide carbonyls is the most likely primary bonding mechanism, because all ß-sheets possess backbone carbonyls. Various amino acid residues may form secondary bonds to the dye via any of three van der Waals forces. It is possible that Congo red binds within the Y grooves, but that would not produce the characteristic staining features that are the diagnostic hallmarks of amyloid. Binding in the X grooves would produce a tightly packed series of dye molecules over the entire length of the fibril. This would account for the signature staining of amyloid by Congo red: dichroic birefringence, enhanced intensity of fluorescence and a shift in visible absorption wavelength.
Collapse
|
1179
|
Activation of α7 nicotinic acetylcholine receptor alleviates Aβ1-42-induced neurotoxicity via downregulation of p38 and JNK MAPK signaling pathways. Neurochem Int 2018; 120:238-250. [DOI: 10.1016/j.neuint.2018.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 01/08/2023]
|
1180
|
Choi ML, Gandhi S. Crucial role of protein oligomerization in the pathogenesis of Alzheimer's and Parkinson's diseases. FEBS J 2018; 285:3631-3644. [PMID: 29924502 DOI: 10.1111/febs.14587] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/11/2018] [Accepted: 06/14/2018] [Indexed: 12/16/2022]
Abstract
Misfolding and aggregation of the proteins amyloid-β, tau and alpha-synuclein is the predominant pathology underlying the neurodegenerative disorders, Alzheimer's and Parkinson's disease. While end stage insoluble products of aggregation have been well characterised in human and animal models of disease, accumulating evidence from biophysical, cellular and in vivo studies has shown that soluble intermediates of aggregation, or oligomers, may be the key species that mediate toxicity and underlie seeding and spreading in disease. Here, we review the process of protein misfolding, and the intrinsic and extrinsic processes that cause the native states of the key aggregating proteins to undergo conformational change to form oligomers and ultimately fibrils. We discuss the structural features of the key toxic intermediate, and describe the putative mechanisms by which oligomers may cause cell toxicity. Finally, we explore the potential therapeutic approaches raised by the oligomer hypothesis in neurodegenerative disease.
Collapse
Affiliation(s)
- Minee L Choi
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK.,The Francis Crick Institute, London, UK
| | - Sonia Gandhi
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London, UK.,The Francis Crick Institute, London, UK
| |
Collapse
|
1181
|
Li G, Yang WY, Zhao YF, Chen YX, Hong L, Li YM. Differential Modulation of the Aggregation of N-Terminal Truncated Aβ using Cucurbiturils. Chemistry 2018; 24:13647-13653. [DOI: 10.1002/chem.201802655] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Gao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry; Tsinghua University; 100084 Beijing China
| | - Wu-Yue Yang
- Zhou Pei-Yuan Center for Applied Mathematics; Department of, Mathematical Sciences; Tsinghua University; 100084 Beijing China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry; Tsinghua University; 100084 Beijing China
| | - Yong-Xiang Chen
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry; Tsinghua University; 100084 Beijing China
| | - Liu Hong
- Zhou Pei-Yuan Center for Applied Mathematics; Department of, Mathematical Sciences; Tsinghua University; 100084 Beijing China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry; Tsinghua University; 100084 Beijing China
- Beijing Institute for Brain Disorders; 100069 Beijing China
| |
Collapse
|
1182
|
Meker S, Chin H, Sut TN, Cho NJ. Amyloid-β Peptide Triggers Membrane Remodeling in Supported Lipid Bilayers Depending on Their Hydrophobic Thickness. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:9548-9560. [PMID: 30021071 DOI: 10.1021/acs.langmuir.8b01196] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Amyloid-β (Aβ) peptide has been implicated in Alzheimer's disease, which is a leading cause of death worldwide. The interaction of Aβ peptides with the lipid bilayers of neuronal cells is a critical step in disease pathogenesis. Recent evidence indicates that lipid bilayer thickness influences Aβ membrane-associated aggregation, while understanding how Aβ interacts with lipid bilayers remains elusive. To address this question, we employed supported lipid bilayer (SLB) platforms composed of different-length phosphatidylcholine (PC) lipids (C12:0 DLPC, C18:1 DOPC, C18:1-C16:0 POPC), and characterized the resulting interactions with soluble Aβ monomers. Quartz crystal microbalance-dissipation (QCM-D) experiments identified concentration-dependent Aβ peptide adsorption onto all tested SLBs, which was corroborated by fluorescence recovery after photobleaching (FRAP) experiments indicating that higher Aβ concentrations led to decreased membrane fluidity. These commonalities pointed to strong Aβ peptide-membrane interactions in all cases. Notably, time-lapsed fluorescence microscopy revealed major differences in Aβ-induced membrane morphological responses depending on SLB hydrophobic thickness. For thicker DOPC and POPC SLBs, membrane remodeling involved the formation of elongated tubule and globular structures as a passive means to regulate membrane stress depending on Aβ concentration. In marked contrast, thin DLPC SLBs were not able to accommodate extensive membrane remodeling. Taken together, our findings reveal that membrane thickness influences the membrane morphological response triggered upon Aβ adsorption.
Collapse
Affiliation(s)
- Sigalit Meker
- School of Materials Science and Engineering , Nanyang Technological University , 639798 , Singapore
- Centre for Biomimetic Sensor Science , Nanyang Technological University , 637553 , Singapore
| | - Hokyun Chin
- School of Materials Science and Engineering , Nanyang Technological University , 639798 , Singapore
- Centre for Biomimetic Sensor Science , Nanyang Technological University , 637553 , Singapore
| | - Tun Naw Sut
- School of Materials Science and Engineering , Nanyang Technological University , 639798 , Singapore
- Centre for Biomimetic Sensor Science , Nanyang Technological University , 637553 , Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering , Nanyang Technological University , 639798 , Singapore
- Centre for Biomimetic Sensor Science , Nanyang Technological University , 637553 , Singapore
- School of Chemical and Biomedical Engineering , Nanyang Technological University , 637459 , Singapore
| |
Collapse
|
1183
|
Berry BJ, Smith AST, Long CJ, Martin CC, Hickman JJ. Physiological Aβ Concentrations Produce a More Biomimetic Representation of the Alzheimer's Disease Phenotype in iPSC Derived Human Neurons. ACS Chem Neurosci 2018; 9:1693-1701. [PMID: 29746089 DOI: 10.1021/acschemneuro.8b00067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by slow, progressive neurodegeneration leading to severe neurological impairment, but current drug development efforts are limited by the lack of robust, human-based disease models. Amyloid-β (Aβ) is known to play an integral role in AD progression as it has been shown to interfere with neurological function. However, studies into AD pathology commonly apply Aβ to neurons for short durations at nonphysiological concentrations to induce an exaggerated dysfunctional phenotype. Such methods are unlikely to elucidate early stage disease dysfunction, when treatment is still possible, since damage to neurons by these high concentrations is extensive. In this study, we investigated chronic, pathologically relevant Aβ oligomer concentrations to induce an electrophysiological phenotype that is more representative of early AD progression compared to an acute high-dose application in human cortical neurons. The high, acute oligomer dose resulted in severe neuronal toxicity as well as upregulation of tau and phosphorylated tau. Chronic, low-dose treatment produced significant functional impairment without increased cell death or accumulation of tau protein. This in vitro phenotype more closely mirrors the status of early stage neural decline in AD pathology and could provide a valuable tool to further understanding of early stage AD pathophysiology and for screening potential therapeutic compounds.
Collapse
Affiliation(s)
- Bonnie J. Berry
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826 United States
| | - Alec S. T. Smith
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826 United States
| | - Christopher J. Long
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826 United States
| | - Candace C. Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826 United States
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826 United States
| |
Collapse
|
1184
|
Seynnaeve D, Vecchio MD, Fruhmann G, Verelst J, Cools M, Beckers J, Mulvihill DP, Winderickx J, Franssens V. Recent Insights on Alzheimer's Disease Originating from Yeast Models. Int J Mol Sci 2018; 19:E1947. [PMID: 29970827 PMCID: PMC6073265 DOI: 10.3390/ijms19071947] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/29/2018] [Accepted: 06/30/2018] [Indexed: 01/28/2023] Open
Abstract
In this review article, yeast model-based research advances regarding the role of Amyloid-β (Aβ), Tau and frameshift Ubiquitin UBB+1 in Alzheimer’s disease (AD) are discussed. Despite having limitations with regard to intercellular and cognitive AD aspects, these models have clearly shown their added value as complementary models for the study of the molecular aspects of these proteins, including their interplay with AD-related cellular processes such as mitochondrial dysfunction and altered proteostasis. Moreover, these yeast models have also shown their importance in translational research, e.g., in compound screenings and for AD diagnostics development. In addition to well-established Saccharomyces cerevisiae models, new upcoming Schizosaccharomyces pombe, Candida glabrata and Kluyveromyces lactis yeast models for Aβ and Tau are briefly described. Finally, traditional and more innovative research methodologies, e.g., for studying protein oligomerization/aggregation, are highlighted.
Collapse
Affiliation(s)
- David Seynnaeve
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Mara Del Vecchio
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Gernot Fruhmann
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Joke Verelst
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Melody Cools
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Jimmy Beckers
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Daniel P Mulvihill
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, Kent, UK.
| | - Joris Winderickx
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| | - Vanessa Franssens
- Functional Biology, KU Leuven, Kasteelpark Arenberg 31, 3000 Leuven, Belgium.
| |
Collapse
|
1185
|
Metaxakis A, Ploumi C, Tavernarakis N. Autophagy in Age-Associated Neurodegeneration. Cells 2018; 7:cells7050037. [PMID: 29734735 PMCID: PMC5981261 DOI: 10.3390/cells7050037] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/23/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
The elimination of abnormal and dysfunctional cellular constituents is an essential prerequisite for nerve cells to maintain their homeostasis and proper function. This is mainly achieved through autophagy, a process that eliminates abnormal and dysfunctional cellular components, including misfolded proteins and damaged organelles. Several studies suggest that age-related decline of autophagy impedes neuronal homeostasis and, subsequently, leads to the progression of neurodegenerative disorders due to the accumulation of toxic protein aggregates in neurons. Here, we discuss the involvement of autophagy perturbation in neurodegeneration and present evidence indicating that upregulation of autophagy holds potential for the development of therapeutic interventions towards confronting neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
| | - Christina Ploumi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion 70013, Crete, Greece.
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Crete, Greece.
- Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion 70013, Crete, Greece.
| |
Collapse
|
1186
|
Secondo A, Bagetta G, Amantea D. On the Role of Store-Operated Calcium Entry in Acute and Chronic Neurodegenerative Diseases. Front Mol Neurosci 2018; 11:87. [PMID: 29623030 PMCID: PMC5874322 DOI: 10.3389/fnmol.2018.00087] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022] Open
Abstract
In both excitable and non-excitable cells, calcium (Ca2+) signals are maintained by a highly integrated process involving store-operated Ca2+ entry (SOCE), namely the opening of plasma membrane (PM) Ca2+ channels following the release of Ca2+ from intracellular stores. Upon depletion of Ca2+ store, the stromal interaction molecule (STIM) senses Ca2+ level reduction and migrates from endoplasmic reticulum (ER)-like sites to the PM where it activates the channel proteins Orai and/or the transient receptor potential channels (TRPC) prompting Ca2+ refilling. Accumulating evidence suggests that SOCE dysregulation may trigger perturbation of intracellular Ca2+ signaling in neurons, glia or hematopoietic cells, thus participating to the pathogenesis of diverse neurodegenerative diseases. Under acute conditions, such as ischemic stroke, neuronal SOCE can either re-establish Ca2+ homeostasis or mediate Ca2+ overload, thus providing a non-excitotoxic mechanism of ischemic neuronal death. The dualistic role of SOCE in brain ischemia is further underscored by the evidence that it also participates to endothelial restoration and to the stabilization of intravascular thrombi. In Parkinson's disease (PD) models, loss of SOCE triggers ER stress and dysfunction/degeneration of dopaminergic neurons. Disruption of neuronal SOCE also underlies Alzheimer's disease (AD) pathogenesis, since both in genetic mouse models and in human sporadic AD brain samples, reduced SOCE contributes to synaptic loss and cognitive decline. Unlike the AD setting, in the striatum from Huntington's disease (HD) transgenic mice, an increased STIM2 expression causes elevated synaptic SOCE that was suggested to underlie synaptic loss in medium spiny neurons. Thus, pharmacological inhibition of SOCE is beneficial to synapse maintenance in HD models, whereas the same approach may be anticipated to be detrimental to cortical and hippocampal pyramidal neurons. On the other hand, up-regulation of SOCE may be beneficial during AD. These intriguing findings highlight the importance of further mechanistic studies to dissect the molecular pathways, and their corresponding targets, involved in synaptic dysfunction and neuronal loss during aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Napoli, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| | - Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, Section of Preclinical and Translational Pharmacology, University of Calabria, Cosenza, Italy
| |
Collapse
|
1187
|
Rahimi F. Aptamers Selected for Recognizing Amyloid β-Protein-A Case for Cautious Optimism. Int J Mol Sci 2018; 19:ijms19030668. [PMID: 29495486 PMCID: PMC5877529 DOI: 10.3390/ijms19030668] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023] Open
Abstract
Aptamers are versatile oligonucleotide ligands used for molecular recognition of diverse targets. However, application of aptamers to the field of amyloid β-protein (Aβ) has been limited so far. Aβ is an intrinsically disordered protein that exists in a dynamic conformational equilibrium, presenting time-dependent ensembles of short-lived, metastable structures and assemblies that have been generally difficult to isolate and characterize. Moreover, despite understanding of potential physiological roles of Aβ, this peptide has been linked to the pathogenesis of Alzheimer disease, and its pathogenic roles remain controversial. Accumulated scientific evidence thus far highlights undesirable or nonspecific interactions between selected aptamers and different Aβ assemblies likely due to the metastable nature of Aβ or inherent affinity of RNA oligonucleotides to β-sheet-rich fibrillar structures of amyloidogenic proteins. Accordingly, lessons drawn from Aβ–aptamer studies emphasize that purity and uniformity of the protein target and rigorous characterization of aptamers’ specificity are important for realizing and garnering the full potential of aptamers selected for recognizing Aβ or other intrinsically disordered proteins. This review summarizes studies of aptamers selected for recognizing different Aβ assemblies and highlights controversies, difficulties, and limitations of such studies.
Collapse
Affiliation(s)
- Farid Rahimi
- Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|