1
|
Mondal R, Deb S, Shome G, Sarkar V, Lahiri D, Datta SS, Benito-León J. Molecular dynamics of amyloid-β transport in Alzheimer's disease: Exploring therapeutic plasma exchange with albumin replacement - Current insights and future perspectives. Neurologia 2025; 40:306-328. [PMID: 40280630 DOI: 10.1016/j.nrleng.2025.03.008] [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: 05/04/2023] [Accepted: 11/07/2023] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The complex process of amyloid-β (Aβ) transportation across the blood-brain and blood-cerebrospinal fluid barriers is crucial for preventing Aβ accumulation, which linked to dementia and neurodegeneration. This review explores therapeutic plasma exchange with albumin replacement in Alzheimer's disease, based on the dynamics of amyloid-β between the brain, plasma, and cerebrospinal fluid. METHODOLOGY A comprehensive literature review was conducted using PubMed/Medline, Cochrane Library, and open databases (bioRxiv, MedRixv, preprint.org) up to April 30, 2023. The first search utilized the following MeSH terms and keywords: 'Plasma Exchange', 'Plasmapheresis', 'Therapeutic plasma exchange', 'Apheresis', 'Aβ', 'p-tau', 'Total-tau', 'Alzheimer's disease', 'Cognitive dysfunction', 'neurodegenerative diseases', 'centrifugation', 'membranous', and 'filtration' in the Title/Abstract, yielding 146 results. A second search with the keywords: 'Albumin', 'Aβ', 'BBB', 'Alzheimer's dementia', and 'Nerve degeneration' resulted in 125 additional articles for analysis. Finally, a third search using keywords: 'Albumin structural domains', 'Albumin-Aβ interactions', 'Albumin-endothelial interactions', and 'Post-Translational Modification' produced 193 results for further review. RESULTS/DISCUSSION Therapeutic plasma exchange shows potential as a disease-modifying therapy for dementia, specifically for Alzheimer's disease. Additionally, the promising role of albumin supplementation in cognitive improvement has attracted attention. However, clinical evidence supporting therapeutic plasma exchange for dementia remains limited, necessitating further research and development to mitigate potential adverse effects. A deeper understanding of the molecular dynamics of Aβ transportation and the mechanisms of therapeutic plasma exchange is essential. A critical evaluation of existing evidence highlights the importance of balancing potential benefits with associated risks, which will guide the development and application of these treatments in neurodegenerative diseases.
Collapse
Affiliation(s)
- R Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - S Deb
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - G Shome
- Department of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - V Sarkar
- Department of Neuroscience, SN Pradhan Center for Neuroscience, University of Calcutta, Kolkata 700019, India
| | - D Lahiri
- Baycrest Academy of Research and Education, Toronto, Canada; Rotman Research Institute, Toronto, Canada; Temerty Faculty of Medicine, University of Toronto, Canada; Department of Neurology, Institute of Neurosciences, Kolkata, India
| | - S S Datta
- Department of Transfusion Medicine, Tata Medical Center, Kolkata 700160, India
| | - J Benito-León
- Department of Neurology, University Hospital "12 de Octubre", Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
2
|
Jain A, Lopus M, Kishore N. From Self-Assembly to Drug Delivery: Understanding and Exploring Protein Fibrils. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:473-495. [PMID: 39745783 DOI: 10.1021/acs.langmuir.4c03745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
It is crucial to comprehend protein misfolding and aggregation in the domains of biomedicine, pharmaceuticals, and proteins. Amyloid fibrils are formed when proteins misfold and assemble, resulting in the debilitating illness known as "amyloidosis". This work investigates lysozyme fibrillation with pluronics (F68 and F127). The effect of pluronics on protein aggregation and fibrillation has been studied mechanistically using a combination of calorimetric and spectroscopic techniques. TEM images and the ThT binding experiment were used to analyze the conformation of protein fibrils, and the results showed that pluronics accelerated the fibrillation process. When pluronics interact with protein at different stages of fibrillation, their pre- and postmicellar concentrations show a decrease in ΔHm° value as the time of incubation increases. This indicates the formation of amorphous aggregates due to which endothermic enthalpy is observed. As a consequence, it was investigated if these generated aggregates can also act as drug delivery vehicle; therefore, the work was carried out with 5-fluorouracil and cytarabine. The endothermic enthalpy of interaction suggests that hydrophobic interaction is more prevalent when cytarabine is employed with protein fibrils, whereas the electrostatic interaction is more prevalent when 5-fluorouracil is combined with it. The former drug, however, showed a greater adsorption than the latter on the surface of protein fibrils. It is therefore determined that 5-fluorouracil has relatively significant adsorption on fibril surfaces, whereas cytarabine has weak adsorption and is easily desorbed in cells. Consequently, the combination of LFF127 and 5-FU is lethal to malignant cells. The drug encapsulation and delivery aspect of protein fibrils/aggregates needs further exploration.
Collapse
Affiliation(s)
- Anu Jain
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai 400098, India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
3
|
Juković M, Ratkaj I, Kalafatovic D, Bradshaw NJ. Amyloids, amorphous aggregates and assemblies of peptides - Assessing aggregation. Biophys Chem 2024; 308:107202. [PMID: 38382283 DOI: 10.1016/j.bpc.2024.107202] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 02/23/2024]
Abstract
Amyloid and amorphous aggregates represent the two major categories of aggregates associated with diseases, and although exhibiting distinct features, researchers often treat them as equivalent, which demonstrates the need for more thorough characterization. Here, we compare amyloid and amorphous aggregates based on their biochemical properties, kinetics, and morphological features. To further decipher this issue, we propose the use of peptide self-assemblies as minimalistic models for understanding the aggregation process. Peptide building blocks are significantly smaller than proteins that participate in aggregation, however, they make a plausible means to bridge the gap in discerning the aggregation process at the more complex, protein level. Additionally, we explore the potential use of peptide-inspired models to research the liquid-liquid phase separation as a feasible mechanism preceding amyloid formation. Connecting these concepts can help clarify our understanding of aggregation-related disorders and potentially provide novel drug targets to impede and reverse these serious illnesses.
Collapse
Affiliation(s)
- Maja Juković
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Ratkaj
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia
| | - Daniela Kalafatovic
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia.
| | - Nicholas J Bradshaw
- Faculty of Biotechnology and Drug Development, University of Rijeka, 51000 Rijeka, Croatia.
| |
Collapse
|
4
|
Polis B, Samson AO. Addressing the Discrepancies Between Animal Models and Human Alzheimer's Disease Pathology: Implications for Translational Research. J Alzheimers Dis 2024; 98:1199-1218. [PMID: 38517793 DOI: 10.3233/jad-240058] [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] [Indexed: 03/24/2024]
Abstract
Animal models, particularly transgenic mice, are extensively used in Alzheimer's disease (AD) research to emulate key disease hallmarks, such as amyloid plaques and neurofibrillary tangles formation. Although these models have contributed to our understanding of AD pathogenesis and can be helpful in testing potential therapeutic interventions, their reliability is dubious. While preclinical studies have shown promise, clinical trials often yield disappointing results, highlighting a notable gap and disparity between animal models and human AD pathology. Existing models frequently overlook early-stage human pathologies and other key AD characteristics, thereby limiting their application in identifying optimal therapeutic interventions. Enhancing model reliability necessitates rigorous study design, comprehensive behavioral evaluations, and biomarker utilization. Overall, a nuanced understanding of each model's neuropathology, its fidelity to human AD, and its limitations is essential for accurate interpretation and successful translation of findings. This article analyzes the discrepancies between animal models and human AD pathology that complicate the translation of findings from preclinical studies to clinical applications. We also delve into AD pathogenesis and attributes to propose a new perspective on this pathology and deliberate over the primary limitations of key experimental models. Additionally, we discuss several fundamental problems that may explain the translational failures and suggest some possible directions for more effective preclinical studies.
Collapse
Affiliation(s)
- Baruh Polis
- Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel
| | | |
Collapse
|
5
|
Zhang O, Haghighatlari M, Li J, Liu ZH, Namini A, Teixeira JMC, Forman-Kay JD, Head-Gordon T. Learning to evolve structural ensembles of unfolded and disordered proteins using experimental solution data. J Chem Phys 2023; 158:174113. [PMID: 37144719 PMCID: PMC10163956 DOI: 10.1063/5.0141474] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/11/2023] [Indexed: 05/06/2023] Open
Abstract
The structural characterization of proteins with a disorder requires a computational approach backed by experiments to model their diverse and dynamic structural ensembles. The selection of conformational ensembles consistent with solution experiments of disordered proteins highly depends on the initial pool of conformers, with currently available tools limited by conformational sampling. We have developed a Generative Recurrent Neural Network (GRNN) that uses supervised learning to bias the probability distributions of torsions to take advantage of experimental data types such as nuclear magnetic resonance J-couplings, nuclear Overhauser effects, and paramagnetic resonance enhancements. We show that updating the generative model parameters according to the reward feedback on the basis of the agreement between experimental data and probabilistic selection of torsions from learned distributions provides an alternative to existing approaches that simply reweight conformers of a static structural pool for disordered proteins. Instead, the biased GRNN, DynamICE, learns to physically change the conformations of the underlying pool of the disordered protein to those that better agree with experiments.
Collapse
Affiliation(s)
- Oufan Zhang
- Kenneth S. Pitzer Theory Center and Department of Chemistry, University of California, Berkeley, California 94720, USA
| | - Mojtaba Haghighatlari
- Kenneth S. Pitzer Theory Center and Department of Chemistry, University of California, Berkeley, California 94720, USA
| | - Jie Li
- Kenneth S. Pitzer Theory Center and Department of Chemistry, University of California, Berkeley, California 94720, USA
| | | | - Ashley Namini
- Molecular Medicine Program, Hospital for Sick Children, Toronto, Ontario M5S 1A8, Canada
| | | | | | | |
Collapse
|
6
|
Judy E, Kishore N. Prevention of insulin fibrillation by biocompatible choline-amino acid based ionic liquids: Biophysical insights. Biochimie 2023; 207:20-32. [PMID: 36471542 DOI: 10.1016/j.biochi.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
We have synthesized biocompatible ionic liquids (ILs) with choline as cation and amino acids as anions to explore their potential towards prevention of fibrillation in insulin and the obtain corresponding mechanistic insights. This has been achieved by examining the effect of these ILs on insulin at the nucleation, elongation and maturation stages of the fibrillation process. A combination of high sensitivity isothermal titration calorimetry (ITC) and differential scanning calorimetry (DSC) have been employed along with spectroscopy and microscopy to evaluate interaction of the ILs at each stage of fibrillation quantitatively. Choline glycinate is observed to provide maximum stabilization to insulin compared to that provided by choline prolinate, choline leucinate, and choline valinate. This increased thermal stabilization has direct correlation with the extent of reduction in the fibrillation of insulin by ILs determined using Thioflavin T and 8-anilinonaphthalene sulfonate based fluorescence assays. ITC has permitted understanding nature of interaction of the ILs with the protein at different fibrillation stages in terms of standard molar enthalpy of interaction whereas DSC has enabled understanding the extent of reduction in thermal stability of the protein at these stages. These ILs are able to completely inhibit formation of insulin aggregates at a concentration of 50 mM. Stabilization of proteins by ILs could be explained based on involvement of preferential hydration process. The work provides biocompatible IL based approach in achieving stability and prevention of fibrillation in insulin.
Collapse
Affiliation(s)
- Eva Judy
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
7
|
AlResaini S, Malik A, Alonazi M, Alhomida A, Khan JM. SDS induces amorphous, amyloid-fibril, and alpha-helical structures in the myoglobin in a concentration-dependent manner. Int J Biol Macromol 2023; 231:123237. [PMID: 36639087 DOI: 10.1016/j.ijbiomac.2023.123237] [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: 11/14/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Amyloid fibrils have been linked to a number of diseases. Surfactants imitate plasma membrane lipids and induce amyloid fibrils. This study examined the effects of the anionic surfactant sodium dodecyl sulfate (SDS) at pH 4.5 on equine skeletal muscle myoglobin (E-Mb). To analyze the effect of SDS on aggregation and amyloid-fibril formation to E-Mb, we used various spectroscopic techniques (turbidity, light scattering, intrinsic fluorescence, ThT fluorescence, and circular dichroism (CD)), electrophoretic, and microscopic techniques. Turbidity, SDS-PAGE, and light scattering all indicated the formation of E-Mb aggregates at SDS concentrations ranging from 0.2 mM to 1.0 mM. In the presence of 0.4 mM SDS, far-UV CD and TEM data indicate that E-MB forms amorphous aggregates. ThT binding, Far-UV CD, and TEM findings indicate that E-Mb forms amyloid-like structures in the presence of 0.6-1.0 mM SDS. However, no aggregation was seen at SDS concentrations above 1 mM. In the presence of high SDS concentrations (> 1 mM), the E-Mb exhibited native-like α-helical structure. As a result, SDS exhibited three distinct behaviors: amorphous aggregates, amyloid-fibrils, and helix-inducer. These findings also shed light on how amyloid fibrils are formed when anionic surfactants are introduced, which is a significant takeaway.
Collapse
Affiliation(s)
- Sundus AlResaini
- Department of Biochemistry, Collage of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ajamaluddin Malik
- Department of Biochemistry, Collage of Science, King Saud University, Riyadh, Saudi Arabia.
| | - Mona Alonazi
- Department of Biochemistry, Collage of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Alhomida
- Department of Biochemistry, Collage of Science, King Saud University, Riyadh, Saudi Arabia
| | - Javed Masood Khan
- Department of Food and Nutrition, Facility of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Sundaria N, Upadhyay A, Prasad A, Prajapati VK, Poluri KM, Mishra A. Neurodegeneration & imperfect ageing: Technological limitations and challenges? Mech Ageing Dev 2021; 200:111574. [PMID: 34562507 DOI: 10.1016/j.mad.2021.111574] [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: 05/30/2021] [Revised: 08/29/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022]
Abstract
Cellular homeostasis is regulated by the protein quality control (PQC) machinery, comprising multiple chaperones and enzymes. Studies suggest that the loss of the PQC mechanisms in neurons may lead to the formation of abnormal inclusions that may lead to neurological disorders and defective aging. The questions could be raised how protein aggregate formation precisely engenders multifactorial molecular pathomechanism in neuronal cells and affects different brain regions? Such questions await thorough investigation that may help us understand how aberrant proteinaceous bodies lead to neurodegeneration and imperfect aging. However, these studies face multiple technological challenges in utilizing available tools for detailed characterizations of the protein aggregates or amyloids and developing new techniques to understand the biology and pathology of proteopathies. The lack of detection and analysis methods has decelerated the pace of the research in amyloid biology. Here, we address the significance of aggregation and inclusion formation, followed by exploring the evolutionary contribution of these structures. We also provide a detailed overview of current state-of-the-art techniques and advances in studying amyloids in the diseased brain. A comprehensive understanding of the structural, pathological, and clinical characteristics of different types of aggregates (inclusions, fibrils, plaques, etc.) will aid in developing future therapies.
Collapse
Affiliation(s)
- Naveen Sundaria
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH‑8 Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology, Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|
9
|
Mak S, Li W, Fu H, Luo J, Cui W, Hu S, Pang Y, Carlier PR, Tsim KW, Pi R, Han Y. Promising tacrine/huperzine A-based dimeric acetylcholinesterase inhibitors for neurodegenerative disorders: From relieving symptoms to modifying diseases through multitarget. J Neurochem 2021; 158:1381-1393. [PMID: 33930191 PMCID: PMC8458250 DOI: 10.1111/jnc.15379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022]
Abstract
Neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease, are devastating diseases in the elderly world, which are closely associated with progressive neuronal loss induced by a variety of genetic and/or environmental factors. Unfortunately, currently available treatments for neurodegenerative disorders can only relieve the symptoms but not modify the pathological processes. Over the past decades, our group by collaborating with Profs. Yuan-Ping Pang and Paul R. Carlier has developed three series of homo/hetero dimeric acetylcholinesterase inhibitors derived from tacrine and/or huperzine A. The representative dimers bis(3)-Cognitin (B3C), bis(12)-hupyridone, and tacrine(10)-hupyridone might possess disease-modifying effects through the modulation of N-methyl-d-aspartic acid receptors, the activation of myocyte enhancer factor 2D gene transcription, and the promotion of neurotrophic factor secretion. In this review, we summarize that the representative dimers, such as B3C, provide neuroprotection against a variety of neurotoxins via multiple targets, including the inhibitions of N-methyl-d-aspartic acid receptor with pathological-activated potential, neuronal nitric oxide synthase, and β-amyloid cascades synergistically. More importantly, B3C might offer disease-modifying potentials by activating myocyte enhancer factor 2D transcription, inducing neuritogenesis, and promoting the expressions of neurotrophic factors in vitro and in vivo. Taken together, the novel dimers might offer synergistic disease-modifying effects, proving that dimerization might serve as one of the strategies to develop new generation of therapeutics for neurodegenerative disorders.
Collapse
Affiliation(s)
- Shinghung Mak
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
- Division of Life Science and Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wenming Li
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, The Ohio State University, Columbus, OH, USA
| | - Jialie Luo
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Wei Cui
- Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo Key Laboratory of Behavioral Neuroscience, School of Medicine, Ningbo University, Ningbo, China
| | - Shengquan Hu
- Shenzhen Institute of Geriatrics, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yuanping Pang
- Mayo Cancer Center, Department of Pharmacology, Mayo Clinic, Rochester, MN, USA
| | | | - Karl Wahkeung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China
- Division of Life Science and Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
10
|
Hanif S, Muhammad P, Niu Z, Ismail M, Morsch M, Zhang X, Li M, Shi B. Nanotechnology‐Based Strategies for Early Diagnosis of Central Nervous System Disorders. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Sumaira Hanif
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Pir Muhammad
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Zheng Niu
- Province's Key Lab of Brain Targeted Bionanomedicine School of Pharmacy Henan University Kaifeng Henan 475004 China
| | - Muhammad Ismail
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Marco Morsch
- Department of Biomedical Sciences Macquarie University Centre for Motor Neuron Disease Research Macquarie University NSW 2109 Australia
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine Henan Provincial People's Hospital Zhengzhou Henan 450003 China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine The Third Affiliated Hospital Sun Yat-sen University Guangzhou Guangdong 510630 China
| | - Bingyang Shi
- Department of Biomedical Sciences Faculty of Medicine & Health & Human Sciences Macquarie University NSW 2109 Australia
| |
Collapse
|
11
|
Niu B, Li M, Jia J, Zhang C, Fan YY, Li W. Hydrophobin-enhanced stability, dispersions and release of curcumin nanoparticles in water. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:1793-1805. [PMID: 32510282 DOI: 10.1080/09205063.2020.1775761] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Most chemotherapeutic drugs commonly suffer from low aqueous solubility that can potentially limit drugs absorption. Drug nanomerization is an advanced approach to overcoming their poor water-solubility. In this study, class I hydrophobin recombinant HGFI (rHGFI)-based curcumin (Cur) nanoparticles (rHGFI-Cur) were prepared by freeze-drying method. The rHGFI-Cur nanocomposites were characterized by contact angle, transmission electron microscopy, fluorescence microscopy and dynamic light scattering. The results showed that rHGFI could lead to the wettability conversion and stability improved of Cur in water. X-ray photoelectron spectroscopy and Fourier transform infrared suggested that rHGFI could non-covalently bind to Cur to render them hydrophilic through hydrophobic forces. Additionally, drug release and cytotoxicity assays illustrated that rHGFI-Cur nanoparticles could facilitate Cur release and exhibited higher cytotoxicity than free Cur for human esophageal cancer cells TE-1. Thus, it suggested that rHGFI has a great potential application for hydrophobic drug delivery without toxicity.[Formula: see text].
Collapse
Affiliation(s)
- Baolong Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, P. R. China.,College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, P. R. China
| | - Meilin Li
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, P. R. China
| | - Jianhong Jia
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, P. R. China
| | - Ce Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, P. R. China
| | - Yan-Ying Fan
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, P. R. China
| | - Wenfeng Li
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan, P. R. China
| |
Collapse
|
12
|
Mujeeb AA, Alam KFB, Alshameri AWF, Jamal F, Farheen S, Kashif M, Ahmed A, Ghazi IA, Owais M. Chaperone Like Attributes of Biogenic Fluorescent Gold Nanoparticles: Potential to Alleviate Toxicity Induced by Intermediate State Fibrils Against Neuroblastoma Cells. Front Chem 2019; 7:787. [PMID: 31799242 PMCID: PMC6878823 DOI: 10.3389/fchem.2019.00787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 11/01/2019] [Indexed: 12/03/2022] Open
Abstract
In general, neurodegenerative disorders have a great deal of correlation with the misfolded as well as aggregated forms of protein-based macromolecules. Among various species formed during the aggregation process, protein oligomers have been classified as most toxic entities against several types of living cells. A series of chemicals have been developed to inhibit protein aggregation as a measure to regulate neurodegenerative diseases. Recently, various classes of nanoparticles have also been reported to inhibit protein aggregation. In the present study, we synthesized fluorescent gold nanoparticles (B-AuNPs) employing Olax scandens leaf extract. Next, an in vitro study was performed to assess the effect of as-synthesized B-AuNPs on the aggregation behavior of the ovalbumin (OVA) and other related model proteins. We performed an extensive study to elucidate anti-amyloidogenic properties of nano-sized entities and established that small-sized B-AuNPs manifest chaperone potential against protein aggregation. Further, we exploited as-synthesized B-AuNPs as a mean to prevent protein aggregation mediated toxicity in neuroblastoma cells.
Collapse
Affiliation(s)
- Anzar Abdul Mujeeb
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | | | | | - Fauzia Jamal
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Saba Farheen
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Mohd Kashif
- CSIR-National Botanical Research Institute, Lucknow, India
| | - Anees Ahmed
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Irfan Ahmad Ghazi
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Mohammad Owais
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
13
|
Training Microglia to Resist Alzheimer's Disease. J Neurosci 2018; 37:477-479. [PMID: 28100733 DOI: 10.1523/jneurosci.3345-16.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/20/2022] Open
|
14
|
Mittal S, Bravo-Rodriguez K, Sanchez-Garcia E. Mechanism of Inhibition of Beta Amyloid Toxicity by Supramolecular Tweezers. J Phys Chem B 2018; 122:4196-4205. [DOI: 10.1021/acs.jpcb.7b10530] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sumit Mittal
- University of Duisburg-Essen, Universitätsstraße 2, 45141 Essen, Germany
| | | | | |
Collapse
|
15
|
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
|
16
|
Malik R, Di J, Nair G, Attar A, Taylor K, Teng E, Klärner FG, Schrader T, Bitan G. Using Molecular Tweezers to Remodel Abnormal Protein Self-Assembly and Inhibit the Toxicity of Amyloidogenic Proteins. Methods Mol Biol 2018; 1777:369-386. [PMID: 29744849 DOI: 10.1007/978-1-4939-7811-3_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Molecular tweezers (MTs) are broad-spectrum inhibitors of abnormal protein self-assembly, which act by binding selectively to lysine and arginine residues. Through this unique mechanism of action, MTs inhibit formation of toxic oligomers and aggregates. Their efficacy and safety have been demonstrated in vitro, in cell culture, and in animal models. Here, we discuss the application of MTs in diverse in vitro and in vivo systems, the experimental details, the scope of their use, and the limitations of the approach. We also consider methods for administration of MTs in animal models to measure efficacy, pharmacokinetic, and pharmacodynamic parameters in proteinopathies.
Collapse
Affiliation(s)
- Ravinder Malik
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jing Di
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Gayatri Nair
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Aida Attar
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA
| | - Karen Taylor
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Edmond Teng
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.,Greater Los Angeles Healthcare System, Veterans Hospital, West Los Angeles, CA, USA
| | | | - Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA. .,Brain Research Institute, University of California at Los Angeles, Los Angeles, CA, USA. .,Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Wolff M, Zhang-Haagen B, Decker C, Barz B, Schneider M, Biehl R, Radulescu A, Strodel B, Willbold D, Nagel-Steger L. Aβ42 pentamers/hexamers are the smallest detectable oligomers in solution. Sci Rep 2017; 7:2493. [PMID: 28559586 PMCID: PMC5449387 DOI: 10.1038/s41598-017-02370-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 04/11/2017] [Indexed: 12/14/2022] Open
Abstract
Amyloid β (Aβ) oligomers may play a decisive role in Alzheimer's disease related neurodegeneration, but their structural properties are poorly understood. In this report, sedimentation velocity centrifugation, small angle neutron scattering (SANS) and molecular modelling were used to identify the small oligomeric species formed by the 42 amino acid residue long isoform of Aβ (Aβ42) in solution, characterized by a sedimentation coefficient of 2.56 S, and a radius of gyration between 2 and 4 nm. The measured sedimentation coefficient is in close agreement with the sedimentation coefficient calculated for Aβ42 hexamers using MD simulations at µM concentration. To the best of our knowledge this is the first report detailing the Aβ42 oligomeric species by SANS measurements. Our results demonstrate that the smallest detectable species in solution are penta- to hexamers. No evidences for the presence of dimers, trimers or tetramers were found, although the existence of those Aβ42 oligomers at measurable quantities had been reported frequently.
Collapse
Affiliation(s)
- Martin Wolff
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
- Physikalische Biochemie, University Potsdam, 14476, Golm, Germany
| | - Bo Zhang-Haagen
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
- Jülich Centre for Neutron Science & Institute of Complex Systems, Neutron Scattering (JCNS-1&ICS-1), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christina Decker
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Bogdan Barz
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Mario Schneider
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
| | - Ralf Biehl
- Jülich Centre for Neutron Science & Institute of Complex Systems, Neutron Scattering (JCNS-1&ICS-1), Forschungszentrum Jülich, 52425, Jülich, Germany
- Jülich Centre for Neutron Science, Outstation at MLZ (JCNS-MLZ), Forschungszentrum Jülich, 85747, Garching, Germany
| | - Aurel Radulescu
- Jülich Centre for Neutron Science, Outstation at MLZ (JCNS-MLZ), Forschungszentrum Jülich, 85747, Garching, Germany
| | - Birgit Strodel
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Luitgard Nagel-Steger
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225, Düsseldorf, Germany.
- Institute of Complex Systems, Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425, Jülich, Germany.
| |
Collapse
|
18
|
Schrader T, Bitan G, Klärner FG. Molecular tweezers for lysine and arginine - powerful inhibitors of pathologic protein aggregation. Chem Commun (Camb) 2016; 52:11318-34. [PMID: 27546596 PMCID: PMC5026632 DOI: 10.1039/c6cc04640a] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Molecular tweezers represent the first class of artificial receptor molecules that have made the way from a supramolecular host to a drug candidate with promising results in animal tests. Due to their unique structure, only lysine and arginine are well complexed with exquisite selectivity by a threading mechanism, which unites electrostatic, hydrophobic and dispersive attraction. However, tweezer design must avoid self-dimerization, self-inclusion and external guest binding. Moderate affinities of molecular tweezers towards sterically well accessible basic amino acids with fast on and off rates protect normal proteins from potential interference with their biological function. However, the early stages of abnormal Aβ, α-synuclein, and TTR assembly are redirected upon tweezer binding towards the generation of amorphous non-toxic materials that can be degraded by the intracellular and extracellular clearance mechanisms. Thus, specific host-guest chemistry between aggregation-prone proteins and lysine/arginine binders rescues cell viability and restores animal health in models of AD, PD, and TTR amyloidosis.
Collapse
Affiliation(s)
- Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, Essen, Germany.
| | | | | |
Collapse
|
19
|
Bourgade K, Dupuis G, Frost EH, Fülöp T. Anti-Viral Properties of Amyloid-β Peptides. J Alzheimers Dis 2016; 54:859-878. [DOI: 10.3233/jad-160517] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Karine Bourgade
- Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gilles Dupuis
- Department of Biochemistry, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric H. Frost
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Tamàs Fülöp
- Department of Medicine, Research Center on Aging, Graduate Program in Immunology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
20
|
Abstract
The aggregation of peptides/proteins is intimately related to a number of human diseases. More than 20 have been identified which aggregate into fibrils containing extensive β-sheet structures, and species generated in the aggregation processes (i.e., oligomers and fibrils) contribute to disease development. Amyloid-β peptide (designated Aβ), related to Alzheimer's disease (AD), is the representative example. The intensive aggregation property of Aβ also leads to difficulty in its synthesis. To improve the synthetic problem, we developed an O-acyl isopeptide of Aβ1-42, in which the N-acyl linkage (amide bond) of Ser(26) was replaced with an O-acyl linkage (ester bond) at the side chain. The O-acyl isopeptide demonstrated markedly higher water-solubility than that of Aβ1-42, while it quickly converted to intact monomer Aβ1-42 via an O-to-N acyl rearrangement under physiological conditions. Inhibition of the pathogenic aggregation of Aβ1-42 might be a therapeutic strategy for curing AD. We succeeded in the rational design and identification of a small molecule aggregation inhibitor based on a pharmacophore motif obtained from cyclo[-Lys-Leu-Val-Phe-Phe-]. Moreover, the inhibition of Aβ aggregation was achieved via oxygenation (i.e., incorporation of oxygen atoms to Aβ) using an artificial catalyst. We identified a selective, cell-compatible photo-oxygenation catalyst of Aβ, a flavin catalyst attached to an Aβ-binding peptide, which markedly decreased the aggregation potency and neurotoxicity of Aβ.
Collapse
Affiliation(s)
- Youhei Sohma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
21
|
Roche J, Shen Y, Lee JH, Ying J, Bax A. Monomeric Aβ(1-40) and Aβ(1-42) Peptides in Solution Adopt Very Similar Ramachandran Map Distributions That Closely Resemble Random Coil. Biochemistry 2016; 55:762-75. [PMID: 26780756 PMCID: PMC4750080 DOI: 10.1021/acs.biochem.5b01259] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
The
pathogenesis of Alzheimer’s disease is characterized
by the aggregation and fibrillation of amyloid peptides Aβ1–40 and Aβ1–42 into amyloid
plaques. Despite strong potential therapeutic interest, the structural
pathways associated with the conversion of monomeric Aβ peptides
into oligomeric species remain largely unknown. In particular, the
higher aggregation propensity and associated toxicity of Aβ1–42 compared to that of Aβ1–40 are poorly understood. To explore in detail the structural propensity
of the monomeric Aβ1–40 and Aβ1–42 peptides in solution, we recorded a large set of nuclear magnetic
resonance (NMR) parameters, including chemical shifts, nuclear Overhauser
effects (NOEs), and J couplings. Systematic comparisons
show that at neutral pH the Aβ1–40 and Aβ1–42 peptides populate almost indistinguishable coil-like
conformations. Nuclear Overhauser effect spectra collected at very
high resolution remove assignment ambiguities and show no long-range
NOE contacts. Six sets of backbone J couplings (3JHNHα, 3JC′C′, 3JC′Hα, 1JHαCα, 2JNCα, and 1JNCα) recorded
for Aβ1–40 were used as input for the recently
developed MERA Ramachandran map analysis, yielding residue-specific
backbone ϕ/ψ torsion angle distributions that closely
resemble random coil distributions, the absence of a significantly
elevated propensity for β-conformations in the C-terminal region
of the peptide, and a small but distinct propensity for αL at K28. Our results suggest that the self-association of
Aβ peptides into toxic oligomers is not driven by elevated propensities
of the monomeric species to adopt β-strand-like conformations.
Instead, the accelerated disappearance of Aβ NMR signals in
D2O over H2O, particularly pronounced for Aβ1–42, suggests that intermolecular interactions between
the hydrophobic regions of the peptide dominate the aggregation process.
Collapse
Affiliation(s)
- Julien Roche
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland 20892-0510, United States
| | - Yang Shen
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland 20892-0510, United States
| | - Jung Ho Lee
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland 20892-0510, United States
| | - Jinfa Ying
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland 20892-0510, United States
| | - Ad Bax
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland 20892-0510, United States
| |
Collapse
|
22
|
SDS-induced Peptide Conformational Changes: From Triglycyl-glycine to Amyloid-β Oligomers Associated with Alzheimer’s Disease. Int J Pept Res Ther 2015. [DOI: 10.1007/s10989-015-9483-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
23
|
Di Natale C, Scognamiglio PL, Cascella R, Cecchi C, Russo A, Leone M, Penco A, Relini A, Federici L, Di Matteo A, Chiti F, Vitagliano L, Marasco D. Nucleophosmin contains amyloidogenic regions that are able to form toxic aggregates under physiological conditions. FASEB J 2015; 29:3689-701. [PMID: 25977257 DOI: 10.1096/fj.14-269522] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/04/2015] [Indexed: 01/08/2023]
Abstract
Nucleophosmin (NPM)-1 is a multifunctional protein involved in a variety of biologic processes and has been implicated in the pathogenesis of several human malignancies. To gain insight into the role of isolated fragments in NPM1 activities, we dissected the C-terminal domain (CTD) into its helical fragments. In this study, we observed the unexpected structural behavior of the peptide fragment corresponding to helix (H)2 (residues 264-277). This peptide has a strong tendency to form amyloidlike assemblies endowed with fibrillar morphology and β-sheet structure, under physiologic conditions, as shown by circular dichroism, thioflavin T, and Congo red binding assays; dynamic light scattering; and atomic force microscopy. The aggregates are also toxic to neuroblastoma cells, as determined using 3-(4;5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction and Ca(2+) influx assays. We also found that the extension of the H2 sequence beyond its N terminus, comprising the connecting loop with H1, delayed aggregation and its associated cytotoxicity, suggesting that contiguous regions of H2 have a protective role in preventing aggregation. Our findings and those in the literature suggest that the helical structures present in the CTD are important in preventing harmful aggregation. These findings could elucidate the pathogenesis of acute myeloid leukemia (AML) caused by NPM1 mutants. Because the CTD is not properly folded in these mutants, we hypothesize that the aggregation propensity of this NPM1 region is involved in the pathogenesis of AML. Preliminary assays on NPM1-Cter-MutA, the most frequent AML-CTD mutation, revealed its significant propensity for aggregation. Thus, the aggregation phenomena should be seriously considered in studies aimed at unveiling the molecular mechanisms of this pathology.
Collapse
Affiliation(s)
- Concetta Di Natale
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Pasqualina Liana Scognamiglio
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Roberta Cascella
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Cristina Cecchi
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Anna Russo
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Marilisa Leone
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Amanda Penco
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Annalisa Relini
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Luca Federici
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Adele Di Matteo
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Fabrizio Chiti
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Luigi Vitagliano
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Daniela Marasco
- *Department of Pharmacy, Diagnostica e Farmaceutica Molecolari-Società Cooperativa a Responsabilità Limitata, Centro Interuniversitario di Ricerca sui Peptidi Bioattivi, University of Naples "Federico II," Naples, Italy; Section of Biochemistry, Department of Biomedical Experimental and Clinical Sciences "Mario Serio," University of Florence, Florence, Italy; Institute of Biostructures and Bioimaging, Consiglio Nazionale delle Ricerche, Naples, Italy; Department of Physics, University of Genoa, Genoa, Italy; Department of Medical, Oral, and Biotechnological Sciences, University of Chieti "G. d'Annunzio," Chieti, Italy; and Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Rome, Italy
| |
Collapse
|
24
|
Uptake of raft components into amyloid β-peptide aggregates and membrane damage. Anal Biochem 2015; 481:18-26. [PMID: 25908557 DOI: 10.1016/j.ab.2015.04.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/13/2015] [Accepted: 04/12/2015] [Indexed: 01/26/2023]
Abstract
Amyloid aggregation and deposition of amyloid β-peptide (Aβ) are pathologic characteristics of Alzheimer's disease (AD). Recent reports have shown that the association of Aβ with membranes containing ganglioside GM1 (GM1) plays a pivotal role in amyloid deposition and the pathogenesis of AD. However, the molecular interactions responsible for membrane damage associated with Aβ deposition are not fully understood. In this study, we microscopically observed amyloid aggregation of Aβ in the presence of lipid vesicles and on a substrate-supported planar membrane containing raft components and GM1. The experimental system enabled us to observe lipid-associated aggregation of Aβ, uptake of the raft components into Aβ aggregates, and relevant membrane damage. The results indicate that uptake of raft components from the membrane into Aβ deposits induces macroscopic heterogeneity of the membrane structure.
Collapse
|
25
|
Zheng X, Liu D, Klärner FG, Schrader T, Bitan G, Bowers MT. Amyloid β-protein assembly: The effect of molecular tweezers CLR01 and CLR03. J Phys Chem B 2015; 119:4831-41. [PMID: 25751170 PMCID: PMC4415044 DOI: 10.1021/acs.jpcb.5b00692] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
The early oligomerization of amyloid
β-protein (Aβ)
has been shown to be an important event in the pathology of Alzheimer’s
disease (AD). Designing small molecule inhibitors targeting Aβ
oligomerization is one attractive and promising strategy for AD treatment.
Here we used ion mobility spectrometry coupled to mass spectrometry
(IMS-MS) to study the different effects of the molecular tweezers
CLR01 and CLR03 on Aβ self-assembly. CLR01 was found to bind
to Aβ directly and disrupt its early oligomerization. Moreover,
CLR01 remodeled the early oligomerization of Aβ42 by compacting
the structures of dimers and tetramers and as a consequence eliminated
higher-order oligomers. Unexpectedly, the negative-control derivative,
CLR03, which lacks the hydrophobic arms of the tweezer structure,
was found to facilitate early Aβ oligomerization. Our study
provides an example of IMS as a powerful tool to study and better
understand the interaction between small molecule modulators and Aβ
oligomerization, which is not attainable by other methods, and provides
important insights into therapeutic development of molecular tweezers
for AD treatment.
Collapse
Affiliation(s)
- Xueyun Zheng
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Deyu Liu
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Frank-Gerrit Klärner
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Thomas Schrader
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Gal Bitan
- §Department of Neurology, David Geffen School of Medicine, Brain Research Institute, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Michael T Bowers
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
26
|
Reale M, Di Nicola M, Velluto L, D'Angelo C, Costantini E, Lahiri DK, Kamal MA, Yu QS, Greig NH. Selective acetyl- and butyrylcholinesterase inhibitors reduce amyloid-β ex vivo activation of peripheral chemo-cytokines from Alzheimer's disease subjects: exploring the cholinergic anti-inflammatory pathway. Curr Alzheimer Res 2015; 11:608-22. [PMID: 24359497 DOI: 10.2174/1567205010666131212113218] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/28/2013] [Accepted: 11/02/2013] [Indexed: 12/22/2022]
Abstract
Increasing evidence suggests that elevated production and/or reduced clearance of amyloid-β peptide (Aβ) drives the early pathogenesis of Alzheimer's disease (AD). Aβ soluble oligomers trigger a neurotoxic cascade that leads to neuronal dysfunction, neurodegeneration and, ultimately, clinical dementia. Inflammation, both within brain and systemically, together with a deficiency in the neurotransmitter acetylcholine (ACh) that underpinned the development of anticholinesterases for AD symptomatic treatment, are invariable hallmarks of the disease. The inter-relation between Aβ, inflammation and cholinergic signaling is complex, with each feeding back onto the others to drive disease progression. To elucidate these interactions plasma samples and peripheral blood mononuclear cells (PBMCs) were evaluated from healthy controls (HC) and AD patients. Plasma levels of acetylcholinesterase (AChE), butyrylcholinesterase (BuChE) and Aβ were significantly elevated in AD vs. HC subjects, and ACh showed a trend towards reduced levels. Aβ challenge of PBMCs induced a greater release of inflammatory cytokines interleukin-1β (IL-1β), monocyte chemotactic protein-1 (MCP-1) and tumor necrosis factor-alpha (TNF-α) from AD vs. HC subjects, with IL-10 being similarly affected. THP-1 monocytic cells, a cell culture counterpart of PBMCs and brain microglial cells, responded similarly to Aβ as well as to phytohaemagglutinin (PHA) challenge, to allow preliminary analysis of the cellular and molecular pathways underpinning Aβ-induced changes in cytokine expression. As amyloid-β precursor protein expression, and hence Aβ, has been reported regulated by particular cytokines and anticholinesterases, the latter were evaluated on Aβ- and PHA-induced chemocytokine expression. Co-incubation with selective AChE/BuChE inhibitors, (-)-phenserine (AChE) and (-)-cymserine analogues (BuChE), mitigated the rise in cytokine levels and suggest that augmentation of the cholinergic anti-inflammatory pathway may prove valuable in AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nigel H Greig
- Dept. of Experimental and Clinical Sciences, Unit ofImmunodiagnostic and Molecular Pathology, University "G. D'Annunzio", N.P.D., Ed. C, III lev., Via dei Vestini, 31, 66123 Chieti, Italy.
| |
Collapse
|
27
|
Yoshiya T, Maruno T, Uemura T, Kubo S, Kiso Y, Sohma Y, Yoshizawa-Kumagaye K, Kobayashi Y, Nishiuchi Y. Non-pretreated O-acyl isopeptide of amyloid β peptide 1-42 is monomeric with a random coil structure but starts to aggregate in a concentration-dependent manner. Bioorg Med Chem Lett 2014; 24:3861-4. [PMID: 25017031 DOI: 10.1016/j.bmcl.2014.06.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 06/17/2014] [Accepted: 06/19/2014] [Indexed: 11/25/2022]
Abstract
An isopeptide of amyloid β peptide 1-42 (isoAβ42) was considered as a non-aggregative precursor molecule for the highly aggregative Aβ42. It has been applied to biological studies after several pretreatments. Here we report that isoAβ42 is monomeric with a random coil structure at 40 μM without any pretreatment. But we also found that isoAβ42 retains a slight aggregative nature, which is significantly weaker than that of the native Aβ42.
Collapse
Affiliation(s)
- Taku Yoshiya
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan.
| | - Takahiro Maruno
- Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Tsuyoshi Uemura
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan
| | - Shigeru Kubo
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan
| | - Yoshiaki Kiso
- Laboratory of Peptide Science, Nagahama Institute of Bio-Science and Technology, Shiga 526-0829, Japan
| | - Youhei Sohma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | | | - Yuji Kobayashi
- Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Yuji Nishiuchi
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki-Shi, Osaka 567-0085, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| |
Collapse
|
28
|
Goure WF, Krafft GA, Jerecic J, Hefti F. Targeting the proper amyloid-beta neuronal toxins: a path forward for Alzheimer's disease immunotherapeutics. ALZHEIMERS RESEARCH & THERAPY 2014; 6:42. [PMID: 25045405 PMCID: PMC4100318 DOI: 10.1186/alzrt272] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Levels of amyloid-beta monomer and deposited amyloid-beta in the Alzheimer’s
disease brain are orders of magnitude greater than soluble amyloid-beta oligomer
levels. Monomeric amyloid-beta has no known direct toxicity. Insoluble fibrillar
amyloid-beta has been proposed to be an in vivo mechanism for removal of
soluble amyloid-beta and exhibits relatively low toxicity. In contrast, soluble
amyloid-beta oligomers are widely reported to be the most toxic amyloid-beta form,
both causing acute synaptotoxicity and inducing neurodegenerative processes. None of
the amyloid-beta immunotherapies currently in clinical development selectively target
soluble amyloid-beta oligomers, and their lack of efficacy is not unexpected
considering their selectivity for monomeric or fibrillar amyloid-beta (or both)
rather than soluble amyloid-beta oligomers. Because they exhibit acute,
memory-compromising synaptic toxicity and induce chronic neurodegenerative toxicity
and because they exist at very low in vivo levels in the Alzheimer’s
disease brain, soluble amyloid-beta oligomers constitute an optimal immunotherapeutic
target that should be pursued more aggressively.
Collapse
Affiliation(s)
- William F Goure
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Grant A Krafft
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Jasna Jerecic
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| | - Franz Hefti
- Acumen Pharmaceuticals, Inc., 4453 North First Street, #360, Livermore, CA 94551, USA
| |
Collapse
|
29
|
Amiri H, Saeidi K, Borhani P, Manafirad A, Ghavami M, Zerbi V. Alzheimer's disease: pathophysiology and applications of magnetic nanoparticles as MRI theranostic agents. ACS Chem Neurosci 2013; 4:1417-29. [PMID: 24024702 PMCID: PMC3837373 DOI: 10.1021/cn4001582] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 09/05/2013] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. During the recent decade, nanotechnology has been widely considered, as a promising tool, for theranosis (diagnosis and therapy) of AD. Here we first discuss pathophysiology and characteristics of AD with a focus on the amyloid cascade hypothesis. Then magnetic nanoparticles (MNPs) and recent works on their applications in AD, focusing on the superparamagnetic iron oxide nanoparticles (SPIONs), are reviewed. Furthermore, the amyloid-nanoparticle interaction is highlighted, with the scope to be highly considered by the scientists aiming for diagnostics and/or treatment of AD employing nanoparticles. Furthermore, recent findings on the "ignored" parameters (e.g., effect of protein "corona" at the surface of nanoparticles on amyloid-β (Aβ) fibrillation process) are discussed.
Collapse
Affiliation(s)
- Houshang Amiri
- Department of Radiology and Department
of Anatomy, Donders Institute for Brain,
Cognition and Behaviour, Radboud University
Nijmegen Medical Centre, 6500HB Nijmegen, The Netherlands
| | - Kolsoum Saeidi
- Department of Medical Genetics and Department of Radiological
Sciences, Kerman University of Medical Sciences, 7618747653 Kerman, Iran
| | - Parvin Borhani
- Department of Medical Genetics and Department of Radiological
Sciences, Kerman University of Medical Sciences, 7618747653 Kerman, Iran
| | - Arash Manafirad
- National Cell Bank, Pasteur Institute of Iran, 13164 Tehran, Iran
| | - Mahdi Ghavami
- National Cell Bank, Pasteur Institute of Iran, 13164 Tehran, Iran
| | - Valerio Zerbi
- Department of Radiology and Department
of Anatomy, Donders Institute for Brain,
Cognition and Behaviour, Radboud University
Nijmegen Medical Centre, 6500HB Nijmegen, The Netherlands
| |
Collapse
|
30
|
Kroker KS, Moreth J, Kussmaul L, Rast G, Rosenbrock H. Restoring long-term potentiation impaired by amyloid-beta oligomers: Comparison of an acetylcholinesterase inhibitior and selective neuronal nicotinic receptor agonists. Brain Res Bull 2013; 96:28-38. [DOI: 10.1016/j.brainresbull.2013.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/12/2013] [Accepted: 04/15/2013] [Indexed: 12/25/2022]
|
31
|
Camilleri A, Zarb C, Caruana M, Ostermeier U, Ghio S, Högen T, Schmidt F, Giese A, Vassallo N. Mitochondrial membrane permeabilisation by amyloid aggregates and protection by polyphenols. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2532-43. [PMID: 23817009 DOI: 10.1016/j.bbamem.2013.06.026] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease and Parkinson's disease are neurodegenerative disorders characterised by the misfolding of proteins into soluble prefibrillar aggregates. These aggregate complexes disrupt mitochondrial function, initiating a pathophysiological cascade leading to synaptic and neuronal degeneration. In order to explore the interaction of amyloid aggregates with mitochondrial membranes, we made use of two in vitro model systems, namely: (i) lipid vesicles with defined membrane compositions that mimic those of mitochondrial membranes, and (ii) respiring mitochondria isolated from neuronal SH-SY5Y cells. External application of soluble prefibrillar forms, but not monomers, of amyloid-beta (Aβ42 peptide), wild-type α-synuclein (α-syn), mutant α-syn (A30P and A53T) and tau-441 proteins induced a robust permeabilisation of mitochondrial-like vesicles, and triggered cytochrome c release (CCR) from isolated mitochondrial organelles. Importantly, the effect on mitochondria was shown to be dependent upon cardiolipin, an anionic phospholipid unique to mitochondria and a well-known key player in mitochondrial apoptosis. Pharmacological modulators of mitochondrial ion channels failed to inhibit CCR. Thus, we propose a generic mechanism of thrilling mitochondria in which soluble amyloid aggregates have the intrinsic capacity to permeabilise mitochondrial membranes, without the need of any other protein. Finally, six small-molecule compounds and black tea extract were tested for their ability to inhibit permeation of mitochondrial membranes by Aβ42, α-syn and tau aggregate complexes. We found that black tea extract and rosmarinic acid were the most potent mito-protectants, and may thus represent important drug leads to alleviate mitochondrial dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Angelique Camilleri
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Moreth J, Mavoungou C, Schindowski K. Passive anti-amyloid immunotherapy in Alzheimer's disease: What are the most promising targets? IMMUNITY & AGEING 2013; 10:18. [PMID: 23663286 PMCID: PMC3681567 DOI: 10.1186/1742-4933-10-18] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 05/01/2013] [Indexed: 12/31/2022]
Abstract
Alzheimer’s disease (AD) is the most common dementia in the industrialized world, with prevalence rates well over 30% in the over 80-years-old population. The dementia causes enormous costs to the social healthcare systems, as well as personal tragedies for the patients, families and caregivers. AD is strongly associated with Amyloid-beta (Aβ) protein aggregation, which results in extracellular plaques in the brain, and according to the amyloid cascade hypothesis appeared to be a promising target for the development of AD therapeutics. Within the past decade convincing data has arisen positioning the soluble prefibrillar Aβ-aggregates as the prime toxic agents in AD. However, different Aβ aggregate species are described but their remarkable metastability hampers the identification of a target species for immunization. Passive immunotherapy with monoclonal antibodies (mAbs) against Aβ is in late clinical development but recently the two most advanced mAbs, Bapineuzumab and Solanezumab, targeting an N-terminal or central epitope, respectively, failed to meet their target of improving or stabilizing cognition and function. Preliminary data from off-label treatment of a small cohort for 3 years with intravenous polyclonal immunoglobulins (IVIG) that appear to target different conformational epitopes indicate a cognitive stabilization. Thus, it might be the more promising strategy reducing the whole spectrum of Aβ-aggregates than to focus on a single aggregate species for immunization.
Collapse
Affiliation(s)
- Jens Moreth
- Institute of Applied Biotechnology, Faculty for Biotechnology, Biberach University of Applied Science, Karlstrasse 11, Biberach/Riss, D-88400, Germany.
| | | | | |
Collapse
|
33
|
Sarafian TA, Ryan CM, Souda P, Masliah E, Kar UK, Vinters HV, Mathern GW, Faull KF, Whitelegge JP, Watson JB. Impairment of mitochondria in adult mouse brain overexpressing predominantly full-length, N-terminally acetylated human α-synuclein. PLoS One 2013; 8:e63557. [PMID: 23667637 PMCID: PMC3646806 DOI: 10.1371/journal.pone.0063557] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 04/02/2013] [Indexed: 12/24/2022] Open
Abstract
While most forms of Parkinson's Disease (PD) are sporadic in nature, a small percentage of PD have genetic causes as first described for dominant, single base pair changes as well as duplication and triplication in the α-synuclein gene. The α-synuclein gene encodes a 140 amino acid residue protein that interacts with a variety of organelles including synaptic vesicles, lysosomes, endoplasmic reticulum/Golgi vesicles and, reported more recently, mitochondria. Here we examined the structural and functional interactions of human α-synuclein with brain mitochondria obtained from an early, pre-manifest mouse model for PD over-expressing human α-synuclein (ASOTg). The membrane potential in ASOTg brain mitochondria was decreased relative to wildtype (WT) mitochondria, while reactive oxygen species (ROS) were elevated in ASOTg brain mitochondria. No selective interaction of human α-synuclein with mitochondrial electron transport complexes cI-cV was detected. Monomeric human α-synuclein plus carboxyl terminally truncated forms were the predominant isoforms detected in ASOTg brain mitochondria by 2-dimensional PAGE (Native/SDS) and immunoblotting. Oligomers or fibrils were not detected with amyloid conformational antibodies. Mass spectrometry of human α-synuclein in both ASOTg brain mitochondria and homogenates from surgically resected human cortex demonstrated that the protein was full-length and postranslationally modified by N-terminal acetylation. Overall the study showed that accumulation of full-length, N-terminally acetylated human α-synuclein was sufficient to disrupt brain mitochondrial function in adult mice.
Collapse
Affiliation(s)
- Theodore A. Sarafian
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Christopher M. Ryan
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Puneet Souda
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Eliezer Masliah
- Department of Neuroscience, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| | - Upendra K. Kar
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Gary W. Mathern
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Kym F. Faull
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Julian P. Whitelegge
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Joseph B. Watson
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
34
|
Amylin uncovered: a review on the polypeptide responsible for type II diabetes. BIOMED RESEARCH INTERNATIONAL 2013; 2013:826706. [PMID: 23607096 PMCID: PMC3626316 DOI: 10.1155/2013/826706] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 02/21/2013] [Indexed: 11/17/2022]
Abstract
Amylin is primarily responsible for classifying type II diabetes as an amyloid (protein misfolding) disease as it has great potential to aggregate into toxic nanoparticles, thereby resulting in loss of pancreatic β-cells. Although type II diabetes is on the increase each year, possibly due to bad eating habits of modern society, research on the culprit for this disease is still in its early days. In addition, unlike the culprit for Alzheimer's disease, amyloid β-peptide, amylin has failed to receive attention worthy of being featured in an abundance of review articles. Thus, the aim of this paper is to shine the spotlight on amylin in an attempt to put it onto the top of researchers' to-do list since the secondary complications of type II diabetes have far-reaching and severe consequences on public health both in developing and fully developed countries alike. This paper will cover characteristics of the amylin aggregates, mechanisms of toxicity, and a particular focus on inhibitors of toxicity and techniques used to assess these inhibitors.
Collapse
|
35
|
Advances in electrochemical detection for study of neurodegenerative disorders. Anal Bioanal Chem 2013; 405:5725-41. [DOI: 10.1007/s00216-013-6904-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 03/05/2013] [Accepted: 03/06/2013] [Indexed: 12/30/2022]
|
36
|
Abstract
O-Acyl isopeptides, in which the N-acyl linkage on the hydroxyamino acid residue (e.g., Ser and Thr) is replaced with an O-acyl linkage, generally possess superior water-solubility to their corresponding native peptides, as well as other distinct physicochemical properties. In addition, O-acyl isopeptides can be rapidly converted into their corresponding native peptide under neutral aqueous conditions through an O-to-N acyl migration. By exploiting these characteristics, researchers have applied the O-acyl isopeptide method to various peptide-synthesis fields, such as the synthesis of aggregative peptides and convergent peptide synthesis. This O-acyl-isopeptide approach also serves as a means to control the biological function of the peptide in question. Herein, we report the synthesis of O-acyl isopeptides and some of their applications.
Collapse
Affiliation(s)
- Youhei Sohma
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Tokyo 113-0033, Japan.
| | | |
Collapse
|
37
|
Sohma Y, Yamasaki M, Kawashima H, Taniguchi A, Yamashita M, Akaji K, Mukai H, Kiso Y. Comparative properties of Aβ1-42, Aβ11-42, and [Pyr¹¹]Aβ11-42 generated from O-acyl isopeptides. Bioorg Med Chem Lett 2013; 23:1326-9. [PMID: 23352512 DOI: 10.1016/j.bmcl.2012.12.082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 12/22/2012] [Accepted: 12/27/2012] [Indexed: 10/27/2022]
Abstract
The use of water-soluble O-acyl isopeptides enabled us to investigate the biochemical properties of Aβ11-42 species, by preparing highly concentrated stock solutions after a pretreatment. Aβ11-42 and [Pyr(11)]Aβ11-42 showed comparable aggregation capability and cytotoxicity, suggesting that the pyroglutamate modification at Glu(11) does not have a crucial role in these events. However, given that Aβ11-42 is converted to [Pyr(11)]Aβ11-42 by a glutamyl cyclase in vivo, the potential aggregative and cytotoxic nature of [Pyr(11)]Aβ11-42 that was observed in the present study provides valuable insights into the pathological functions of pyroglutamate-modified Aβ species in Alzheimer's disease.
Collapse
Affiliation(s)
- Youhei Sohma
- Center for Frontier Research in Medicinal Science, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto 607-8412, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Moreth J, Kroker KS, Schwanzar D, Schnack C, von Arnim CAF, Hengerer B, Rosenbrock H, Kussmaul L. Globular and protofibrillar aβ aggregates impair neurotransmission by different mechanisms. Biochemistry 2013; 52:1466-76. [PMID: 23374097 DOI: 10.1021/bi3016444] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In Alzheimer's disease, substantial evidence indicates the causative role of soluble amyloid β (Aβ) aggregates. Although a variety of Aβ assemblies have been described, the debate about their individual relevance is still ongoing. One critical issue hampering this debate is the use of different methods for the characterization of endogenous and synthetic peptide and their intrinsic limitations for distinguishing Aβ aggregates. Here, we used different protocols for the establishment of prefibrillar Aβ assemblies with varying morphologies and sizes and compared them in a head-to-head fashion. Aggregation was characterized via the monomeric peptide over time until spheroidal, protofibrillar, or fibrillar Aβ aggregates were predominant. It could be shown that a change in the ionic environment induced a structural rearrangement, which consequently confounds the delineation of a measured neurotoxicity toward a distinct Aβ assembly. Here, neuronal binding and hippocampal neurotransmission were found to be suitable to account for the synaptotoxicity to different Aβ assemblies, based on the stability of the applied Aβ aggregates in these settings. In contrast to monomeric or fibrillar Aβ, different prefibrillar Aβ aggregates targeted neurons and impaired hippocampal neurotransmission with nanomolar potency, albeit by different modalities. Spheroidal Aβ aggregates inhibited NMDAR-dependent long-term potentiation, as opposed to protofibrillar Aβ aggregates, which inhibited AMPAR-dominated basal neurotransmission. In addition, a provoked structural conversion of spheroidal to protofibrillar Aβ assemblies resulted in a time-dependent suppression of basal neurotransmission, indicative of a mechanistic switch in synaptic impairment. Thus, we emphasize the importance of addressing the metastability of prefacto characterized Aβ aggregates in assigning a biological effect.
Collapse
Affiliation(s)
- Jens Moreth
- Department of CNS Diseases Research Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse, Biberach an der Riss D-88397, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Platt V, Lee DY, Canaria C, Frankel K, Bernstein S, McMurray C. Towards understanding region-specificity of triplet repeat diseases: coupled immunohistology and mass spectrometry imaging. Methods Mol Biol 2013; 1010:213-30. [PMID: 23754228 PMCID: PMC7191641 DOI: 10.1007/978-1-62703-411-1_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Many trinucleotide repeat disorders exhibit region-specific toxicity within tissues, the basis of which cannot be explained by traditional methods. For example, in Huntington's Disease (HD), the toxic disease-causing protein is ubiquitously expressed. However, only the medium spiny neurons in the striatum are initially targeted for death. Many changes are likely to initiate in these cells at an intracellular and microstructural level long before there is a measureable phenotype, but why some regions of the brain are more susceptible to death is unknown. This chapter describes a method to detect functional changes among brain regions and cell types, and link them directly with region-specific physiology. Due to the neurodegeneration that accompanies many triplet repeat disorders, we focus on the brain, although the methods described in this chapter can be translated to other tissue types. We integrate immunohistology and traditional mass spectrometry with a novel mass spectrometry imaging technique, called nanostructure initiated mass spectrometry (NIMS). When used together, these tools offer unique insights into region-specific physiology of the brain, and a basis for understanding the region-specific toxicity associated with triplet repeat disorders.
Collapse
Affiliation(s)
- Virginia Platt
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720
| | - Do Yup Lee
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720
| | - Christie Canaria
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720
| | - Ken Frankel
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720
| | - Susan Bernstein
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720
| | - Cynthia McMurray
- Lawrence Berkeley National Laboratories, Life Sciences Division, 1 Cyclotron Rd., Berkeley, CA 94720,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic and Foundation, 200 First St., Rochester, MN 55905,Department of Biochemistry and Molecular Biology, Mayo Clinic and Foundation, 200 First St., Rochester, MN 55905,Corresponding authors.
| |
Collapse
|
40
|
Abstract
AbstractAbnormal protein folding and self-assembly causes over 30 cureless human diseases for which no disease-modifying therapies are available. The common side to all these diseases is formation of aberrant toxic protein oligomers and amyloid fibrils. Both types of assemblies are drug targets, yet each presents major challenges to drug design, discovery, and development. In this review, we focus on two small molecules that inhibit formation of toxic amyloid protein assemblies — the green-tea derivative (−)-epigallocatechin-3-gallate (EGCG), which was identified through a combination of epidemiologic data and a compound library screen, and the molecular tweezer CLR01, whose inhibitory activity was discovered in our group based on rational reasoning, and subsequently confirmed experimentally. Both compounds act in a manner that is not specific to one particular protein and thus are useful against a multitude of amyloidogenic proteins, yet they act via distinct putative mechanisms. CLR01 disrupts protein aggregation through specific binding to lysine residues, whereas the mechanisms underlying the activity of EGCG are only recently beginning to unveil. We discuss current in vitro and, where available, in vivo literature related to EGCG and CLR01’s effects on amyloid β-protein, α-synuclein, transthyretin, islet amyloid polypeptide, and calcitonin. We also describe the toxicity, pharmacokinetics, and mechanism of action of each compound.
Collapse
|
41
|
Matveeva EG, Rudolph A, Moll JR, Thompson RB. Structure-selective anisotropy assay for amyloid Beta oligomers. ACS Chem Neurosci 2012. [PMID: 23181170 DOI: 10.1021/cn3001262] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Amyloid β (Abeta) peptides in their oligomeric form have been proposed as major toxic species in Alzheimer's disease (AD). There are a limited number of anti-Abeta antibodies specific to oligomeric forms of Abeta compared to the monomeric form, and accurate measurement of oligomeric forms in biological samples, cerebrospinal fluid (CSF), or brain extracts remains challenging. We introduce an oligomer-specific (in preference to monomers or fibrils) fluorescence assay based on a conformationally sensitive bis-pyrene-labeled peptide that contains amino acid residues 16-35 of the human amyloid beta protein (pronucleon peptide, PP). This peptide exhibits a shift in fluorescence emission from pyrene excimer to pyrene monomer emission resulting from a conformational change. Specific binding of PP to oligomeric forms of Abeta can be monitored in solution by a change in fluorescence spectrum as well as a change in pyrene monomer fluorescence anisotropy (or polarization). The mechanism of binding and its relation to anisotropy and fluorescence lifetime changes are discussed. The development of a simple, rapid, anisotropy assay for measurement of Abeta oligomers is important for further study of the oligomers' role in AD, and specific detection of oligomers in biological samples, such as cerebrospinal fluid.
Collapse
Affiliation(s)
- Evgenia G. Matveeva
- University of Maryland School of Medicine, Department of Biochemistry and Molecular
Biology, Baltimore, Maryland 21201, United States
- Adlyfe, Inc., Rockville,
Maryland 20850, United States
| | - Alan Rudolph
- Adlyfe, Inc., Rockville,
Maryland 20850, United States
| | | | - Richard B. Thompson
- University of Maryland School of Medicine, Department of Biochemistry and Molecular
Biology, Baltimore, Maryland 21201, United States
| |
Collapse
|
42
|
Bruggink KA, Jongbloed W, Biemans EALM, Veerhuis R, Claassen JAHR, Kuiperij HB, Verbeek MM. Amyloid-β oligomer detection by ELISA in cerebrospinal fluid and brain tissue. Anal Biochem 2012; 433:112-20. [PMID: 23022042 DOI: 10.1016/j.ab.2012.09.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 09/11/2012] [Accepted: 09/12/2012] [Indexed: 12/27/2022]
Abstract
Amyloid-β (Aβ) deposits are important pathological hallmarks of Alzheimer's disease (AD). Aβ aggregates into fibrils; however, the intermediate oligomers are believed to be the most neurotoxic species and, therefore, are of great interest as potential biomarkers. Here, we have developed an enzyme-linked immunosorbent assay (ELISA) specific for Aβ oligomers by using the same capture and (labeled) detection antibody. The ELISA predominantly recognizes relatively small oligomers (10-25 kDa) and not monomers. In brain tissue of APP/PS1 transgenic mice, we found that Aβ oligomer levels increase with age. However, for measurements in human samples, pretreatment to remove human anti-mouse antibodies (HAMAs) was required. In HAMA-depleted human hippocampal extracts, the Aβ oligomer concentration was significantly increased in AD compared with nondemented controls. Aβ oligomer levels could also be quantified in pretreated cerebrospinal fluid (CSF) samples; however, no difference was detected between AD and control groups. Our data suggest that levels of small oligomers might not be suitable as biomarkers for AD. In addition, we demonstrate the importance of avoiding HAMA interference in assays to quantify Aβ oligomers in human body fluids.
Collapse
Affiliation(s)
- Kim A Bruggink
- Department of Neurology, Department of Laboratory Medicine, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Centre, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
43
|
Solomonov I, Korkotian E, Born B, Feldman Y, Bitler A, Rahimi F, Li H, Bitan G, Sagi I. Zn2+-Aβ40 complexes form metastable quasi-spherical oligomers that are cytotoxic to cultured hippocampal neurons. J Biol Chem 2012; 287:20555-64. [PMID: 22528492 DOI: 10.1074/jbc.m112.344036] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The roles of metal ions in promoting amyloid β-protein (Aβ) oligomerization associated with Alzheimer disease are increasingly recognized. However, the detailed structures dictating toxicity remain elusive for Aβ oligomers stabilized by metal ions. Here, we show that small Zn(2+)-bound Aβ1-40 (Zn(2+)-Aβ40) oligomers formed in cell culture medium exhibit quasi-spherical structures similar to native amylospheroids isolated recently from Alzheimer disease patients. These quasi-spherical Zn(2+)-Aβ40 oligomers irreversibly inhibit spontaneous neuronal activity and cause massive cell death in primary hippocampal neurons. Spectroscopic and x-ray diffraction structural analyses indicate that despite their non-fibrillar morphology, the metastable Zn(2+)-Aβ40 oligomers are rich in β-sheet and cross-β structures. Thus, Zn(2+) promotes Aβ40 neurotoxicity by structural organization mechanisms mediated by coordination chemistry.
Collapse
Affiliation(s)
- Inna Solomonov
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Motamedi-Shad N, Garfagnini T, Penco A, Relini A, Fogolari F, Corazza A, Esposito G, Bemporad F, Chiti F. Rapid oligomer formation of human muscle acylphosphatase induced by heparan sulfate. Nat Struct Mol Biol 2012; 19:547-54, S1-2. [PMID: 22522822 DOI: 10.1038/nsmb.2286] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 03/22/2012] [Indexed: 11/09/2022]
Abstract
Many human diseases are caused by the conversion of proteins from their native state into amyloid fibrils that deposit in the extracellular space. Heparan sulfate, a component of the extracellular matrix, is universally associated with amyloid deposits and promotes fibril formation. The formation of cytotoxic prefibrillar oligomers is challenging to study because of its rapidity, transient appearance and the heterogeneity of species generated. The process is even more complex with agents such as heparan sulfate. Here we have used a stopped-flow device coupled to turbidometry detection to monitor the rapid conversion of human muscle acylphosphatase into oligomers with varying heparan sulfate and protein concentrations. We also analyzed mutants of the 15 basic amino acids of acylphosphatase, identifying the residues primarily involved in heparan sulfate-induced oligomerization of this protein and tracing the process with unprecedented molecular detail.
Collapse
|
45
|
Zampagni M, Cascella R, Casamenti F, Grossi C, Evangelisti E, Wright D, Becatti M, Liguri G, Mannini B, Campioni S, Chiti F, Cecchi C. A comparison of the biochemical modifications caused by toxic and non-toxic protein oligomers in cells. J Cell Mol Med 2012; 15:2106-16. [PMID: 21155974 PMCID: PMC4394221 DOI: 10.1111/j.1582-4934.2010.01239.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Peptides and proteins can convert from their soluble forms into highly ordered fibrillar aggregates, giving rise to pathological conditions ranging from neurodegenerative disorders to systemic amyloidoses. It is increasingly recognized that protein oligomers forming early in the process of fibril aggregation represent the pathogenic species in protein deposition diseases. The N-terminal domain of the HypF protein from Escherichia coli (HypF-N) has previously been shown to form, under distinct conditions, two types of HypF-N oligomers with indistinguishable morphologies but distinct structural features at the molecular level. Only the oligomer type exposing hydrophobic surfaces and possessing sufficient structural plasticity is toxic (type A), whereas the other type is benign to cultured cells (type B). Here we show that only type A oligomers are able to induce a Ca2+ influx from the cell medium to the cytosol, to penetrate the plasma membrane, to increase intracellular reactive oxygen species production, lipid peroxidation and release of intracellular calcein, resulting in the activation of the apoptotic pathway. Remarkably, these oligomers can also induce a loss of cholinergic neurons when injected into rat brains. By contrast, markers of cellular stress and viability were unaffected in cultured and rat neuronal cells exposed to type B oligomers. The analysis of the time scales of such effects indicates that the difference of toxicity between the two oligomer types involve the early events of the toxicity cascade, shedding new light on the mechanism of action of protein oligomers and on the molecular targets for the therapeutic intervention against protein deposition diseases.
Collapse
|
46
|
Application of photochemical cross-linking to the study of oligomerization of amyloidogenic proteins. Methods Mol Biol 2012; 849:11-21. [PMID: 22528080 DOI: 10.1007/978-1-61779-551-0_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Assembly of amyloidogenic proteins into toxic oligomers and fibrils is an important pathogenic feature of over 30 amyloid-related diseases. Understanding the structures and mechanisms involved in the assembly process is necessary for rational approaches geared at inhibiting formation of these toxic species. Here, we review the application of photo-induced cross-linking of unmodified proteins (PICUP) to two disease-related amyloidogenic proteins (1) islet amyloid polypeptide (IAPP), whose toxic oligomers are thought to cause the demise of pancreatic β-cells in type-2 diabetes mellitus and (2) α-synuclein, which aggregates into toxic oligomers and precipitates in Lewy bodies in Parkinson's disease. PICUP is an effective method allowing chemical "freezing" of dynamically changing oligomers and subsequent study of the oligomer size distribution that existed before cross-linking. The method has provided insights into the factors controlling early oligomerization, which could not be obtained by other means. We discuss sample preparation, experimental details, optimization of parameters, and troubleshooting.
Collapse
|
47
|
Abstract
In this chapter we provided the overall background to the subject of protein aggregation and fibrillogenesis in amyloidogenesis, with introduction and brief discussion of the various topics that are included with the coming chapters. The division of the book into basic science and clinical science sections enables correlation of the topics to be made. The many proteins and peptides that have currently been found to undergo fibrillogenesis are tabulated. A broad technical survey is made, to indicate the vast array of techniques currently available to study aspects of protein oligomerization, aggregation and fibrillogenesis. These are split into three groups and tabulated, as the microscopical techniques, the analytical and biophysical methods, and the biochemical and cellular techniques. A few techniques are discussed, but in most cases only a link to relevant recent literature is provided.
Collapse
|
48
|
Gessel MM, Wu C, Li H, Bitan G, Shea JE, Bowers MT. Aβ(39-42) modulates Aβ oligomerization but not fibril formation. Biochemistry 2011; 51:108-17. [PMID: 22129303 DOI: 10.1021/bi201520b] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recently, certain C-terminal fragments (CTFs) of Aβ42 have been shown to be effective inhibitors of Aβ42 toxicity. Here, we examine the interactions between the shortest CTF in the original series, Aβ(39-42), and full-length Aβ. Mass spectrometry results indicate that Aβ(39-42) binds directly to Aβ monomers and to the n = 2, 4, and 6 oligomers. The Aβ42:Aβ(39-42) complex is further probed using molecular dynamics simulations. Although the CTF was expected to bind to the hydrophobic C-terminus of Aβ42, the simulations show that Aβ(39-42) binds at several locations on Aβ42, including the C-terminus, other hydrophobic regions, and preferentially in the N-terminus. Ion mobility-mass spectrometry (IM-MS) and electron microscopy experiments indicate that Aβ(39-42) disrupts the early assembly of full-length Aβ. Specifically, the ion-mobility results show that Aβ(39-42) prevents the formation of large decamer/dodecamer Aβ42 species and, moreover, can remove these structures from solution. At the same time, thioflavin T fluorescence and electron microscopy results show that the CTF does not inhibit fibril formation, lending strong support to the hypothesis that oligomers and not amyloid fibrils are the Aβ form responsible for toxicity. The results emphasize the role of small, soluble assemblies in Aβ-induced toxicity and suggest that Aβ(39-42) inhibits Aβ-induced toxicity by a unique mechanism, modulating early assembly into nontoxic hetero-oligomers, without preventing fibril formation.
Collapse
Affiliation(s)
- Megan Murray Gessel
- Department of Chemistry and Biochemistry, University of California at Santa Barbara, Santa Barbara, California 93106, United States
| | | | | | | | | | | |
Collapse
|
49
|
Prion protein at the crossroads of physiology and disease. Trends Neurosci 2011; 35:92-103. [PMID: 22137337 DOI: 10.1016/j.tins.2011.10.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 10/20/2011] [Accepted: 10/20/2011] [Indexed: 11/23/2022]
Abstract
The presence of the cellular prion protein (PrP(C)) on the cell surface is critical for the neurotoxicity of prions. Although several biological activities have been attributed to PrP(C), a definitive demonstration of its physiological function remains elusive. In this review, we discuss some of the proposed functions of PrP(C), focusing on recently suggested roles in cell adhesion, regulation of ionic currents at the cell membrane and neuroprotection. We also discuss recent evidence supporting the idea that PrP(C) may function as a receptor for soluble oligomers of the amyloid β peptide and possibly other toxic protein aggregates. These data suggest surprising new connections between the physiological function of PrP(C) and its role in neurodegenerative diseases beyond those caused by prions.
Collapse
|
50
|
Li H, Du Z, Lopes DHJ, Fradinger EA, Wang C, Bitan G. C-terminal tetrapeptides inhibit Aβ42-induced neurotoxicity primarily through specific interaction at the N-terminus of Aβ42. J Med Chem 2011; 54:8451-60. [PMID: 22087474 DOI: 10.1021/jm200982p] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibition of amyloid β-protein (Aβ)-induced toxicity is a promising therapeutic strategy for Alzheimer's disease (AD). Previously, we reported that the C-terminal tetrapeptide Aβ(39-42) is a potent inhibitor of neurotoxicity caused by Aβ42, the form of Aβ most closely associated with AD. Here, initial structure-activity relationship studies identified key structural requirements, including chirality, side-chain structure, and a free N-terminus, which control Aβ(39-42) inhibitory activity. To elucidate the binding site(s) of Aβ(39-42) on Aβ42, we used intrinsic tyrosine (Y) fluorescence and solution-state NMR. The data suggest that Aβ(39-42) binds at several sites, of which the predominant one is located in the N-terminus of Aβ42, in agreement with recent modeling predictions. Thus, despite the small size of Aβ(39-42) and the hydrophobic, aliphatic nature of all four side-chains, the interaction of Aβ(39-42) with Aβ42 is controlled by specific intermolecular contacts requiring a combination of hydrophobic and electrostatic interactions and a particular stereochemistry.
Collapse
Affiliation(s)
- Huiyuan Li
- Department of Neurology, David Geffen School of Medicine, Brain Research Institute, University of California, Los Angeles, 635 Charles E. Young Drive South, Los Angeles, California 90095-7334, United States
| | | | | | | | | | | |
Collapse
|