1
|
Naranda J, Bračič M, Vogrin M, Maver U, Trojner T. Practical Use of Quartz Crystal Microbalance Monitoring in Cartilage Tissue Engineering. J Funct Biomater 2022; 13:jfb13040159. [PMID: 36278628 PMCID: PMC9590066 DOI: 10.3390/jfb13040159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022] Open
Abstract
Quartz crystal microbalance (QCM) is a real-time, nanogram-accurate technique for analyzing various processes on biomaterial surfaces. QCM has proven to be an excellent tool in tissue engineering as it can monitor key parameters in developing cellular scaffolds. This review focuses on the use of QCM in the tissue engineering of cartilage. It begins with a brief discussion of biomaterials and the current state of the art in scaffold development for cartilage tissue engineering, followed by a summary of the potential uses of QCM in cartilage tissue engineering. This includes monitoring interactions with extracellular matrix components, adsorption of proteins onto biomaterials, and biomaterial–cell interactions. In the last part of the review, the material selection problem in tissue engineering is highlighted, emphasizing the importance of surface nanotopography, the role of nanofilms, and utilization of QCM as a “screening” tool to improve the material selection process. A step-by-step process for scaffold design is proposed, as well as the fabrication of thin nanofilms in a layer-by-layer manner using QCM. Finally, future trends of QCM application as a “screening” method for 3D printing of cellular scaffolds are envisioned.
Collapse
Affiliation(s)
- Jakob Naranda
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
- Department of Orthopaedics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
- Correspondence: (J.N.); (M.B.); Tel.: +386-2-321-1541 (J.N.); +386-2-220-7929 (M.B.)
| | - Matej Bračič
- Laboratory for Characterisation and Processing of Polymers (LCPP), Faculty of Mechanical Engineering, University of Maribor, SI-2000 Maribor, Slovenia
- Correspondence: (J.N.); (M.B.); Tel.: +386-2-321-1541 (J.N.); +386-2-220-7929 (M.B.)
| | - Matjaž Vogrin
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
- Department of Orthopaedics, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
- Department of Pharmacology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia
| | - Teodor Trojner
- Department of Orthopaedics, University Medical Centre Maribor, SI-2000 Maribor, Slovenia
| |
Collapse
|
2
|
Multifaceted neuroprotective effects of (-)-epigallocatechin-3-gallate (EGCG) in Alzheimer’s disease: an overview of pre-clinical studies focused on β-amyloid peptide. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
3
|
Al Adem K, Shanti A, Srivastava A, Homouz D, Thomas SA, Khair M, Stefanini C, Chan V, Kim TY, Lee S. Linking Alzheimer’s Disease and Type 2 Diabetes: Characterization and Inhibition of Cytotoxic Aβ and IAPP Hetero-Aggregates. Front Mol Biosci 2022; 9:842582. [PMID: 35372522 PMCID: PMC8968156 DOI: 10.3389/fmolb.2022.842582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic self-aggregation of β-amyloid (Aβ) peptide and islet amyloid polypeptide (IAPP) is implicated in the pathogenesis of Alzheimer’s disease (AD) and Type 2 diabetes (T2D), respectively. Increasing evidence, particularly the co-deposition of Aβ and IAPP in both brain and pancreatic tissues, suggests that Aβ and IAPP cross-interaction may be responsible for a pathological link between AD and T2D. Here, we examined the nature of IAPP-Aβ40 co-aggregation and its inhibition by small molecules. In specific, we characterized the kinetic profiles, morphologies, secondary structures and toxicities of IAPP-Aβ40 hetero-assemblies and compared them to those formed by their homo-assemblies. We demonstrated that monomeric IAPP and Aβ40 form stable hetero-dimers and hetero-assemblies that further aggregate into β-sheet-rich hetero-aggregates that are toxic (cell viability <50%) to both PC-12 cells, a neuronal cell model, and RIN-m5F cells, a pancreatic cell model for β-cells. We then selected polyphenolic candidates to inhibit IAPP or Aβ40 self-aggregation and examined the inhibitory effect of the most potent candidate on IAPP-Aβ40 co-aggregation. We demonstrated that epigallocatechin gallate (EGCG) form inter-molecular hydrogen bonds with each of IAPP and Aβ40. We also showed that EGCG reduced hetero-aggregate formation and resulted in lower β-sheets content and higher unordered structures in IAPP-Aβ40-EGCG samples. Importantly, we showed that EGCG is highly effective in reducing the toxicity of IAPP-Aβ40 hetero-aggregates on both cell models, specifically at concentrations that are equivalent to or are 2.5-fold higher than the mixed peptide concentrations. To the best of our knowledge, this is the first study to report the inhibition of IAPP-Aβ40 co-aggregation by small molecules. We conclude that EGCG is a promising candidate to prevent co-aggregation and cytotoxicity of IAPP-Aβ40, which in turn, contribute to the pathological link between AD and T2D.
Collapse
Affiliation(s)
- Kenana Al Adem
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Aya Shanti
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Amit Srivastava
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dirar Homouz
- Department of Physics, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Physics, University of Houston, Houston, TX, United States
- Center for Theoretical Biological Physics, Rice University, Houston, TX, United States
| | - Sneha Ann Thomas
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Tae-Yeon Kim
- Department of Civil Infrastructure and Environmental Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Sungmun Lee
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Khalifa University’s Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- *Correspondence: Sungmun Lee,
| |
Collapse
|
4
|
Ho T, Ahmadi S, Kerman K. Do glutathione and copper interact to modify Alzheimer's disease pathogenesis? Free Radic Biol Med 2022; 181:180-196. [PMID: 35092854 DOI: 10.1016/j.freeradbiomed.2022.01.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder first described in 1906 that is currently estimated to impact ∼40 million people worldwide. Extensive research activities have led to a wealth of information on the pathogenesis, hallmarks, and risk factors of AD; however, therapeutic options remain extremely limited. The large number of pathogenic factors that have been reported to potentially contribute to AD include copper dyshomeostasis as well as increased oxidative stress, which is related to alterations to molecular antioxidants like glutathione (GSH). While the individual roles of GSH and copper in AD have been studied by many research groups, their interactions have received relatively little attention, although they appear to interact and affect each other's regulation. Existing knowledge on how GSH-copper interactions may affect AD is sparse and lacks focus. This review first highlights the most relevant individual roles that GSH and copper play in physiology and AD, and then collects and assesses research concerning their interactions, in an effort to provide a more accessible and understandable picture of the role of GSH, copper, and their interactions in AD.
Collapse
Affiliation(s)
- Talia Ho
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| | - Soha Ahmadi
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada.
| | - Kagan Kerman
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1265 Military Trail, Toronto, ON, M1C 1A4, Canada; Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada.
| |
Collapse
|
5
|
Wang MD, Hou DY, Lv GT, Li RX, Hu XJ, Wang ZJ, Zhang NY, Yi L, Xu WH, Wang H. Targeted in situ self-assembly augments peptide drug conjugate cell-entry efficiency. Biomaterials 2021; 278:121139. [PMID: 34624753 DOI: 10.1016/j.biomaterials.2021.121139] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/26/2021] [Accepted: 09/18/2021] [Indexed: 11/26/2022]
Abstract
Peptide drug conjugate (PDC) has emerged as one of the new generations of targeted therapeutics for cancer, which owns the advantages of improved drug targetability and reduced adverse effects compared with traditional chemotherapy. However, the poor permeability of PDC drugs regarding tumor cells is an urgent problem to be solved. Herein, we design a PDC drug molecule, which is composed of three modules: targeting motif (RGD target), assembly motif (GNNNQNY) and cytotoxic payload (CPT molecule). This PDC in situ forms nanoclusters upon binding cellular receptor, resulting in improved PDC cell-entry efficiency and treatment efficacy. In addition, the PDC shows increased therapeutic efficacy and raises the maximum tolerance dose of the drug in breast and bladder xenografted mice models. This strategy leverages the assembly principle to promote penetration of peptide molecules into cells and increase intracellular drug bioavailability, which is of great significance for the development of PDC drugs in the future.
Collapse
Affiliation(s)
- Man-Di Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Gan-Tian Lv
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ru-Xiang Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Xing-Jie Hu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China
| | - Zhi-Jia Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Ni-Yuan Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Li Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Wan-Hai Xu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, 100190, Beijing, China; Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China; Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Park S, Yi Y, Lim MH. Reactivity of Flavonoids Containing a Catechol or Pyrogallol Moiety with Metal‐Free and Metal‐Associated Amyloid‐β. B KOREAN CHEM SOC 2020. [DOI: 10.1002/bkcs.12172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Seongmin Park
- Department of Chemistry Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Yelim Yi
- Department of Chemistry Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
7
|
Rezabakhsh A, Rahbarghazi R, Fathi F. Surface plasmon resonance biosensors for detection of Alzheimer's biomarkers; an effective step in early and accurate diagnosis. Biosens Bioelectron 2020; 167:112511. [PMID: 32858422 DOI: 10.1016/j.bios.2020.112511] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
The rapid and direct detection of biomarkers in biofluids at clinically relevant concentrations faces serious limitations to develop diagnostic criteria for neurodegenerative diseases such as Alzheimer's disease (AD). In this regard, the early detection of biomarkers correlated with AD using novel modalities and instruments is at the center of attention. Recently, some newly invented optical-based biosensors namely Surface Plasmon Resonance (SPR) has been extensively investigated for the detection of biomarkers using a label-free method or by checking interaction between ligand and analyte. These approaches can sense a very small amount of target molecules in the blood and cerebrospinal fluids samples. In this review, the different hypothesis related to AD, and the structural properties of AD biomarkers was introduced. Also, we aim to highlight the specific role of available SPR-based sensing methods for early detection of AD biomarkers such as aggregated β-amyloid and tau proteins. Efforts to better understand the accuracy and efficiency of optical-based biosensors in the field of neurodegenerative disease enable us to accelerate the advent of novel modalities in the clinical setting for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Emergency Medicine Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Fathi
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Biosensor Sciences and Technologies Research Center (BSTRC), Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
8
|
Jamerlan A, An SSA, Hulme J. Advances in amyloid beta oligomer detection applications in Alzheimer's disease. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
9
|
Pereira M, Marques AC, Oliveira D, Martins R, Moreira FTC, Sales MGF, Fortunato E. Paper-Based Platform with an In Situ Molecularly Imprinted Polymer for β-Amyloid. ACS OMEGA 2020; 5:12057-12066. [PMID: 32548384 PMCID: PMC7271027 DOI: 10.1021/acsomega.0c00062] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/19/2020] [Indexed: 05/23/2023]
Abstract
Alzheimer's disease (AD) is one of the most common forms of dementia affecting millions of people worldwide. Currently, an easy and effective form of diagnosis is missing, which significantly hinders a possible improvement of the patient's quality of life. In this context, biosensors emerge as a future solution, opening the doors for preventive medicine and allowing the premature diagnosis of numerous pathologies. This work presents a pioneering biosensor that combines a bottom-up design approach using paper as a platform for the electrochemical recognition of peptide amyloid β-42 (Aβ-42), a biomarker for AD present in blood, associated with visible differences in the brain tissue and responsible for the formation of senile plaques. The sensor layer relies on a molecularly imprinted polymer as a biorecognition element, created on the carbon ink electrode's surface by electropolymerizing a mixture of the target analyte (Aβ-42) and a monomer (O-phenylenediamine) at neutral pH 7.2. Next, the template molecule was removed from the polymeric network by enzymatic and acidic treatments. The vacant sites so obtained preserved the shape of the imprinted protein and were able to rebind the target analyte. Morphological and chemical analyses were performed in order to control the surface modification of the materials. The analytical performance of the biosensor was evaluated by an electroanalytical technique, namely, square wave voltammetry. For this purpose, the analytical response of the biosensor was tested with standard solutions ranging from 0.1 ng/mL to 1 μg/mL of Aβ-42. The linear response of the biosensor went down to 0.1 ng/mL. Overall, the developed biosensor offered numerous benefits, such as simplicity, low cost, reproducibility, fast response, and repeatability less than 10%. All together, these features may have a strong impact in the early detection of AD.
Collapse
Affiliation(s)
- Marta
V. Pereira
- BioMark,
Sensor Research/ISEP, School of Engineering, Polytechnic Institute
Porto 4249-015 Porto, Portugal
- CEB,
Centre of Biological Engineering Minho University, 4710-957 Braga, Portugal
- i3N/CENIMAT,
Department of Materials Science, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Ana C. Marques
- BioMark,
Sensor Research/ISEP, School of Engineering, Polytechnic Institute
Porto 4249-015 Porto, Portugal
- i3N/CENIMAT,
Department of Materials Science, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Daniela Oliveira
- BioMark,
Sensor Research/ISEP, School of Engineering, Polytechnic Institute
Porto 4249-015 Porto, Portugal
- CEB,
Centre of Biological Engineering Minho University, 4710-957 Braga, Portugal
| | - Rodrigo Martins
- i3N/CENIMAT,
Department of Materials Science, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Felismina T. C. Moreira
- BioMark,
Sensor Research/ISEP, School of Engineering, Polytechnic Institute
Porto 4249-015 Porto, Portugal
- CEB,
Centre of Biological Engineering Minho University, 4710-957 Braga, Portugal
| | - M. Goreti F. Sales
- BioMark,
Sensor Research/ISEP, School of Engineering, Polytechnic Institute
Porto 4249-015 Porto, Portugal
- CEB,
Centre of Biological Engineering Minho University, 4710-957 Braga, Portugal
| | - Elvira Fortunato
- i3N/CENIMAT,
Department of Materials Science, Faculty of Science and Technology, Universidade NOVA de Lisboa and CEMOP/UNINOVA, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
10
|
Effects of Cu(II) on the aggregation of amyloid-β. J Biol Inorg Chem 2019; 24:1197-1215. [PMID: 31602542 DOI: 10.1007/s00775-019-01727-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/01/2019] [Indexed: 12/27/2022]
Abstract
Aberrant aggregation of the Aβ protein is a hallmark of Alzheimer's disease (AD), but no complete characterization of the molecular level pathogenesis has been achieved. A promising hypothesis is that dysfunction of metal ion homeostasis, and consequently, the undesired interaction of metal ions with Aβ, may be central to the development of AD. Qualitatively, most data indicate that Cu(II) induces rapid self-assembly of both Aβ40 and Aβ42 during the initial phase of the aggregation, while at longer time scales fibrillation may occur, depending on the experimental conditions. For Aβ40 and Cu(II):Aβ ≤ 1, most data imply that low concentration of Aβ40 favors nucleation and rapid fibril elongation, while high concentration of Aβ40 favors formation of amorphous aggregates. However, there are conflicting reports on this issue. For Aβ42 and Cu(II):Aβ ≤ 1, there is consensus that the lag time is extended upon addition of Cu(II). For Cu(II):Aβ > 1, the lag time is increased upon interaction with Cu(II), and in most cases fibrillation is not observed, presumably because Cu(II) occupies a second more solvent-exposed binding site, which is more prone to form metal ion-bridged species and cause rapid formation of non-fibrillar aggregates. The interesting N-terminally truncated Aβ11-40 with high affinity for Cu(II), exhibits delay of fibrillation upon addition of 0.4 eq. Cu(II). In our view, there are still problems achieving reproducible results in this field, and we provide a shortlist of some of the pitfalls. Finally, we propose a consensus model for the effects of Cu(II) on the aggregation kinetics of Aβ.
Collapse
|
11
|
Bocková M, Slabý J, Špringer T, Homola J. Advances in Surface Plasmon Resonance Imaging and Microscopy and Their Biological Applications. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:151-176. [PMID: 30822102 DOI: 10.1146/annurev-anchem-061318-115106] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Surface plasmon resonance microscopy and imaging are optical methods that enable observation and quantification of interactions of nano- and microscale objects near a metal surface in a temporally and spatially resolved manner. This review describes the principles of surface plasmon resonance microscopy and imaging and discusses recent advances in these methods, in particular, in optical platforms and functional coatings. In addition, the biological applications of these methods are reviewed. These include the detection of a broad variety of analytes (nucleic acids, proteins, bacteria), the investigation of biological systems (bacteria and cells), and biomolecular interactions (drug-receptor, protein-protein, protein-DNA, protein-cell).
Collapse
Affiliation(s)
- Markéta Bocková
- Institute of Photonics and Electronics, Czech Academy of Sciences, 18251 Prague, Czech Republic;
| | - Jiří Slabý
- Institute of Photonics and Electronics, Czech Academy of Sciences, 18251 Prague, Czech Republic;
| | - Tomáš Špringer
- Institute of Photonics and Electronics, Czech Academy of Sciences, 18251 Prague, Czech Republic;
| | - Jiří Homola
- Institute of Photonics and Electronics, Czech Academy of Sciences, 18251 Prague, Czech Republic;
| |
Collapse
|
12
|
Biosensors for the Detection of Interaction between Legionella pneumophila Collagen-Like Protein and Glycosaminoglycans. SENSORS 2018; 18:s18082668. [PMID: 30110899 PMCID: PMC6111780 DOI: 10.3390/s18082668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 01/18/2023]
Abstract
The adhesin Legionella collagen-like (Lcl) protein can bind to extracellular matrix components and mediate the binding of Legionella pneumophila to host cells. In this study, electrochemical impedance spectroscopy (EIS) and surface plasmon resonance (SPR)-based biosensors were employed to characterize these interactions between glycosaminoglycans (GAGs) and the adhesin Lcl protein. Fucoidan displayed a high affinity (KD 18 nM) for Lcl protein. Chondroitin sulfate A and dermatan sulfate differ in the position of a carboxyl group replacing D-glucuronate with D-iduronate. Our results indicated that the presence of D-iduronate in dermatan sulfate strongly hindered its interaction with Lcl. These biophysical studies provided valuable information in our understanding of adhesin-ligand interactions related to Legionella pneumophila infections.
Collapse
|
13
|
Savelieff MG, Nam G, Kang J, Lee HJ, Lee M, Lim MH. Development of Multifunctional Molecules as Potential Therapeutic Candidates for Alzheimer’s Disease, Parkinson’s Disease, and Amyotrophic Lateral Sclerosis in the Last Decade. Chem Rev 2018; 119:1221-1322. [DOI: 10.1021/acs.chemrev.8b00138] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Masha G. Savelieff
- SciGency Science Communications, Ann Arbor, Michigan 48104, United States
| | - Geewoo Nam
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Juhye Kang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Misun Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
14
|
Ganesh HVS, Noroozifar M, Kerman K. Epigallocatechin Gallate-Modified Graphite Paste Electrode for Simultaneous Detection of Redox-Active Biomolecules. SENSORS 2017; 18:s18010023. [PMID: 29271930 PMCID: PMC5795891 DOI: 10.3390/s18010023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/07/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
In this study, simultaneous electrochemical detection of ascorbic acid (AA), dopamine (DA), and uric acid (UA) was performed using a modified graphite paste electrode (MGPE) with epigallocatechin gallate (EGCG) and green tea (GT) powder. It was shown that the anodic peak current increased in comparison with that of the graphite paste electrode (GPE) in the cyclic voltammograms. The optimal pH for simultaneous determination of a quaternary mixture of AA–DA–UA was determined to be pH 2. The anodic peak potentials for a mixture containing AA–DA–UA were well separated from each other. The catalytic peak currents obtained at the surface of the MGPE/EGCG were linearly dependent on the AA, DA, and UA concentrations up to 23, 14, and 14 µM, respectively. The detection limits for AA, DA, and UA were 190, 90, and 70 nM, respectively. The analytical performance of this sensor has been evaluated for simultaneous detection of AA, DA, and UA in real samples. Finally, a modified electrode was prepared using GT and used for simultaneous determination of AA, DA, and UA. Based on the results, MPGE/GT showed two oxidation peaks at 0.43 and 0.6 V for DA and UA, respectively, without any oxidation peak for AA. The calibration curves at the surface of MGPE/GT were linear up to 14 µM with a detection limit of 0.18 and 0.33 µM for DA and UA, respectively. MGPEs provide a promising platform for the future development of sensors for multiplexed electrochemical detection of clinically important analytes.
Collapse
Affiliation(s)
- Hashwin V S Ganesh
- Department of Physical and Environmental Sciences, University of Toronto, Scarborough 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| | - Meissam Noroozifar
- Analytical Research Laboratory, Department of Chemistry, University of Sistan and Baluchestan, P.O. Box 98135-674, Zahedan, Iran.
| | - Kagan Kerman
- Department of Physical and Environmental Sciences, University of Toronto, Scarborough 1265 Military Trail, Toronto, ON M1C 1A4, Canada.
| |
Collapse
|
15
|
Yoo YK, Kim J, Kim G, Kim YS, Kim HY, Lee S, Cho WW, Kim S, Lee SM, Lee BC, Lee JH, Hwang KS. A highly sensitive plasma-based amyloid-β detection system through medium-changing and noise cancellation system for early diagnosis of the Alzheimer's disease. Sci Rep 2017; 7:8882. [PMID: 28827785 PMCID: PMC5567090 DOI: 10.1038/s41598-017-09370-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/24/2017] [Indexed: 01/17/2023] Open
Abstract
We developed an interdigitated microelectrode (IME) sensor system for blood-based Alzheimer's disease (AD) diagnosis based on impedimetric detection of amyloid-β (Aβ) protein, which is a representative candidate biomarker for AD. The IME sensing device was fabricated using a surface micromachining process. For highly sensitive detection of several tens to hundreds of picogram/mL of Aβ in blood, medium change from plasma to PBS buffer was utilized with signal cancellation and amplification processing (SCAP) system. The system demonstrated approximately 100-folds higher sensitivity according to the concentrations. A robust antibody-immobilization process was used for stability during medium change. Selectivity of the reaction due to the affinity of Aβ to the antibody and the sensitivity according to the concentration of Aβ were also demonstrated. Considering these basic characteristics of the IME sensor system, the medium change was optimized in relation to the absolute value of impedance change and differentiated impedance changes for real plasma based Aβ detection. Finally, the detection of Aβ levels in transgenic and wild-type mouse plasma samples was accomplished with the designed sensor system and the medium-changing method. The results confirmed the potential of this system to discriminate between patients and healthy controls, which would enable blood-based AD diagnosis.
Collapse
Affiliation(s)
- Yong Kyoung Yoo
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, 02447, Korea
- Department of Electrical Engineering, Kwangwoon University, Seoul, 139-701, South Korea
| | - Jinsik Kim
- Department of Medical Biotechnology, College of Life Science and Biotechnology, Dongguk University, Seoul, Korea
| | - Gangeun Kim
- Center for BioMicrosystems, Korea Institute of Science and Technology (KIST), Seoul, 136-791, South Korea
| | - Young Soo Kim
- Department of Pharmacy & Integrated Science and Engineering Division, Yonsei University, Incheon, 21983, South Korea
| | - Hye Yun Kim
- Department of Pharmacy & Integrated Science and Engineering Division, Yonsei University, Incheon, 21983, South Korea
| | - Sejin Lee
- Department of Pharmacy & Integrated Science and Engineering Division, Yonsei University, Incheon, 21983, South Korea
| | - Won Woo Cho
- CANTIS.co, Sangnok-gu, Ansan-si, Gyeonggi-do, 426-901, South Korea
| | - Seongsoo Kim
- Department of Chemical Engineering, Kangwon National University, Gangwon-do, 200-701, South Korea
| | - Sang-Myung Lee
- Department of Chemical Engineering, Kangwon National University, Gangwon-do, 200-701, South Korea
| | - Byung Chul Lee
- Center for BioMicrosystems, Korea Institute of Science and Technology (KIST), Seoul, 136-791, South Korea
| | - Jeong Hoon Lee
- Department of Electrical Engineering, Kwangwoon University, Seoul, 139-701, South Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, 02447, Korea.
| |
Collapse
|
16
|
Kaushik A, Jayant RD, Tiwari S, Vashist A, Nair M. Nano-biosensors to detect beta-amyloid for Alzheimer's disease management. Biosens Bioelectron 2016; 80:273-287. [PMID: 26851586 PMCID: PMC4786026 DOI: 10.1016/j.bios.2016.01.065] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 01/21/2016] [Accepted: 01/27/2016] [Indexed: 10/22/2022]
Abstract
Beta-amyloid (β-A) peptides are potential biomarkers to monitor Alzheimer's diseases (AD) for diagnostic purposes. Increased β-A level is neurotoxic and induces oxidative stress in brain resulting in neurodegeneration and causes dementia. As of now, no sensitive and inexpensive method is available for β-A detection under physiological and pathological conditions. Although, available methods such as neuroimaging, enzyme-linked immunosorbent assay (ELISA), and polymerase chain reaction (PCR) detect β-A, but they are not yet extended at point-of-care (POC) due to sophisticated equipments, need of high expertize, complicated operations, and challenge of low detection limit. Recently, β-A antibody based electrochemical immuno-sensing approach has been explored to detect β-A at pM levels within 30-40 min compared to 6-8h of ELISA test. The introduction of nano-enabling electrochemical sensing technology could enable rapid detection of β-A at POC and may facilitate fast personalized health care delivery. This review explores recent advancements in nano-enabling electrochemical β-A sensing technologies towards POC application to AD management. These analytical tools can serve as an analytical tool for AD management program to obtain bio-informatics needed to optimize therapeutics for neurodegenerative diseases diagnosis management.
Collapse
Affiliation(s)
- Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of Neuro immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of Neuro immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Sneham Tiwari
- Center for Personalized Nanomedicine, Institute of Neuro immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Arti Vashist
- Center for Personalized Nanomedicine, Institute of Neuro immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Madhavan Nair
- Center for Personalized Nanomedicine, Institute of Neuro immune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
| |
Collapse
|
17
|
Yi X, Feng C, Hu S, Li H, Wang J. Surface plasmon resonance biosensors for simultaneous monitoring of amyloid-beta oligomers and fibrils and screening of select modulators. Analyst 2015; 141:331-6. [PMID: 26613550 DOI: 10.1039/c5an01864a] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oligomeric amyloid-beta (Aβ) peptides are considered as the most toxic species in Alzheimer's disease (AD). Monitoring of the Aβ aggregation profiles is critical for elucidating the oligomer toxicity and may serve as a therapeutic target for AD. By immobilizing the capture antibodies of A11 and OC that are specific to the oligomers and fibrils, respectively, in separate fluidic channels, a novel surface plasmon resonance (SPR) biosensor was designed for monitoring the oligomeric and fibrillar species of Aβ(1-42) simultaneously. The influence of curcumin, Cu(2+) and methylene blue on the amount of toxic oligomers and fibrils was evaluated. The half maximal inhibitory concentration (IC50) of curcumin and methylene blue was determined. The formation of Aβ fibrils was also validated by the thioflavin T (ThT) fluorescence assay. The results demonstrate the utility of SPR as an analytical tool for rapid and comprehensive monitoring of Aβ aggregation and screening of Aβ modulators.
Collapse
Affiliation(s)
- Xinyao Yi
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, P. R. China 410083.
| | | | | | | | | |
Collapse
|
18
|
Yin T, Yang L, Liu Y, Zhou X, Sun J, Liu J. Sialic acid (SA)-modified selenium nanoparticles coated with a high blood-brain barrier permeability peptide-B6 peptide for potential use in Alzheimer's disease. Acta Biomater 2015; 25:172-83. [PMID: 26143603 DOI: 10.1016/j.actbio.2015.06.035] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/17/2015] [Accepted: 06/30/2015] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a formidable gatekeeper toward exogenous substances, playing an important role in brain homeostasis and maintaining a healthy microenvironment for complex neuronal activities. However, it also greatly hinders drug permeability into the brain and limits the management of brain diseases. The development of new drugs that show improved transport across the BBB represents a promising strategy for Alzheimer's disease (AD) intervention. Whereas, previous study of receptor-mediated endogenous BBB transport systems has focused on a strategy of using transferrin to facilitate brain drug delivery system, a system that still suffers from limitations including synthesis procedure, stability and immunological response. In the present study, we synthetised sialic acid (SA)-modified selenium (Se) nanoparticles conjugated with an alternative peptide-B6 peptide (B6-SA-SeNPs, a synthetic selenoprotein analogue), which shows high permeability across the BBB and has the potential to serve as a novel nanomedicine for disease modification in AD. Laser-scanning confocal microscopy, flow cytometry analysis and inductively coupled plasma-atomic emission spectroscopy ICP-AES revealed high cellular uptake of B6-SA-SeNPs by cerebral endothelial cells (bEnd.3). The transport efficiency of B6-SA-SeNPs was evaluated in a Transwell experiment based on in vitro BBB model. It provided direct evidence for B6-SA-SeNPs crossing the BBB and being absorbed by PC12 cells. Moreover, inhibitory effects of B6-SA-SeNPs on amyloid-β peptide (Aβ) fibrillation could be demonstrated in PC12 cells and bEnd3 cells. B6-SA-SeNPs could not only effectively inhibit Aβ aggregation but could disaggregate preformed Aβ fibrils into non-toxic amorphous oligomers. These results suggested that B6-SA-SeNPs may provide a promising platform, particularly for the application of nanoparticles in the treatment of brain diseases. STATEMENT OF SIGNIFICANCE Alzheimer's disease (AD) is the world's most common form of dementia characterized by intracellular neurofibrillary tangles in the brain. Over the past decades, the blood-brain barrier (BBB) limits access of therapeutic or diagnostic agents into the brain, which greatly hinders the development of new drugs for treating AD. In this work, we evaluated the efficiency of B6-SA-SeNPs across BBB and investigated the interactions between B6-SA-SeNPs and amyloid-β peptide (Aβ). We confirm that B6-SA-SeNPs could provide a promising platform because of its high brain delivery efficiency, anti-amyloid properties and anti-oxidant properties, which may serve as a novel nanomedicine for the application in the treatment of brain diseases.
Collapse
|
19
|
Hung VWS, Bressan LP, Seo K, Kerman K. Electroanalysis of Natural Compounds as Copper Chelating Agents for Alzheimer’s Disease Therapy. ELECTROANAL 2015. [DOI: 10.1002/elan.201500138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
20
|
Cheng XR, Wallace GQ, Lagugné-Labarthet F, Kerman K. Au nanostructured surfaces for electrochemical and localized surface plasmon resonance-based monitoring of α-synuclein-small molecule interactions. ACS APPLIED MATERIALS & INTERFACES 2015; 7:4081-4088. [PMID: 25622115 DOI: 10.1021/am507972b] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In this proof-of-concept study, the fabrication of novel Au nanostructured indium tin oxide (Au-ITO) surfaces is described for the development of a dual-detection platform with electrochemical and localized surface plasmon resonance (LSPR)-based biosensing capabilities. Nanosphere lithography (NSL) was applied to fabricate Au-ITO surfaces. Oligomers of α-synuclein (αS) were covalently immobilized to determine the electrochemical and LSPR characteristics of the protein. Cyclic voltammetry (CV) and differential pulse voltammetry (DPV) were performed using the redox probe [Fe(CN)6](3-/4-) to detect the binding of Cu(II) ions and (-)-epigallocatechin-3-gallate (EGCG) to αS on the Au-ITO surface. Electrochemical and LSPR data were complemented by Thioflavin-T (ThT) fluorescence, surface plasmon resonance imaging (SPRi), and transmission electron microscopy (TEM) studies. EGCG was shown to induce the formation of amorphous aggregates that decreased the electrochemical signals. However, the binding of EGCG with αS increased the LSPR absorption band with a bathochromic shift of 10-15 nm. The binding of Cu(II) to αS enhanced the DPV peak current intensity. NSL fabricated Au-ITO surfaces provide a promising dual-detection platform to monitor the interaction of small molecules with proteins using electrochemistry and LSPR.
Collapse
Affiliation(s)
- Xin R Cheng
- Department of Physical and Environmental Sciences, University of Toronto Scarborough , Toronto, Ontario M1C 1A4, Canada
| | | | | | | |
Collapse
|
21
|
Wang C, Xu L, Cheng F, Wang H, Jia L. Curcumin induces structural change and reduces the growth of amyloid-β fibrils: a QCM-D study. RSC Adv 2015. [DOI: 10.1039/c5ra02314a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Curcumin inhibited Aβ fibril growth through leading to the structural conversion of the growing fibril to a more loosely constructed aggregate.
Collapse
Affiliation(s)
- Conggang Wang
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian 116023
- P. R. China
| | - Li Xu
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian 116023
- P. R. China
| | - Fang Cheng
- School of Pharmaceutical Science and Technology
- Dalian University of Technology
- Dalian 116023
- P. R. China
| | - Hanqi Wang
- School of Pharmaceutical Science and Technology
- Dalian University of Technology
- Dalian 116023
- P. R. China
| | - Lingyun Jia
- School of Life Science and Biotechnology
- Dalian University of Technology
- Dalian 116023
- P. R. China
| |
Collapse
|
22
|
Savelieff MG, DeToma AS, Derrick JS, Lim MH. The ongoing search for small molecules to study metal-associated amyloid-β species in Alzheimer's disease. Acc Chem Res 2014; 47:2475-82. [PMID: 25080056 DOI: 10.1021/ar500152x] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The development of a cure for Alzheimer's disease (AD) has been impeded by an inability to pinpoint the root cause of this disorder. Although numerous potential pathological factors have been indicated, acting either individually or mutually, the molecular mechanisms leading to disease onset and progression have not been clear. Amyloid-β (Aβ), generated from proteolytic processing of the amyloid precursor protein (APP), and its aggregated forms, particularly oligomers, are suggested as key pathological features in AD-affected brains. Historically, highly concentrated metals are found colocalized within Aβ plaques. Metal binding to Aβ (metal-Aβ) generates/stabilizes potentially toxic Aβ oligomers, and produces reactive oxygen species (ROS) in vitro (redox active metal ions; plausible contribution to oxidative stress). Consequently, clarification of the relationship between Aβ, metal ions, and toxicity, including oxidative stress via metal-Aβ, can lead to a deeper understanding of AD development. To probe the involvement of metal-Aβ in AD pathogenesis, rationally designed and naturally occurring molecules have been examined as chemical tools to target metal-Aβ species, modulate the interaction between the metal and Aβ, and subsequently redirect their aggregation into nontoxic, off-pathway unstructured aggregates. These ligands are also capable of attenuating the generation of redox active metal-Aβ-induced ROS to mitigate oxidative stress. One rational design concept, the incorporation approach, installs a metal binding site into a framework known to interact with Aβ. This approach affords compounds with the simultaneous ability to chelate metal ions and interact with Aβ. Natural products capable of Aβ interaction have been investigated for their influence on metal-induced Aβ aggregation and have inspired the construction of synthetic analogues. Systematic studies of these synthetic or natural molecules could uncover relationships between chemical structures, metal/Aβ/metal-Aβ interactions, and inhibition of Aβ/metal-Aβ reactivity (i.e., aggregation modes of Aβ/metal-Aβ; associated ROS production), suggesting mechanisms to refine the design strategy. Interdisciplinary investigations have demonstrated that the designed molecules and natural products control the aggregation pathways of metal-Aβ species transforming their size/conformation distribution. The aptitude of these molecules to impact metal-Aβ aggregation pathways, either via inhibition of Aβ aggregate formation, most importantly of oligomers, or disaggregation of preformed fibrils, could originate from their formation of complexes with metal-Aβ. Potentially, these molecules could direct metal-Aβ size/conformational states into alternative nontoxic unstructured oligomers, and control the geometry at the Aβ-ligated metal center for limited ROS formation to lessen the overall toxicity induced by metal-Aβ. Complexation between small molecules and Aβ/metal-Aβ has been observed by nuclear magnetic resonance spectroscopy (NMR) and ion mobility-mass spectrometry (IM-MS) pointing to molecular level interactions, validating the design strategy. In addition, these molecules exhibit other attractive properties, such as antioxidant capacity, prevention of ROS production, potential blood-brain barrier (BBB) permeability, and reduction of Aβ-/metal-Aβ-induced cytotoxicity, making them desirable tools for unraveling AD complexity. In this Account, we summarize the recent development of small molecules, via both rational design and the selection and modification of natural products, as tools for investigating metal-Aβ complexes, to advance our understanding of their relation to AD pathology.
Collapse
Affiliation(s)
| | | | | | - Mi Hee Lim
- Department
of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Korea
| |
Collapse
|
23
|
Cheng XR, Daaboul GG, Ünlü MS, Kerman K. LED-based interferometric reflectance imaging sensor for the detection of amyloid-β aggregation. Analyst 2014; 139:59-65. [DOI: 10.1039/c3an01307c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
24
|
Insights into antiamyloidogenic properties of the green tea extract (-)-epigallocatechin-3-gallate toward metal-associated amyloid-β species. Proc Natl Acad Sci U S A 2013; 110:3743-8. [PMID: 23426629 DOI: 10.1073/pnas.1220326110] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Despite the significance of Alzheimer's disease, the link between metal-associated amyloid-β (metal-Aβ) and disease etiology remains unclear. To elucidate this relationship, chemical tools capable of specifically targeting and modulating metal-Aβ species are necessary, along with a fundamental understanding of their mechanism at the molecular level. Herein, we investigated and compared the interactions and reactivities of the green tea extract, (-)-epigallocatechin-3-gallate [(2R,3R)-5,7-dihydroxy-2-(3,4,5-trihydroxyphenyl)-3,4-dihydro-2H-1-benzopyran-3-yl 3,4,5-trihydroxybenzoate; EGCG], with metal [Cu(II) and Zn(II)]-Aβ and metal-free Aβ species. We found that EGCG interacted with metal-Aβ species and formed small, unstructured Aβ aggregates more noticeably than in metal-free conditions in vitro. In addition, upon incubation with EGCG, the toxicity presented by metal-free Aβ and metal-Aβ was mitigated in living cells. To understand this reactivity at the molecular level, structural insights were obtained by ion mobility-mass spectrometry (IM-MS), 2D NMR spectroscopy, and computational methods. These studies indicated that (i) EGCG was bound to Aβ monomers and dimers, generating more compact peptide conformations than those from EGCG-untreated Aβ species; and (ii) ternary EGCG-metal-Aβ complexes were produced. Thus, we demonstrate the distinct antiamyloidogenic reactivity of EGCG toward metal-Aβ species with a structure-based mechanism.
Collapse
|
25
|
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
|