1
|
Li YY, Yang Y, Zhang RS, Ge RX, Xie SB. Targeted degradation of membrane and extracellular proteins with LYTACs. Acta Pharmacol Sin 2024:10.1038/s41401-024-01364-y. [PMID: 39103530 DOI: 10.1038/s41401-024-01364-y] [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: 04/28/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/07/2024] Open
Abstract
Targeted protein degradation technology has gained substantial momentum over the past two decades as a revolutionary strategy for eliminating pathogenic proteins that are otherwise refractory to treatment. Among the various approaches developed to harness the body's innate protein homeostasis mechanisms for this purpose, lysosome targeting chimeras (LYTACs) that exploit the lysosomal degradation pathway by coupling the target proteins with lysosome-trafficking receptors represent the latest innovation. These chimeras are uniquely tailored to degrade proteins that are membrane-bound and extracellular, encompassing approximately 40% of all proteome. Several novel LYTAC formulas have been developed recently, providing valuable insights for the design and development of therapeutic degraders. This review delineates the recent progresses of LYTAC technology, its practical applications, and the factors that dictate target degradation efficiency. The potential and emerging trends of this technology are discussed as well. LYTAC technology offers a promising avenue for targeted protein degradation, potentially revolutionizing the therapeutic landscape for numerous diseases.
Collapse
Affiliation(s)
- Yu-Yang Li
- Department of Clinical Pathobiology and Immunological Testing, School of Medical Laboratory, Qilu Medical University, Zibo, 255300, China
| | - Yang Yang
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Ren-Shuai Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Rui-Xin Ge
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Song-Bo Xie
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, 300052, China.
| |
Collapse
|
2
|
Porcu S, Corpino R, Carbonaro CM, Ricci PC, Vargiu AV, Sanna AL, Sforazzini G, Chiriu D. Promising Molecular Architectures for Two-Photon Probes in the Diagnosis of α-Synuclein Aggregates. Molecules 2024; 29:2817. [PMID: 38930882 PMCID: PMC11207056 DOI: 10.3390/molecules29122817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The abnormal deposition of protein in the brain is the central factor in neurodegenerative disorders (NDs). These detrimental aggregates, stemming from the misfolding and subsequent irregular aggregation of α-synuclein protein, are primarily accountable for conditions such as Parkinson's disease, Alzheimer's disease, and dementia. Two-photon-excited (TPE) probes are a promising tool for the early-stage diagnosis of these pathologies as they provide accurate spatial resolution, minimal intrusion, and the ability for prolonged observation. To identify compounds with the potential to function as diagnostic probes using two-photon techniques, we explore three distinct categories of compounds: Hydroxyl azobenzene (AZO-OH); Dicyano-vinyl bithiophene (DCVBT); and Tetra-amino phthalocyanine (PcZnNH2). The molecules were structurally and optically characterized using a multi-technique approach via UV-vis absorption, Raman spectroscopy, three-dimensional fluorescence mapping (PLE), time-resolved photoluminescence (TRPL), and pump and probe measurements. Furthermore, quantum chemical and molecular docking calculations were performed to provide insights into the photophysical properties of the compounds as well as to assess their affinity with the α-synuclein protein. This innovative approach seeks to enhance the accuracy of in vivo probing, contributing to early Parkinson's disease (PD) detection and ultimately allowing for targeted intervention strategies.
Collapse
Affiliation(s)
- Stefania Porcu
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| | - Riccardo Corpino
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| | - Carlo Maria Carbonaro
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| | - Pier Carlo Ricci
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| | - Attilio Vittorio Vargiu
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| | - Anna Laura Sanna
- Department of Chemistry and Hearth Science, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy;
| | - Giuseppe Sforazzini
- Department of Chemistry and Hearth Science, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy;
| | - Daniele Chiriu
- Department of Physics, University of Cagliari, Cittadella Universitaria, SP n°8, 09042 Monserrato, CA, Italy; (S.P.); (R.C.); (C.M.C.); (P.C.R.); (A.V.V.)
| |
Collapse
|
3
|
Bisi N, Pinzi L, Rastelli G, Tonali N. Early Diagnosis of Neurodegenerative Diseases: What Has Been Undertaken to Promote the Transition from PET to Fluorescence Tracers. Molecules 2024; 29:722. [PMID: 38338465 PMCID: PMC10856728 DOI: 10.3390/molecules29030722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Alzheimer's Disease (AD) and Parkinson's Disease (PD) represent two among the most frequent neurodegenerative diseases worldwide. A common hallmark of these pathologies is the misfolding and consequent aggregation of amyloid proteins into soluble oligomers and insoluble β-sheet-rich fibrils, which ultimately lead to neurotoxicity and cell death. After a hundred years of research on the subject, this is the only reliable histopathological feature in our hands. Since AD and PD are diagnosed only once neuronal death and the first symptoms have appeared, the early detection of these diseases is currently impossible. At present, there is no effective drug available, and patients are left with symptomatic and inconclusive therapies. Several reasons could be associated with the lack of effective therapeutic treatments. One of the most important factors is the lack of selective probes capable of detecting, as early as possible, the most toxic amyloid species involved in the onset of these pathologies. In this regard, chemical probes able to detect and distinguish among different amyloid aggregates are urgently needed. In this article, we will review and put into perspective results from ex vivo and in vivo studies performed on compounds specifically interacting with such early species. Following a general overview on the three different amyloid proteins leading to insoluble β-sheet-rich amyloid deposits (amyloid β1-42 peptide, Tau, and α-synuclein), a list of the advantages and disadvantages of the approaches employed to date is discussed, with particular attention paid to the translation of fluorescence imaging into clinical applications. Furthermore, we also discuss how the progress achieved in detecting the amyloids of one neurodegenerative disease could be leveraged for research into another amyloidosis. As evidenced by a critical analysis of the state of the art, substantial work still needs to be conducted. Indeed, the early diagnosis of neurodegenerative diseases is a priority, and we believe that this review could be a useful tool for better investigating this field.
Collapse
Affiliation(s)
- Nicolò Bisi
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy; (L.P.); (G.R.)
| | - Nicolò Tonali
- Université Paris-Saclay, CNRS, BioCIS, Bat. Henri Moissan, 17, Av. des Sciences, 91400 Orsay, France
| |
Collapse
|
4
|
Teppang KL, Zhao Q, Yang J. Development of fluorophores for the detection of oligomeric aggregates of amyloidogenic proteins found in neurodegenerative diseases. Front Chem 2023; 11:1343118. [PMID: 38188930 PMCID: PMC10766704 DOI: 10.3389/fchem.2023.1343118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease and Parkinson's disease are the two most common neurodegenerative diseases globally. These neurodegenerative diseases have characteristic late-stage symptoms allowing for differential diagnosis; however, they both share the presence of misfolded protein aggregates which appear years before clinical manifestation. Historically, research has focused on the detection of higher-ordered aggregates (or amyloids); however, recent evidence has shown that the oligomeric state of these protein aggregates plays a greater role in disease pathology, resulting in increased efforts to detect oligomers to aid in disease diagnosis. In this review, we summarize some of the exciting new developments towards the development of fluorescent probes that can detect oligomeric aggregates of amyloidogenic proteins present in Alzheimer's and Parkinson's disease patients.
Collapse
Affiliation(s)
| | | | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
5
|
Jain M, Dhariwal R, Patil N, Ojha S, Tendulkar R, Tendulkar M, Dhanda PS, Yadav A, Kaushik P. Unveiling the Molecular Footprint: Proteome-Based Biomarkers for Alzheimer's Disease. Proteomes 2023; 11:33. [PMID: 37873875 PMCID: PMC10594437 DOI: 10.3390/proteomes11040033] [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: 08/30/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss. Early and accurate diagnosis of AD is crucial for implementing timely interventions and developing effective therapeutic strategies. Proteome-based biomarkers have emerged as promising tools for AD diagnosis and prognosis due to their ability to reflect disease-specific molecular alterations. There is of great significance for biomarkers in AD diagnosis and management. It emphasizes the limitations of existing diagnostic approaches and the need for reliable and accessible biomarkers. Proteomics, a field that comprehensively analyzes the entire protein complement of cells, tissues, or bio fluids, is presented as a powerful tool for identifying AD biomarkers. There is a diverse range of proteomic approaches employed in AD research, including mass spectrometry, two-dimensional gel electrophoresis, and protein microarrays. The challenges associated with identifying reliable biomarkers, such as sample heterogeneity and the dynamic nature of the disease. There are well-known proteins implicated in AD pathogenesis, such as amyloid-beta peptides, tau protein, Apo lipoprotein E, and clusterin, as well as inflammatory markers and complement proteins. Validation and clinical utility of proteome-based biomarkers are addressing the challenges involved in validation studies and the diagnostic accuracy of these biomarkers. There is great potential in monitoring disease progression and response to treatment, thereby aiding in personalized medicine approaches for AD patients. There is a great role for bioinformatics and data analysis in proteomics for AD biomarker research and the importance of data preprocessing, statistical analysis, pathway analysis, and integration of multi-omics data for a comprehensive understanding of AD pathophysiology. In conclusion, proteome-based biomarkers hold great promise in the field of AD research. They provide valuable insights into disease mechanisms, aid in early diagnosis, and facilitate personalized treatment strategies. However, further research and validation studies are necessary to harness the full potential of proteome-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Mukul Jain
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Rupal Dhariwal
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Nil Patil
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Sandhya Ojha
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Reshma Tendulkar
- Vivekanand Education Society, College of Pharmacy, Chembur, Mumbai 400071, India;
| | - Mugdha Tendulkar
- Sardar Vallabhbhai Patel College of Science, Mira Rd (East), Thane 400071, India;
| | | | - Alpa Yadav
- Department of Botany, Indira Gandhi University, Meerpur, Rewari 122502, India;
| | - Prashant Kaushik
- Instituto de Conservacióny Mejora de la Agrodiversidad Valenciana, Universitat Politècnica de València, 46022 Valencia, Spain
| |
Collapse
|
6
|
Kim HY, Chia WK, Hsieh CJ, Guarino DS, Graham TJA, Lengyel-Zhand Z, Schneider M, Tomita C, Lougee MG, Kim HJ, Pagar VV, Lee H, Hou C, Garcia BA, Petersson EJ, O’Shea J, Kotzbauer PT, Mathis CA, Lee VMY, Luk KC, Mach RH. A Novel Brain PET Radiotracer for Imaging Alpha Synuclein Fibrils in Multiple System Atrophy. J Med Chem 2023; 66:12185-12202. [PMID: 37651366 PMCID: PMC10617560 DOI: 10.1021/acs.jmedchem.3c00779] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Abnormal α-synuclein (α-syn) aggregation characterizes α-synucleinopathies, including Parkinson's disease (PD) and multiple system atrophy (MSA). However, no suitable positron emission tomography (PET) radiotracer for imaging α-syn in PD and MSA exists currently. Our structure-activity relationship studies identified 4-methoxy-N-(4-(3-(pyridin-2-yl)-3,8-diazabicyclo[3.2.1]octan-8-yl)phenyl)benzamide (4i) as a PET radiotracer candidate for imaging α-syn. In vitro assays revealed high binding of 4i to recombinant α-syn fibrils (inhibition constant (Ki) = 6.1 nM) and low affinity for amyloid beta (Aβ) fibrils in Alzheimer's disease (AD) homogenates. However, [3H]4i also exhibited high specific binding to AD, progressive supranuclear palsy, and corticobasal degeneration tissues as well as PD and MSA tissues, suggesting notable affinity to tau. Nevertheless, the specific binding to pathologic α-syn aggregates in MSA post-mortem brain tissues was significantly higher than in PD tissues. This finding demonstrated the potential use of [11C]4i as a PET tracer for imaging α-syn in MSA patients. Nonhuman primate PET studies confirmed good brain uptake and rapid washout for [11C]4i.
Collapse
Affiliation(s)
- Ho Young Kim
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Wai Kit Chia
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Chia-Ju Hsieh
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Dinahlee Saturnino Guarino
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Thomas J. A. Graham
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Zsofia Lengyel-Zhand
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Mark Schneider
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Cosette Tomita
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Marshall G. Lougee
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Hee Jong Kim
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6303, USA
| | - Vinayak V. Pagar
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Catherine Hou
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6303, USA
| | - E. James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104-6323, USA
| | - Jennifer O’Shea
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110-1010, USA
| | - Paul T. Kotzbauer
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110-1010, USA
| | - Chester A. Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research, Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-2676, USA
| | - Kelvin C. Luk
- Center for Neurodegenerative Disease Research, Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-2676, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Vagelos Laboratories, 1012, 231 S. 34th Street, Philadelphia, PA 19104-6323, USA
| |
Collapse
|
7
|
Watt KJC, Meade RM, James TD, Mason JM. Development of a hydroxyflavone-labelled 4554W peptide probe for monitoring αS aggregation. Sci Rep 2023; 13:10968. [PMID: 37414785 PMCID: PMC10326036 DOI: 10.1038/s41598-023-37655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023] Open
Abstract
Parkinson's is the second most common neurodegenerative disease, with the number of individuals susceptible due to increase as a result of increasing life expectancy and a growing worldwide population. However, despite the number of individuals affected, all current treatments for PD are symptomatic-they alleviate symptoms, but do not slow disease progression. A major reason for the lack of disease-modifying treatments is that there are currently no methods to diagnose individuals during the earliest stages of the disease, nor are there any methods to monitor disease progression at a biochemical level. Herein, we have designed and evaluated a peptide-based probe to monitor αS aggregation, with a particular focus on the earliest stages of the aggregation process and the formation of oligomers. We have identified the peptide-probe K1 as being suitable for further development to be applied to number of applications including: inhibition of αS aggregation; as a probe to monitor αS aggregation, particularly at the earliest stages before Thioflavin-T is active; and a method to detect early-oligomers. With further development and in vivo validation, we anticipate this probe could be used for the early diagnosis of PD, a method to evaluate the effectiveness of potential therapeutics, and as a tool to help in the understanding of the onset and development of PD.
Collapse
Affiliation(s)
- Kathryn J C Watt
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Richard M Meade
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Tony D James
- Department of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Jody M Mason
- Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
8
|
Tong Y, Zhu W, Chen J, Wen T, Xu F, Pang J. Discovery of Small-Molecule Degraders for Alpha-Synuclein Aggregates. J Med Chem 2023. [PMID: 37267712 DOI: 10.1021/acs.jmedchem.3c00274] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Alpha-synuclein (αSyn) species, especially the oligomers and fibers, are associated with multiple neurodegenerative diseases and cannot be directly targeted under the conventional pharmacological paradigm. Proteolysis-targeting chimera technology confers degradation of various "undruggable" targets; however, hardly any small-molecule degrader for αSyn aggregates has been reported yet. Herein, by using the probe molecule sery308 as a warhead, a series of small-molecule degraders for αSyn aggregates were designed and synthesized. Their degradation effects on αSyn aggregates were evaluated on a modified pre-formed fibril-seeding cell model. Compound 2b exhibited the highest degradation efficiency (DC50 = 7.51 ± 0.53 μM) with high selectivity. Mechanistic exploration revealed that both proteasomal and lysosomal pathways were involved in this kind of degradation. Moreover, the therapeutic effects of 2b were tested on SH-SY5Y (human neuroblastoma cell line) cells and Caenorhabditis elegans. Our results provided a new class of small-molecule candidates against synucleinopathies and broadened the substrate spectrum of PROTAC-based degraders.
Collapse
Affiliation(s)
- Yichen Tong
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Wentao Zhu
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Chen
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianzhi Wen
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Xu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization & Innovative Drug Development of Chinese Ministry of Education (MOE) & Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jiyan Pang
- School of Chemistry, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
9
|
Somavarapu AK, Kleijwegt G, Nagaraj M, Alam P, Nielsen J, Otzen DE. Drug repurposing screens identify compounds that inhibit α-synuclein oligomers' membrane disruption and block antibody interactions. Chem Sci 2023; 14:3030-3047. [PMID: 36937574 PMCID: PMC10016340 DOI: 10.1039/d2sc05534a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Small soluble oligomers of the protein α-synuclein (αSO) have been linked to disruptions in neuronal homeostasis, contributing to the development of Parkinson's Disease (PD). While this makes αSO an obvious drug target, the development of effective therapeutics against αSO is challenged by its low abundance and structural and morphological complexity. Here, we employ two different approaches to neutralize toxic interactions made by αSOs with different cellular components. First, we use available data to identify four neuronal proteins as likely candidates for αSO interactions, namely Cfl1, Uchl1, Sirt2 and SerRS. However, despite promising results when immobilized, all 4 proteins only bind weakly to αSO in solution in microfluidic assays, making them inappropriate for screening. In contrast, the formation of stable contacts formed between αSO and vesicles consisting of anionic lipids not only mimics a likely biological role of αSO but also provided a platform to screen two small molecule libraries for disruptors of these contacts. Of the 7 best leads obtained in this way, 2 significantly impaired αSO contacts with other proteins in a sandwich ELISA assay using αSO-binding monoclonal antibodies and nanobodies. In addition, 5 of these leads suppressed α-synuclein amyloid formation. Thus, a repurposing screening that directly targets a key culprit in PD pathogenesis shows therapeutic potential.
Collapse
Affiliation(s)
- Arun Kumar Somavarapu
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| | - Giulia Kleijwegt
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| | - Madhu Nagaraj
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| | - Parvez Alam
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| | - Janni Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University Gustav Wieds Vej 14, 8000 Aarhus C Denmark
| |
Collapse
|
10
|
Smith JK, Mellick GD, Sykes AM. The role of the endolysosomal pathway in α-synuclein pathogenesis in Parkinson's disease. Front Cell Neurosci 2023; 16:1081426. [PMID: 36704248 PMCID: PMC9871505 DOI: 10.3389/fncel.2022.1081426] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/11/2023] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease that is characterized by a loss of dopaminergic neurons in the substantia nigra pars compacta of the midbrain (SNpc). Extensive studies into genetic and cellular models of PD implicate protein trafficking as a prominent contributor to the death of these dopaminergic neurons. Considerable evidence also suggests the involvement of α-synuclein as a central component of the characteristic cell death in PD and it is a major structural constituent of proteinaceous inclusion bodies (Lewy bodies; LB). α-synuclein research has been a vital part of PD research in recent years, with newly discovered evidence suggesting that α-synuclein can propagate through the brain via prion-like mechanisms. Healthy cells can internalize toxic α-synuclein species and seed endogenous α-synuclein to form large, pathogenic aggregates and form LBs. A better understanding of how α-synuclein can propagate, enter and be cleared from the cell is vital for therapeutic strategies.
Collapse
|
11
|
Kaide S, Watanabe H, Iikuni S, Hasegawa M, Ono M. Synthesis and Evaluation of 18F-Labeled Chalcone Analogue for Detection of α-Synuclein Aggregates in the Brain Using the Mouse Model. ACS Chem Neurosci 2022; 13:2982-2990. [PMID: 36197745 DOI: 10.1021/acschemneuro.2c00473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
In the brains of patients with synucleinopathies such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, α-synuclein (α-syn) aggregates deposit abnormally to induce neurodegeneration, although the mechanism is unclear. Thus, in vivo imaging studies targeting α-syn aggregates have attracted much attention to guide medical intervention against synucleinopathy. In our previous study, a chalcone analogue, [125I]PHNP-3, functioned as a feasible probe in terms of α-syn binding in vitro; however, it did not migrate to the mouse brain, and further improvement of brain uptake was required. In the present study, we designed and synthesized two novel 18F-labeled chalcone analogues, [18F]FHCL-1 and [18F]FHCL-2, using a central nervous system multiparameter optimization (CNS MPO) algorithm with the aim of improving blood-brain barrier permeation in the mouse brain. Then, we evaluated their utility for in vivo imaging of α-syn aggregates using a mouse model. In the competitive inhibition assay, both chalcone analogues exhibited high binding affinity for α-syn aggregates (Ki = 2.6 and 3.4 nM, respectively), while no marked amyloid β (Aβ)-binding was observed. The 18F-labeling reaction was successfully performed. In a biodistribution experiment, brain uptake of both chalcone analogues in normal mice (2.09 and 2.40% injected dose/gram (% ID/g) at 2 min postinjection, respectively) was higher than that of [125I]PHNP-3, suggesting that the introduction of 18F into the chalcone analogue led to an improvement in brain uptake in mice while maintaining favorable binding ability for α-syn aggregates. Furthermore, in an ex vivo autoradiography experiment, [18F]FHCL-2 showed the feasibility of the detection of α-syn aggregates in the mouse brain in vivo. These preclinical studies demonstrated the validity of the design of α-syn-targeting probes based on the CNS MPO score and the possibility of in vivo imaging of α-syn aggregates in a mouse model using 18F-labeled chalcone analogues.
Collapse
Affiliation(s)
- Sho Kaide
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
12
|
REMD Simulations of Full-Length Alpha-Synuclein Together with Ligands Reveal Binding Region and Effect on Amyloid Conversion. Int J Mol Sci 2022; 23:ijms231911545. [PMID: 36232847 PMCID: PMC9569888 DOI: 10.3390/ijms231911545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Alpha-synuclein is a key protein involved in the development and progression of Parkinson’s disease and other synucleinopathies. The intrinsically disordered nature of alpha-synuclein hinders the computational screening of new drug candidates for the treatment of these neurodegenerative diseases. In the present work, replica exchange molecular dynamics simulations of the full-length alpha-synuclein together with low-molecular ligands were utilized to predict the binding site and effect on the amyloid-like conversion of the protein. This approach enabled an accurate prediction of the binding sites for three tested compounds (fasudil, phthalocyanine tetrasulfonate, and spermine), giving good agreement with data from experiments by other groups. Lots of information about the binding and protein conformational ensemble enabled the suggestion of a putative effect of the ligands on the amyloid-like conversion of alpha-synuclein and the mechanism of anti- and pro-amyloid activity of the tested compounds. Therefore, this approach looks promising for testing new drug candidates for binding with alpha-synuclein or other intrinsically disordered proteins and at the same time the estimation of the effect on protein behavior, including amyloid-like aggregation.
Collapse
|
13
|
Nwabufo CK, Aigbogun OP. Diagnostic and therapeutic agents that target alpha-synuclein in Parkinson's disease. J Neurol 2022; 269:5762-5786. [PMID: 35831620 PMCID: PMC9281355 DOI: 10.1007/s00415-022-11267-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
The development of disease-modifying drugs and differential diagnostic agents is an urgent medical need in Parkinson’s disease. Despite the complex pathophysiological pathway, the misfolding of alpha-synuclein has been identified as a putative biomarker for detecting the onset and progression of the neurodegeneration associated with Parkinson’s disease. Identifying the most appropriate alpha-synuclein-based diagnostic modality with clinical translation will revolutionize the diagnosis of Parkinson’s. Likewise, molecules that target alpha-synuclein could alter the disease pathway that leads to Parkinson’s and may serve as first-in class therapeutics compared to existing treatment options such as levodopa and dopamine agonist that do not necessarily modify the disease pathway. Notwithstanding the promising benefits that alpha-synuclein presents to therapeutics and diagnostics development for Parkinson’s disease, finding ways to address potential challenges such as inadequate preclinical models, safety and efficacy will be paramount to achieving clinical translation. In this comprehensive review paper, we described the role of alpha-synuclein in the pathogenesis of Parkinson’s disease, as well as how its structure and function relationship delineate disease onset and progression. We further discussed different alpha-synuclein-based diagnostic modalities including biomolecular assays and molecular imaging. Finally, we presented current small molecules and biologics that are being developed as disease-modifying drugs or positron emission tomography imaging probes for Parkinson’s disease.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada. .,Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| | - Omozojie P Aigbogun
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.,Department of Chemistry, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
14
|
Królicka E, Kieć-Kononowicz K, Łażewska D. Chalcones as Potential Ligands for the Treatment of Parkinson's Disease. Pharmaceuticals (Basel) 2022; 15:ph15070847. [PMID: 35890146 PMCID: PMC9317344 DOI: 10.3390/ph15070847] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/16/2022] Open
Abstract
Along with the increase in life expectancy, a significant increase of people suffering from neurodegenerative diseases (ND) has been noticed. The second most common ND, after Alzheimer’s disease, is Parkinson’s disease (PD), which manifests itself with a number of motor and non-motor symptoms that hinder the patient’s life. Current therapies can only alleviate those symptoms and slow down the progression of the disease, but not effectively cure it. So now, in addition to understanding the mechanism and causes of PD, it is also important to find a powerful way of treatment. It has been proved that in the etiology and course of PD, the essential roles are played by dopamine (DA) (an important neurotransmitter), enzymes regulating its level (e.g., COMT, MAO), and oxidative stress leading to neuroinflammation. Chalcones, due to their “simple” structure and valuable biological properties are considered as promising candidates for treatment of ND, also including PD. Here, we provide a comprehensive review of chalcones and related structures as potential new therapeutics for cure and prevention of PD. For this purpose, three databases (Pubmed, Scopus and Web of Science) were searched to collect articles published during the last 5 years (January 2018–February 2022). Chalcones have been described as promising enzyme inhibitors (MAO B, COMT, AChE), α-synuclein imaging probes, showing anti-neuroinflammatory activity (inhibition of iNOS or activation of Nrf2 signaling), as well as antagonists of adenosine A1 and/or A2A receptors. This review focused on the structure–activity relationships of these compounds to determine how a particular substituent or its position in the chalcone ring(s) (ring A and/or B) affects biological activity.
Collapse
|
15
|
Min JO, Strohäker T, Jeong BC, Zweckstetter M, Lee SJ. Chicago sky blue 6B inhibits α-synuclein aggregation and propagation. Mol Brain 2022; 15:27. [PMID: 35346306 PMCID: PMC8962151 DOI: 10.1186/s13041-022-00913-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/17/2022] [Indexed: 11/10/2022] Open
Abstract
Abnormal deposition of α-synuclein aggregates in Lewy bodies and Lewy neurites is the hallmark lesion in Parkinson’s disease (PD). These aggregates, thought to be the culprit of disease pathogenesis, spread throughout the brain as the disease progresses. Agents that inhibit α-synuclein aggregation and/or spread of aggregates would thus be candidate disease-modifying drugs. Here, we found that Chicago sky blue 6B (CSB) may be such a drug, showing that it inhibits α-synuclein aggregation and cell-to-cell propagation in both in vitro and in vivo models of synucleinopathy. CSB inhibited the fibrillation of α-synuclein in a concentration-dependent manner through direct binding to the N-terminus of α-synuclein. Furthermore, both seeded polymerization and cell-to-cell propagation of α-synuclein were inhibited by CSB treatment. Notably, CSB alleviated behavioral deficits and neuropathological features, such as phospho-α-synuclein and astrogliosis, in A53T α-synuclein transgenic mice. These results indicate that CSB directly binds α-synuclein and inhibits its aggregation, thereby blocking α-synuclein cell-to-cell propagation.
Collapse
Affiliation(s)
- Joo-Ok Min
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, Republic of Korea
| | - Timo Strohäker
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
| | - Byung-Chul Jeong
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, Republic of Korea.,Nutritional Sciences and Toxicology Department, University of California Berkeley, Berkeley, USA
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.,Department for NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
16
|
Alzghool OM, van Dongen G, van de Giessen E, Schoonmade L, Beaino W. α-Synuclein Radiotracer Development and In Vivo Imaging: Recent Advancements and New Perspectives. Mov Disord 2022; 37:936-948. [PMID: 35289424 PMCID: PMC9310945 DOI: 10.1002/mds.28984] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/14/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
α-Synucleinopathies including idiopathic Parkinson's disease, dementia with Lewy bodies and multiple systems atrophy share overlapping symptoms and pathological hallmarks. Selective neurodegeneration and Lewy pathology are the main hallmarks of α-synucleinopathies. Currently, there is no imaging biomarker suitable for a definitive early diagnosis of α-synucleinopathies. Although dopaminergic deficits detected with single-photon emission computed tomography (SPECT) and positron emission tomography (PET) radiotracers can support clinical diagnosis by confirming the presence of dopaminergic neurodegeneration, dopaminergic imaging cannot visualize the preceding disease process, nor distinguish α-synucleinopathies from tauopathies with dopaminergic neurodegeneration, especially at early symptomatic disease stage when clinical presentation is often overlapping. Aggregated α-synuclein (αSyn) could be a suitable imaging biomarker in α-synucleinopathies, because αSyn aggregation and therefore, Lewy pathology is evidently an early driver of α-synucleinopathies pathogenesis. Additionally, several antibodies and small molecule compounds targeting aggregated αSyn are in development for therapy. However, there is no way to directly measure if or how much they lower the levels of aggregated αSyn in the brain. There is clearly a paramount diagnostic and therapeutic unmet medical need. To date, aggregated αSyn and Lewy pathology inclusion bodies cannot be assessed ante-mortem with SPECT or PET imaging because of the suboptimal binding characteristics and/or physicochemical properties of current radiotracers. The aim of this narrative review is to highlight the suitability of aggregated αSyn as an imaging biomarker in α-synucleinopathies, the current limitations with and lessons learned from αSyn radiotracer development, and finally to propose antibody-based ligands for imaging αSyn aggregates as a complementary tool rather than an alternative to small molecule ligands. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
- Obada M Alzghool
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands.,Turku PET Centre, University of Turku, Turku, Finland
| | - Guus van Dongen
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elsmarieke van de Giessen
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Linda Schoonmade
- Medical Library, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wissam Beaino
- Department of Radiology and Nuclear Medicine, Tracer Center Amsterdam, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Kaide S, Watanabe H, Iikuni S, Hasegawa M, Itoh K, Ono M. Chalcone Analogue as New Candidate for Selective Detection of α-Synuclein Pathology. ACS Chem Neurosci 2022; 13:16-26. [PMID: 34910473 DOI: 10.1021/acschemneuro.1c00441] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Deposition of α-synuclein (α-syn) aggregates is one of the neuropathological hallmarks of synucleinopathies including Parkinson's disease, dementia with Lewy bodies, and multiple-system atrophy. In vivo detection of α-syn aggregates with SPECT or PET may be an effective tool for medical intervention against synucleinopathy. In the present study, we designed and synthesized a series of chalcone analogues with different aryl groups to evaluate their potential as α-syn imaging probes. In competitive inhibition assays, aryl groups markedly affected binding affinity and selectivity for recombinant α-syn aggregates. Chalcone analogues with a 4-(dimethylamino)phenyl group bound to both α-syn and amyloid β (Aβ) aggregates while ones with a 4-nitrophenyl group displayed α-syn-selective binding. In fluorescent staining, only chalcone analogues with a 4-nitrophenyl group succeeded in selective detection of human α-syn against Aβ aggregates in patients' brain samples. Among them, PHNP-3 exhibited the most promising binding characteristics for α-syn aggregates (Ki = 0.52 nM), encouraging us to further evaluate its utility. Then, a 125I-labeling reaction was performed to obtain [125I]PHNP-3. In a binding saturation assay, [125I]PHNP-3 bound to α-syn aggregates with high affinity (Kd = 6.9 nM) and selectivity. In a biodistribution study, [125I]PHNP-3 exhibited modest uptake (0.78% ID/g at 2 min after intravenous injection) into a normal mouse brain. Although there is room for improvement of its pharmacokinetics in the brain, encouraging in vitro results in the present study indicate that further structural optimization based on PHNP-3 might lead to the development of a clinically useful probe targeting α-syn aggregates in the future.
Collapse
Affiliation(s)
- Sho Kaide
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kyoko Itoh
- Department of Pathology & Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
18
|
Zhong J, Li M, Xu J, Dong W, Qin Y, Qiu S, Li X, Wang H. Roflupram attenuates α-synuclein-induced cytotoxicity and promotes the mitochondrial translocation of Parkin in SH-SY5Y cells overexpressing A53T mutant α-synuclein. Toxicol Appl Pharmacol 2022; 436:115859. [PMID: 34990728 DOI: 10.1016/j.taap.2021.115859] [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: 09/08/2021] [Revised: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 01/05/2023]
Abstract
We have previously shown that inhibition of cAMP-specific 3',5'-cyclic phosphodiesterase 4 (PDE4) protects against cellular toxicity in neuronal cells. Since α-synuclein (α-syn) toxicity contributes to the neurodegeneration of Parkinson's disease (PD). The aim of this study was to explore the effects and mechanisms of PDE4 on α-syn-induced neuronal toxicity. Using mutant human A53T α-syn overexpressed SH-SY5Y cells, we found that PDE4B knockdown reduced cellular apoptosis. Roflupram (ROF, 20 μM), a selective PDE4 inhibitor, produced similar protective effects and restored the morphological alterations of mitochondria. Mechanistic studies identified that α-syn enhanced the phosphorylation of Parkin at Ser131, followed by the decreased mitochondrial translocation of Parkin. Whereas both PDE4B knockdown and PDE4 inhibition by ROF blocked the effects of α-syn on Parkin phosphorylation and mitochondrial translocation. Moreover, PDE4 inhibition reversed the increase in the phosphorylation of p38 mitogen-activated protein kinase (MAPK) induced by α-syn. ROF treatment also reduced the binding of p38 MAPK to Parkin. Consistently, overexpression of PDE4B blocked the roles of ROF on p38 MAPK phosphorylation, Parkin phosphorylation, and the subsequent mitochondrial translocation of parkin. Furthermore, PDE4B overexpression attenuated the protective role of ROF, as evidenced by reduced mitochondria membrane potential and increased cellular apoptosis. Interestingly, ROF failed to suppress α-syn-induced cytotoxicity in the presence of a protein kinase A (PKA) inhibitor H-89. Our findings indicate that PDE4 facilitates α-syn-induced cytotoxicity via the PKA/p38 MAPK/Parkin pathway in SH-SY5Y cells overexpressing A53T mutant α-synuclein. PDE4 inhibition by ROF is a promising strategy for the prevention and treatment of α-syn-induced neurodegeneration.
Collapse
Affiliation(s)
- Jiahong Zhong
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mengfan Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiangping Xu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| | - Wenli Dong
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yunyun Qin
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuqing Qiu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xing Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haitao Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
19
|
Upcott M, Chaprov KD, Buchman VL. Toward a Disease-Modifying Therapy of Alpha-Synucleinopathies: New Molecules and New Approaches Came into the Limelight. Molecules 2021; 26:molecules26237351. [PMID: 34885933 PMCID: PMC8658846 DOI: 10.3390/molecules26237351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 12/25/2022] Open
Abstract
The accumulation of the various products of alpha-synuclein aggregation has been associated with the etiology and pathogenesis of several neurodegenerative conditions, including both familial and sporadic forms of Parkinson’s disease (PD) and dementia with Lewy bodies (DLB). It is now well established that the aggregation and spread of alpha-synuclein aggregation pathology activate numerous pathogenic mechanisms that contribute to neurodegeneration and, ultimately, to disease progression. Therefore, the development of a safe and effective disease-modifying therapy that limits or prevents the accumulation of the toxic intermediate products of alpha-synuclein aggregation and the spread of alpha-synuclein aggregation pathology could provide significant positive clinical outcomes in PD/DLB cohorts. It has been suggested that this goal can be achieved by reducing the intracellular and/or extracellular levels of monomeric and already aggregated alpha-synuclein. The principal aim of this review is to critically evaluate the potential of therapeutic strategies that target the post-transcriptional steps of alpha-synuclein production and immunotherapy-based approaches to alpha-synuclein degradation in PD/DLB patients. Strategies aimed at the downregulation of alpha-synuclein production are at an early preclinical stage of drug development and, although they have shown promise in animal models of alpha-synuclein aggregation, many limitations need to be resolved before in-human clinical trials can be seriously considered. In contrast, many strategies aimed at the degradation of alpha-synuclein using immunotherapeutic approaches are at a more advanced stage of development, with some in-human Phase II clinical trials currently in progress. Translational barriers for both strategies include the limitations of alpha-synuclein aggregation models, poor understanding of the therapeutic window for the alpha-synuclein knockdown, and variability in alpha-synuclein pathology across patient cohorts. Overcoming such barriers should be the main focus of further studies. However, it is already clear that these strategies do have the potential to achieve a disease-modifying effect in PD and DLB.
Collapse
Affiliation(s)
- Matthew Upcott
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK;
| | - Kirill D. Chaprov
- Institute of Physiologically Active Compounds Russian Academy of Sciences (IPAC RAS), 1 Severniy Proezd, Chernogolovk, 142432 Moscow, Russia;
- Belgorod State National Research University, 85 Pobedy Street, 308015 Belgorod, Russia
| | - Vladimir L. Buchman
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK;
- Institute of Physiologically Active Compounds Russian Academy of Sciences (IPAC RAS), 1 Severniy Proezd, Chernogolovk, 142432 Moscow, Russia;
- Belgorod State National Research University, 85 Pobedy Street, 308015 Belgorod, Russia
- Correspondence:
| |
Collapse
|
20
|
Dong WL, Zhong JH, Chen YQ, Xie JF, Qin YY, Xu JP, Cai NB, Li MF, Liu L, Wang HT. Roflupram protects against rotenone-induced neurotoxicity and facilitates α-synuclein degradation in Parkinson's disease models. Acta Pharmacol Sin 2021; 42:1991-2003. [PMID: 34531546 PMCID: PMC8632895 DOI: 10.1038/s41401-021-00768-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023] Open
Abstract
We have previously shown that roflupram (ROF) protects against MPP+-induced neuronal damage in models of Parkinson's disease (PD). Since impaired degradation of α-synuclein (α-syn) is one of the key factors that lead to PD, here we investigated whether and how ROF affects the degradation of α-syn in rotenone (ROT)-induced PD models in vivo and in vitro. We showed that pretreatment with ROF (10 μM) significantly attenuated cell apoptosis and reduced the level of α-syn in ROT-treated SH-SY5Y cells. Furthermore, ROF significantly enhanced the lysosomal function, as evidenced by the increased levels of mature cathepsin D (CTSD) and lysosomal-associated membrane protein 1 (LAMP1) through increasing NAD+/NADH and the expression of sirtuin 1 (SIRT1). Pretreatment with an SIRT1 inhibitor selisistat (SELI, 10 μM) attenuated the neuroprotection of ROF, ROF-reduced expression of α-syn, and ROF-increased expression levels of LAMP1 and mature CTSD. Moreover, inhibition of CTSD by pepstatin A (20 μM) attenuated ROF-reduced expression of α-syn. In vivo study was conducted in mice exposed to ROT (10 mg·kg-1·d-1, i.g.) for 6 weeks; then, ROT-treated mice received ROF (0.5, 1, or 2 mg·kg-1·d-1; i.g.) for four weeks. ROF significantly ameliorated motor deficits, which was accompanied by increased expression levels of tyrosine hydroxylase, SIRT1, mature CTSD, and LAMP1, and a reduced level of α-syn in the substantia nigra pars compacta. Taken together, these results demonstrate that ROF exerts a neuroprotective action and reduces the α-syn level in PD models. The mechanisms underlying ROF neuroprotective effects appear to be associated with NAD+/SIRT1-dependent activation of lysosomal function.
Collapse
Affiliation(s)
- Wen-Li Dong
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Hong Zhong
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yun-Qing Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jin-Feng Xie
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Pharmacy, Guangdong Women and Children Hospital, Guangzhou, 511400, China
| | - Yun-Yun Qin
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jiang-Ping Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China.
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China.
| | - Ning-Bo Cai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Meng-Fan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lu Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hai-Tao Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China.
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Pharmacophore modeling and 3D-QSAR study for the design of novel α-synuclein aggregation inhibitors. J Mol Model 2021; 27:260. [PMID: 34432157 DOI: 10.1007/s00894-021-04881-3] [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: 05/19/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
Alpha-synuclein (α-syn), as a highly soluble presynaptic protein expressed in the brain, plays an important role in recycling synaptic vesicles and regulating the synthesis, storage, and release of neurotransmitters. Accumulation of α-syn in Lewy bodies and Lewy neurites is the pathological hallmark of Parkinson's disease (PD), so inhibition of α-syn aggregation may provide a novel approach for treating PD. In this study, the 3D structure of α-syn was downloaded from Protein Data Bank (PDB ID: 2N0A). A ligand-based pharmacophore model was conducted on a set of 43 diverse α-syn ligands, and the results suggested that two hydrogen-bond acceptors, one hydrophobic group, and two aromatic rings were significant to the inhibition of α-syn aggregation. A ligand-based 3D-QSAR model was also established with good statistical significance (R2 = 0.920) and excellent predictive ability (Q2 = 0.752). Novel indolinone derivatives were designed and synthesized based on the pharmacophore model. Subsequently, the 3D-QSAR model was used to predict the inhibitory activities towards α-syn aggregation, and the actual inhibitory activities were evaluated by thioflavin-T assay in vitro with the best inhibitory activity reaching 45.08%. The fitting results indicated that the built pharmacophore and 3D-QSAR models provided better reliability and accuracy for compound modification and prediction of the activity thereof. A ligand-based pharmacophore modeling and 3D-QSAR study have been performed on a set of 43 diverse ligands for α-synuclein for the first time. Based on the best pharmacophore modeling, novel indolinone derivatives were designed and synthesized, and the inhibitory activities for α-synuclein aggregation were evaluated by thioflavin-T assay in vitro, which preliminary indicated that five pharmacophore sites (two hydrogen bond acceptors (A), a hydrophobic group (H), and two aromatic rings (R)) in compounds contribute to the inhibitory activities. In the study, the built pharmacophore modeling and 3D-QSAR provided better reliability and accuracy for compound modification and prediction of the activity thereof.
Collapse
|
22
|
Ehrlich RS, Shiao AL, Li M, Teppang KL, Jeoung KY, Theodorakis EA, Yang J. Exploring the Effect of Aliphatic Substituents on Aryl Cyano Amides on Enhancement of Fluorescence upon Binding to Amyloid-β Aggregates. ACS Chem Neurosci 2021; 12:2946-2952. [PMID: 34270227 DOI: 10.1021/acschemneuro.1c00334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The self-assembly of amyloid-β (Aβ) peptides into amyloid aggregates is a pathological hallmark of Alzheimer's Disease. We previously reported a fluorescent Aryl Cyano Amide (ARCAM) probe that exhibits an increase in fluorescence emission upon binding to Aβ aggregates in solution and in neuronal tissue. Here, we investigate the effect of introducing small aliphatic substituents on the spectroscopic properties of ARCAM both free in solution and when bound to aggregated Aβ. We found that introducing substituents designed to hinder the rotation of bonds between the electron donor and acceptor on these fluorophores can affect the overall brightness of fluorescence emission of the probes in amyloid-free solutions, but the relative fluorescence enhancement of these probes in amyloid-containing solutions is dependent on the location of the substituents on the ARCAM scaffold. We also observed the capability to tune the excitation or emission wavelength of these probes by introducing electron-donating or -withdrawing substituents that putatively affect either the energy required for photoexcitation or the stability of the photoexcited state. These studies reveal new design principles for developing ARCAM-based fluorescent Aβ-binding probes with an enhanced fluorescence signal compared to background and tunable spectroscopic properties, which may lead to improved chemical tools for aiding in the diagnosis of amyloid-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Rachel S. Ehrlich
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Alexander L. Shiao
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Meihan Li
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Kristine L. Teppang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Kun Yong Jeoung
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Emmanuel A. Theodorakis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, United States
| |
Collapse
|
23
|
Bisi N, Feni L, Peqini K, Pérez-Peña H, Ongeri S, Pieraccini S, Pellegrino S. α-Synuclein: An All-Inclusive Trip Around its Structure, Influencing Factors and Applied Techniques. Front Chem 2021; 9:666585. [PMID: 34307295 PMCID: PMC8292672 DOI: 10.3389/fchem.2021.666585] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
Alpha-synuclein (αSyn) is a highly expressed and conserved protein, typically found in the presynaptic terminals of neurons. The misfolding and aggregation of αSyn into amyloid fibrils is a pathogenic hallmark of several neurodegenerative diseases called synucleinopathies, such as Parkinson’s disease. Since αSyn is an Intrinsically Disordered Protein, the characterization of its structure remains very challenging. Moreover, the mechanisms by which the structural conversion of monomeric αSyn into oligomers and finally into fibrils takes place is still far to be completely understood. Over the years, various studies have provided insights into the possible pathways that αSyn could follow to misfold and acquire oligomeric and fibrillar forms. In addition, it has been observed that αSyn structure can be influenced by different parameters, such as mutations in its sequence, the biological environment (e.g., lipids, endogenous small molecules and proteins), the interaction with exogenous compounds (e.g., drugs, diet components, heavy metals). Herein, we review the structural features of αSyn (wild-type and disease-mutated) that have been elucidated up to present by both experimental and computational techniques in different environmental and biological conditions. We believe that this gathering of current knowledge will further facilitate studies on αSyn, helping the planning of future experiments on the interactions of this protein with targeting molecules especially taking into consideration the environmental conditions.
Collapse
Affiliation(s)
- Nicolò Bisi
- BioCIS, CNRS, Université Paris Saclay, Châtenay-Malabry Cedex, France
| | - Lucia Feni
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| | - Kaliroi Peqini
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| | - Helena Pérez-Peña
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Sandrine Ongeri
- BioCIS, CNRS, Université Paris Saclay, Châtenay-Malabry Cedex, France
| | | | - Sara Pellegrino
- DISFARM-Dipartimento di Scienze Farmaceutiche, Sezione Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
24
|
Gao L, Wang W, Wang X, Yang F, Xie L, Shen J, Brimble MA, Xiao Q, Yao SQ. Fluorescent probes for bioimaging of potential biomarkers in Parkinson's disease. Chem Soc Rev 2021; 50:1219-1250. [DOI: 10.1039/d0cs00115e] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This review comprehensively summarizes various types of fluorescent probes for PD and their applications for detection of various PD biomarkers.
Collapse
Affiliation(s)
- Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
- Department of Chemistry
| | - Wei Wang
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
- Department of Chemistry
| | - Xuan Wang
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
| | - Fen Yang
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
| | - Liuxing Xie
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
| | - Jun Shen
- Department of Radiology
- Sun Yat-Sen Memorial Hospital
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Margaret A. Brimble
- School of Chemical Sciences
- The University of Auckland
- Auckland 1010
- New Zealand
| | - Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen)
- Sun Yat-sen University
- Shenzhen, 518107
- P. R. China
- Department of Chemistry
| | - Shao Q. Yao
- Department of Chemistry
- National University of Singapore
- Singapore
| |
Collapse
|
25
|
Kaide S, Watanabe H, Shimizu Y, Iikuni S, Nakamoto Y, Hasegawa M, Itoh K, Ono M. Identification and Evaluation of Bisquinoline Scaffold as a New Candidate for α-Synuclein-PET Imaging. ACS Chem Neurosci 2020; 11:4254-4261. [PMID: 33258582 DOI: 10.1021/acschemneuro.0c00523] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
α-Synuclein (α-syn) aggregates are pathologically associated with the hallmarks found in brains affected by synucleinopathies such as Parkinson's disease (PD) and multiple system atrophy (MSA). Therefore, the in vivo detection of α-syn aggregates using radiolabeled probes is useful for the comprehension of and medical intervention for synucleinopathies. In the present study, we identified a bisquinoline scaffold as a new promising structure for targeting α-syn aggregates by a screening assay. Then, based on the scaffold, novel bisquinoline derivatives, BQ1 and BQ2, were designed and synthesized, and we evaluated their utilities as α-syn imaging probes. Both compounds showed high affinity for recombinant α-syn aggregates in binding assays in vitro and clearly detected α-syn aggregates in human brain sections. BQ2 showed higher affinity for α-syn aggregates than BQ1, leading to performing 18F-labeling to obtain [18F]BQ2. In a biodistribution study using normal mice, [18F]BQ2 displayed moderate uptake (1.59% ID/g at 2 min postinjection) into but subsequent retention (1.35% ID/g at 60 min postinjection) in the brain. The results of this study suggest that a bisquinoline derivative may be a new candidate as an α-syn-PET imaging probe after appropriate structure modification for further improvement in the pharmacokinetics.
Collapse
Affiliation(s)
- Sho Kaide
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoichi Shimizu
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masato Hasegawa
- Department of Brain and Neurosciences, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Kyoko Itoh
- Department of Pathology & Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
26
|
Emerging novel approaches to drug research and diagnosis of Parkinson's disease. Acta Pharmacol Sin 2020; 41:439-441. [PMID: 32203079 PMCID: PMC7471400 DOI: 10.1038/s41401-020-0369-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/11/2022] Open
|