1
|
Ono K, Noguchi-Shinohara M, Watanabe-Nakayama T. The basis of anti-Aβ antibody therapy: The toxicity of Aβ aggregates and the mechanism of action of anti-Aβ antibodies. Intern Med 2024:4569-24. [PMID: 39370248 DOI: 10.2169/internalmedicine.4569-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Abstract
In the pathophysiology of Alzheimer's disease (AD), the amyloid hypothesis, which posits that amyloid β-protein (Aβ) abnormally aggregates and damages neurons with tau, has been proposed. It was originally thought that the accumulation of insoluble amyloid fibrils in the brain leads to AD-inducing neurotoxicity; however, in recent years, the positioning of early and intermediate aggregates has also been emphasized. In particular, following the positive results of phase 3 clinical trials of lecanemab and its approval in Japan and the United States, the pathology of protofibrils, which are the target molecules of lecanemab, has attracted attention. Using high-speed atomic force microscopy, we have previously reported that lecanemab, which has a high affinity for protofibrils, binds to and surrounds them. Donanemab, a recombinant monoclonal antibody that primarily targets fibrils composed of N3pG Aβ, has also attracted attention because of its efficacy in phase 3 clinical trials in patients with early stage AD.
Collapse
Affiliation(s)
- Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Japan
| | - Moeko Noguchi-Shinohara
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Japan
| | | |
Collapse
|
2
|
Dabas A, Goyal B. Structural Reorganization Mechanism of the Aβ 42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353. ACS Chem Neurosci 2024; 15:3136-3151. [PMID: 39158263 DOI: 10.1021/acschemneuro.4c00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer's disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ42 fibril (ΔGbinding= -142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ42 fibril. The illumination of key interactions responsible for the destabilization of the Aβ42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.
Collapse
Affiliation(s)
- Arushi Dabas
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| |
Collapse
|
3
|
Kamburova K, Dimitrov IL, Hodzhaoglu F, Milkova V. Investigation of the Aggregation of Aβ Peptide (1-40) in the Presence of κ-Carrageenan-Stabilised Liposomes Loaded with Homotaurine. Molecules 2024; 29:3460. [PMID: 39124866 PMCID: PMC11313970 DOI: 10.3390/molecules29153460] [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: 07/07/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
The kinetics of amyloid aggregation was studied indirectly by monitoring the changes in the polydispersity of mixed dispersion of amyloid β peptide (1-40) and composite liposomes. The liposomes were prepared from the 1,2-dioleoyl-sn-glicero-3-phoshocholine (DOPC) phospholipid and stabilised by the electrostatic adsorption of κ-carrageenan. The produced homotaurine-loaded and unloaded liposomes had a highly negative electrokinetic potential and remarkable stability in phosphate buffer (pH 4 and 7.4). For the first time, the appearance and evolution of the aggregation of Aβ were presented through the variation in the standard percentile readings (D10, D50, and D90) obtained from the particle size distribution analysis. The kinetic experiments indicated the appearance of the first aggregates almost 30 min after mixing the liposomes and peptide solution. It was observed that by adding unloaded liposomes, the size of 90% of the particles in the dispersion (D90) increased. In contrast, the addition of homotaurine-loaded liposomes had almost minimal impact on the size of the fractions of larger particles during the kinetic experiments. Despite the specific bioactivity of homotaurine in the presence of natural cell membranes, this study reported an additional inhibitory effect of the compound on the amyloid peptide aggregation due to the charge effects and 'molecular crowding'.
Collapse
Affiliation(s)
| | | | | | - Viktoria Milkova
- Institute of Physical Chemistry ‘Acad. R. Kaischew’, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (K.K.); (I.L.D.); (F.H.)
| |
Collapse
|
4
|
Sánchez-Puebla L, López-Cuenca I, Salobrar-García E, González-Jiménez M, Arias-Vázquez A, Matamoros JA, Ramírez AI, Fernández-Albarral JA, Elvira-Hurtado L, Saido TC, Saito T, Nieto-Vaquero C, Cuartero MI, Moro MA, Salazar JJ, de Hoz R, Ramírez JM. Retinal Vascular and Structural Changes in the Murine Alzheimer's APPNL-F/NL-F Model from 6 to 20 Months. Biomolecules 2024; 14:828. [PMID: 39062542 PMCID: PMC11274728 DOI: 10.3390/biom14070828] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) may manifest retinal changes preceding brain pathology. A transversal case-control study utilized spectral-domain OCT angiography (SD-OCTA) and Angio-Tool software 0.6a to assess retinal vascular structures and OCT for inner and outer retina thickness in the APPNL-F/NL-F AD model at 6, 9, 12, 15, 17, and 20 months old. Comparisons to age-matched wild type (WT) were performed. The analysis focused on the three vascular plexuses using AngiooTool and on retinal thickness, which was represented with the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Compared to WT, the APPNL-F/NL-F group exhibited both vascular and structural changes as early as 6 months persisting and evolving at 15, 17, and 20 months. Significant vascular alterations, principally in the superficial vascular complex (SVC), were observed. There was a significant decrease in the vessel area and the total vessel length in SVC, intermediate, and deep capillary plexus. The inner retina in the APPNL-F/NL-F group predominantly decreased in thickness while the outer retina showed increased thickness in most analyzed time points compared to the control group. There are early vascular and structural retinal changes that precede the cognitive changes, which appear at later stages. Therefore, the natural history of the APPNL-F/NL-F model may be more similar to human AD than other transgenic models.
Collapse
Affiliation(s)
- Lidia Sánchez-Puebla
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Inés López-Cuenca
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Elena Salobrar-García
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - María González-Jiménez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Alberto Arias-Vázquez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - José A. Matamoros
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana I. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José A. Fernández-Albarral
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, Brain Science Institute, RIKEN, Wako 351-0198, Japan;
| | - Takashi Saito
- Institute of Brain Science, Faculty of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan;
| | - Carmen Nieto-Vaquero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María I. Cuartero
- Hospital 12 de Octubre Research Institute (i + 12), 28029 Madrid, Spain;
- University Institute for Research in Neurochemistry, Complutense University of Madrid (UCM), 28040 Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - María A. Moro
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Neurovascular Pathophysiology, Cardiovascular Risk Factor and Brain Function Programme, 28029 Madrid, Spain; (C.N.-V.); (M.A.M.)
| | - Juan J. Salazar
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - Rosa de Hoz
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Faculty of Optics and Optometry, Complutense University of Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Ramon Castroviejo Institute for Ophthalmic Research, Complutense University of Madrid, 28040 Madrid, Spain; (L.S.-P.); (I.L.-C.); (E.S.-G.); (M.G.-J.); (A.A.-V.); (J.A.M.); (A.I.R.); (J.A.F.-A.); (L.E.-H.); (J.J.S.)
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
5
|
Kaku T, Ikebukuro K, Tsukakoshi K. Structure of cytotoxic amyloid oligomers generated during disaggregation. J Biochem 2024; 175:575-585. [PMID: 38430131 PMCID: PMC11155694 DOI: 10.1093/jb/mvae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Amyloidosis is characterized by the abnormal accumulation of amyloid proteins. The causative proteins aggregate from monomers to oligomers and fibrils, among which some intermediate oligomers are considered as major toxins. Cytotoxic oligomers are generated not only by aggregation but also via fibril disaggregation. However, little is known about the structural characteristics and generation conditions of cytotoxic oligomers produced during disaggregation. Herein, we summarized the structural commonalities of cytotoxic oligomers formed under various disaggregation conditions, including the addition of heat shock proteins or small compounds. In vitro experimental data demonstrated the presence of high-molecular-weight oligomers (protofibrils or protofilaments) that exhibited a fibrous morphology and β-sheet structure. Molecular dynamics simulations indicated that the distorted β-sheet structure contributed to their metastability. The tendency of these cytotoxic oligomers to appear under mild disaggregation conditions, implied formation during the early stages of disaggregation. This review will aid researchers in exploring the characteristics of highly cytotoxic oligomers and developing drugs that target amyloid aggregates.
Collapse
Affiliation(s)
- Toshisuke Kaku
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
6
|
Liu ZL, Hua FF, Qu L, Yan N, Zhang HF. Evaluating serum CXCL12, sCD22, Lp-PLA2 levels and ratios as biomarkers for diagnosis of Alzheimer's disease. World J Psychiatry 2024; 14:380-387. [PMID: 38617987 PMCID: PMC11008386 DOI: 10.5498/wjp.v14.i3.380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/15/2024] [Accepted: 02/04/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Grasping the underlying mechanisms of Alzheimer's disease (AD) is still a work in progress, and existing diagnostic techniques encounter various obstacles. Therefore, the discovery of dependable biomarkers is essential for early detection, tracking the disease's advancement, and steering treatment strategies. AIM To explore the diagnostic potential of serum CXCL12, sCD22, Lp-PLA2, and their ratios in AD, aiming to enhance early detection and inform targeted treatment strategies. METHODS The study was conducted in Dongying people's Hospital from January 2021 to December 2022. Participants included 60 AD patients (AD group) and 60 healthy people (control group). Using a prospective case-control design, the levels of CXCL12, sCD22 and Lp-PLA2 and their ratios were detected by enzyme-linked immunosorbent assay kit in the diagnosis of AD. The differences between the two groups were analyzed by statistical methods, and the corresponding ratio was constructed to improve the specificity and sensitivity of diagnosis. RESULTS Serum CXCL12 levels were higher in the AD group (47.2 ± 8.5 ng/mL) than the control group (32.8 ± 5.7 ng/mL, P < 0.001), while sCD22 levels were lower (14.3 ± 2.1 ng/mL vs 18.9 ± 3.4 ng/mL, P < 0.01). Lp-PLA2 levels were also higher in the AD group (112.5 ± 20.6 ng/mL vs 89.7 ± 15.2 ng/mL, P < 0.05). Significant differences were noted in CXCL12/sCD22 (3.3 vs 1.7, P < 0.001) and Lp-PLA2/sCD22 ratios (8.0 vs 5.2, P < 0.05) between the groups. Receiver operating characteristic analysis confirmed high sensitivity and specificity of these markers and their ratios in distinguishing AD, with area under the curves ranging from 0.568 to 0.787. CONCLUSION Serum CXCL12 and Lp-PLA2 levels were significantly increased, while sCD22 were significantly decreased, as well as increases in the ratios of CXCL12/sCD22 and Lp-PLA2/sCD22, are closely related to the onset of AD. These biomarkers and their ratios can be used as potential diagnostic indicators for AD, providing an important clinical reference for early intervention and treatment.
Collapse
Affiliation(s)
- Zeng-Ling Liu
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Fei-Fei Hua
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Lei Qu
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Na Yan
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| | - Hui-Fang Zhang
- Department of Neurology, Dongying People's Hospital, Dongying 257000, Shandong Province, China
| |
Collapse
|
7
|
Wang K, Cai W. Binding mechanism of full-length Aβ40 peptide to a mixed lipid bilayer. Front Chem 2024; 12:1367793. [PMID: 38449479 PMCID: PMC10914957 DOI: 10.3389/fchem.2024.1367793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
The destructive effect of Aβ peptides on membranes is an important source of its cytotoxicity in the pathogenesis of Alzheimer's disease. We have investigated the binding mechanism between the Aβ42 peptide and bilayer in our former work. However, as another abundant form of Aβ peptides in the physiological environment, the binding mechanism between Aβ40 peptide and the lipid bilayer still remains ambiguous. Hence, we performed all-atom simulations on the Aβ40 peptides with the lipid bilayer herein using replica exchange with the solute tempering 2 method. We obtained four major binding models with the hydrophobic C-terminus as the most preferable binding region. Hydrophobic residues and positively charged residues are the principal residues involved in the peptide-bilayer interactions. Aβ40 peptides in our simulation mainly adopt a β-rich conformation in both bound and unbound states. Besides, we determined peptide-water interactions and found that bound peptides prefer forming hydrogen bonds with water molecules than unbound peptides. Our findings herein may provide new insights for the in-depth understanding of the membrane-destructive mechanism of Aβ peptides.
Collapse
Affiliation(s)
| | - Wensheng Cai
- Research Center for Analytical Sciences, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin, China
| |
Collapse
|
8
|
Tsukano C, Uchino A, Irie K. Synthesis and applications of symmetric amino acid derivatives. Org Biomol Chem 2024; 22:411-428. [PMID: 37877370 DOI: 10.1039/d3ob01379k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Symmetric α-amino acid derivatives can be used for the synthesis of intermolecularly linked peptides such as dimer-type peptides, and modified peptides in which two amino acids are intramolecularly linked. They are also synthetic intermediates for the total synthesis of natural products and functional molecules. These symmetric amino acid derivatives must be prepared based on organic synthesis. It is necessary to develop an optimal synthetic strategy for constructing the target symmetric amino acid derivative. In this review, we will introduce strategies for synthesizing symmetric amino acid derivatives. Additionally, selected applications of these amino acids in the life sciences will be described.
Collapse
Affiliation(s)
- Chihiro Tsukano
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Ayumi Uchino
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
9
|
Chen C, Yan ZS, Ma YQ, Ding HM. Effect of Terahertz Waves on the Structure of the Aβ42 Monomer, Dimer, and Protofibril: Insights from Molecular Dynamics Simulations. ACS Chem Neurosci 2023; 14:4128-4138. [PMID: 37983764 DOI: 10.1021/acschemneuro.3c00485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023] Open
Abstract
Amyloid-β (Aβ) and its assemblies play important roles in the pathogenesis of Alzheimer's disease (AD). Recent studies conducted by experimental and computational researchers have extensively explored the structure, assembly, and influence of biomolecules and cell membranes on Aβ. However, the impact of terahertz waves on the structures of Aβ monomers and aggregates remains largely unexplored. In this study, we systematically investigate the molecular mechanisms by which terahertz waves affect the structure of the Aβ42 monomer, dimer, and tetramer through all-atom molecular dynamics (MD) simulations. Our findings indicate that terahertz waves at a specific frequency (42.55 THz) can enhance intramolecular and intermolecular interactions in the Aβ42 monomer and dimer, respectively, by resonating with the symmetric stretching mode of the -COO- groups and the symmetric bending/stretching mode of -CH3 groups. Consequently, the β-structure content of the Aβ42 monomer is greatly increased, and the binding energy between the monomers in the Aβ42 dimer is significantly enhanced. Additionally, our observations suggest that terahertz waves can mildly stabilize the structure of tetrameric protofibrils by enhancing the interactions among peripheral peptides. Furthermore, we also investigated the effect of the frequency of terahertz waves on the structure of Aβ42. The present study contributes to a better understanding of the impact of external fields on the biobehavior of Aβ42 peptides and may shed some light on the potential risks associated with electromagnetic field radiation.
Collapse
Affiliation(s)
- Chen Chen
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Zeng-Shuai Yan
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu-Qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| |
Collapse
|
10
|
Akasaka T, Watanabe H, Ono M. In Vivo Near-Infrared Fluorescence Imaging Selective for Soluble Amyloid β Aggregates Using y-Shaped BODIPY Derivative. J Med Chem 2023; 66:14029-14046. [PMID: 37824378 DOI: 10.1021/acs.jmedchem.3c01057] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Soluble amyloid β (Aβ) aggregates, suggested to be the most toxic forms of Aβ, draw attention as therapeutic targets and biomarkers of Alzheimer's disease (AD). As soluble Aβ aggregates are transient and diverse, imaging their diverse forms in vivo is expected to have a marked impact on research and diagnosis of AD. Herein, we report a near-infrared fluorescent (NIRF) probe, BAOP-16, targeting diverse soluble Aβ aggregates. BAOP-16, whose molecular shape resembles "y", showed a marked selective increase in fluorescence intensity upon binding to soluble Aβ aggregates in the near-infrared region and a high binding affinity for them. Additionally, BAOP-16 could detect Aβ oligomers in the brains of Aβ-inoculated model mice. In an in vivo fluorescence imaging study of BAOP-16, brains of AD model mice displayed significantly higher fluorescence signals than those of wild-type mice. These results indicate that BAOP-16 could be useful for the in vivo NIRF imaging of diverse soluble Aβ aggregates.
Collapse
Affiliation(s)
- Takahiro Akasaka
- 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
| | - 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
|
11
|
Taniwa K, Murakami K, Sakaguchi Y, Izuo N, Hanaki M, Sampa N, Kume T, Shimizu T, Irie K. Detection of Dietary Chalcone and Flavonoid Metabolites in Mice Using UPLC-MS/MS and Their Modulatory Effects on Amyloid β Aggregation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14289-14299. [PMID: 37702279 DOI: 10.1021/acs.jafc.3c02598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Amyloid β-protein (Aβ42) aggregates have been demonstrated to induce cognitive decline and neurodegeneration in Alzheimer's disease (AD). Thus, functional food ingredients that inhibit Aβ42 aggregation are valuable for AD prevention. Although several food ingredients have been studied for their anti-aggregation activity, information on their bioavailability in the brain, incorporated forms, and relevance to AD etiology is limited. Here, we first detected the sulfate- and glucuronic-acid-conjugated forms of green perilla-derived chalcone (1) and taxifolin (2), which inhibit Aβ42 aggregation, in the brain, small intestine, and plasma of mice (1 and 2 were administered orally) using ultra-performance liquid chromatography-tandem mass spectrometry. We observed that the conjugated metabolites (sulfate (4) and glucuronide (5)) of 1 prevented the fibrillization and oligomerization of Aβ42. These findings imply that the conjugated metabolites of 1 can prove beneficial for AD treatment.
Collapse
Affiliation(s)
- Kota Taniwa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Yoshiki Sakaguchi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Naotaka Izuo
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Mizuho Hanaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Nobuaki Sampa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Takahiko Shimizu
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
12
|
Ito N, Tsuji M, Adachi N, Nakamura S, Sarkar AK, Ikenaka K, Aguirre C, Kimura AM, Kiuchi Y, Mochizuki H, Teplow DB, Ono K. Extracellular high molecular weight α-synuclein oligomers induce cell death by disrupting the plasma membrane. NPJ Parkinsons Dis 2023; 9:139. [PMID: 37770475 PMCID: PMC10539356 DOI: 10.1038/s41531-023-00583-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/20/2023] [Indexed: 09/30/2023] Open
Abstract
α-Synuclein (αS), the causative protein of Parkinson's disease and other α-synucleinopathies, aggregates from a low molecular weight form (LMW-αS) to a high molecular weight αS oligomer (HMW-αSo). Aggregated αS accumulates intracellularly, induces intrinsic apoptosis, is released extracellularly, and appears to propagate disease through prion-like spreading. Whether extracellular αS aggregates are cytotoxic, damage cell wall, or induce cell death is unclear. We investigated cytotoxicity and cell death caused by HMW-αSo or LMW-αS. Extracellular HMW-αSo was more cytotoxic than LMW-αS and was a crucial factor for inducing plasma membrane damage and cell death. HMW-αSo induced reactive oxygen species production and phospholipid peroxidation in the membrane, thereby impairing calcium homeostasis and disrupting plasma membrane integrity. HMW-αSo also induced extrinsic apoptosis and cell death by activating acidic sphingomyelinase. Thus, as extracellular HMW-αSo causes neuronal injury and death via cellular transmission and direct plasma membrane damage, we propose an additional disease progression pathway for α-synucleinopathies.
Collapse
Affiliation(s)
- Naohito Ito
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan.
| | - Naoki Adachi
- Department of Physiology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Avijite Kumer Sarkar
- Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229-3026, USA
| | - Kensuke Ikenaka
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - César Aguirre
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Atsushi Michael Kimura
- Brain Research Institute Center for Integrated Human Brain Science, Department of Functional Neurology and Neurosurgery, Niigata University, Niigata, 951-8122, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - David B Teplow
- Department of Neurology, David Geffen School of Medicine, University of California-Los Angeles (UCLA), Los Angeles, LA, 10833, USA
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8640, Japan.
| |
Collapse
|
13
|
Santacruz CA, Vincent JL, Imbault V, Bruneau M, Creteur J, Brimioulle S, Vincent R, Communi D, Taccone FS. Cerebral apolipoprotein E and amyloid precursor-like protein 1 as risk factors for chronic neurodegeneration after non-traumatic acute brain injury (ABI). Crit Care 2023; 27:249. [PMID: 37355618 PMCID: PMC10290338 DOI: 10.1186/s13054-023-04538-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023] Open
Affiliation(s)
- Carlos A Santacruz
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.
- Department of Intensive and Critical Care Medicine, Fundación Santa Fe de Bogota, Bogota, Colombia.
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Virginie Imbault
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels, Belgium
| | - Michael Bruneau
- Laboratory for the Structure and Function of Biological Membranes, Center for Structural Biology and Bioinformatics, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Creteur
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Serge Brimioulle
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Raussens Vincent
- Structural Biology and Bioinformatics Center, Structure and Function of Biological Membranes, Faculty of Science, Université Libre de Bruxelles, Brussels, Belgium
| | - David Communi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabio S Taccone
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
14
|
Gupta S, Dasmahapatra AK. Enhanced stability of a disaggregated Aβ fibril on removal of ligand inhibits refibrillation: An all atom Molecular Dynamics simulation study. Int J Biol Macromol 2023; 240:124481. [PMID: 37076062 DOI: 10.1016/j.ijbiomac.2023.124481] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
The extraneuronally deposited senile plaques, composed of neurotoxic aggregates of Aβ fibril, define Alzheimer's disease (AD). Natural compounds have been tested for their destabilization potential on Aβ fibril, thereby curing AD. However, the resultant destabilized Aβ fibril, needs to be checked for its irreversibility to the native organized state after removal of the ligand. Herein, we assessed the stability of a destabilized fibril after the ligand (ellagic acid represented as REF) is removed from the complex. The study has been conducted via Molecular Dynamics (MD) simulation of 1 μs for both Aβ-Water (control) and Aβ-REF″ (test or REF removed) system. The increased value of RMSD, Rg, SASA, lower β-sheet content and reduced number of H-bonds explains enhanced destabilization observed in Aβ-REF″ system. The increased inter-chain distance demonstrates breaking of the residual contacts, testifying the drift of terminal chains from the pentamer. The increased SASA along with the ∆Gps(polar solvation energy) accounts for the reduced interaction amongst residues, and more with solvent molecules, governing irreversibility to native state. The higher Gibb's free energy of the misaligned structure of Aβ-REF″ ensures irreversibility to the organized structure due to its inability to cross such high energy barrier. The observed stability of the disaggregated structure, despite ligand elimination, establishes the effectiveness of the destabilization technique as a promising therapeutic approach towards treating AD.
Collapse
Affiliation(s)
- Shivani Gupta
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Ashok Kumar Dasmahapatra
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India; Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
15
|
Momma Y, Tsuji M, Oguchi T, Ohashi H, Nohara T, Ito N, Yamamoto K, Nagata M, Kimura AM, Nakamura S, Kiuchi Y, Ono K. The Curcumin Derivative GT863 Protects Cell Membranes in Cytotoxicity by Aβ Oligomers. Int J Mol Sci 2023; 24:ijms24043089. [PMID: 36834498 PMCID: PMC9960433 DOI: 10.3390/ijms24043089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
In Alzheimer's disease (AD), accumulation of amyloid β-protein (Aβ) is one of the major mechanisms causing neuronal cell damage. Disruption of cell membranes by Aβ has been hypothesized to be the important event associated with neurotoxicity in AD. Curcumin has been shown to reduce Aβ-induced toxicity; however, due to its low bioavailability, clinical trials showed no remarkable effect on cognitive function. As a result, GT863, a derivative of curcumin with higher bioavailability, was synthesized. The purpose of this study is to clarify the mechanism of the protective action of GT863 against the neurotoxicity of highly toxic Aβ oligomers (Aβo), which include high-molecular-weight (HMW) Aβo, mainly composed of protofibrils in human neuroblastoma SH-SY5Y cells, focusing on the cell membrane. The effect of GT863 (1 μM) on Aβo-induced membrane damage was assessed by phospholipid peroxidation of the membrane, membrane fluidity, membrane phase state, membrane potential, membrane resistance, and changes in intracellular Ca2+ ([Ca2+]i). GT863 inhibited the Aβo-induced increase in plasma-membrane phospholipid peroxidation, decreased membrane fluidity and resistance, and decreased excessive [Ca2+]i influx, showing cytoprotective effects. The effects of GT863 on cell membranes may contribute in part to its neuroprotective effects against Aβo-induced toxicity. GT863 may be developed as a prophylactic agent for AD by targeting inhibition of membrane disruption caused by Aβo exposure.
Collapse
Affiliation(s)
- Yutaro Momma
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
- Correspondence: (M.T.); (K.O.)
| | - Tatsunori Oguchi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Hideaki Ohashi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Tetsuhito Nohara
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Naohito Ito
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Ken Yamamoto
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Miki Nagata
- Department of Hospital Pharmaceutics, School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | - Atsushi Michael Kimura
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
- Correspondence: (M.T.); (K.O.)
| |
Collapse
|
16
|
Gupta S, Dasmahapatra AK. Destabilization of Aβ fibrils by omega-3 polyunsaturated fatty acids: a molecular dynamics study. J Biomol Struct Dyn 2023; 41:581-598. [PMID: 34856889 DOI: 10.1080/07391102.2021.2009915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The senile plaques of neurotoxic aggregates of Aβ protein, deposited extraneuronally, mark the pathological hallmark of Alzheimer's disease (AD). The natural compounds such as omega-3 (ω-3) polyunsaturated fatty acids (PUFAs), which can access blood-brain barrier, are believed to be potential disruptors of preformed Aβ fibrils to cure AD with unknown mechanism. Herein, we present the destabilization potential of three ω-3 PUFAs, viz. Eicosapentaenoic acid (EPA), Docosahexaenoic acid (HXA), and α-linolenic acid (LNL) by molecular dynamics simulation. After an initial testing of 300 ns, EPA and HXA have been considered further for extended production run time, 500 ns. The increased value of root mean square deviation (RMSD), radius of gyration, and solvent-accessible surface area (SASA), the reduced number of H-bonds and β-sheet content, and disruption of salt bridges and hydrophobic contacts establish the binding of these ligands to Aβ fibril leading to destabilization. The polar head was found to interact with positively charged lysine (K28) residue in the fibril. However, the hydrophobicity of the long aliphatic tail competes with the intrinsic hydrophobic interactions of Aβ fibril. This amphiphilic nature of EPA and HXA led to the breaking of inherent hydrophobic contacts and formation of new bonds between the tail of PUFA and hydrophobic residues of Aβ fibril, leading to the destabilization of fibril. The Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) results explain the binding of EPA and HXA to Aβ fibril by interacting with different residues. The destabilization potential of EPA and HXA establishes them as promising drug leads to cure AD, and encourages prospecting of other fatty acids for therapeutic intervention in AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shivani Gupta
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Ashok Kumar Dasmahapatra
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.,Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
17
|
Noguchi-Shinohara M, Hamaguchi T, Sakai K, Komatsu J, Iwasa K, Horimoto M, Nakamura H, Yamada M, Ono K. Effects of Melissa officinalis Extract Containing Rosmarinic Acid on Cognition in Older Adults Without Dementia: A Randomized Controlled Trial. J Alzheimers Dis 2023; 91:805-814. [PMID: 36502333 DOI: 10.3233/jad-220953] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous in vitro and in vivo studies on Alzheimer's disease (AD) models have reported that rosmarinic acid (RA) can inhibit the formation of amyloid-β fibrils as well as the oligomerization and deposition of amyloid-β protein. Melissa officinalis (M. officinalis) extract containing 500 mg of RA is tolerable and safe in healthy individuals and patients with mild AD dementia. OBJECTIVE This randomized placebo-controlled double-blind trial aimed to assess the effects of M. officinalis extract on cognition in older adults without dementia. METHODS This study included individuals who were diagnosed with subjective or mild cognitive impairment (n = 323). The trial involved M. officinalis extract supplementation (500 mg of RA per day) period of 96 weeks followed by a washout period of 24 weeks. The primary endpoint was the Alzheimer's Disease Assessment Scale-cognitive subscale score, and the secondary endpoints were other cognitive measure results as well as safety and tolerability. RESULTS There were no significant differences in cognitive measures between the placebo and M. officinalis groups from baseline to 96 weeks. However, based on the analysis of Clinical Dementia Rating Sum of Boxes scores in participants without hypertension, the score was found to be increased by 0.006 and decreased by 0.085 in the M. officinalis and placebo groups, respectively; this difference was statistically significant (p = 0.036). Furthermore, there were no differences in vital signs, physical and neurological measures, or hippocampal volume between the two groups. CONCLUSION These results indicate that M. officinalis extract may help prevent cognitive decline in older adults without hypertension.
Collapse
Affiliation(s)
- Moeko Noguchi-Shinohara
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Neurology, Kanazawa Medical University, Uchinada, Kahoku, Ishikawa, Japan
| | - Kenji Sakai
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Junji Komatsu
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Preemptive Medicine for Dementia, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kazuo Iwasa
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University, Hakui, Ishikawa, Japan
| | - Mai Horimoto
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Preemptive Medicine for Dementia, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hiroyuki Nakamura
- Department of Environmental and Preventive Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masahito Yamada
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Kudanzaka Hospital, Tokyo, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
18
|
Wang J, Liu YM, Hu J, Chen C. Trained immunity in monocyte/macrophage: Novel mechanism of phytochemicals in the treatment of atherosclerotic cardiovascular disease. Front Pharmacol 2023; 14:1109576. [PMID: 36895942 PMCID: PMC9989041 DOI: 10.3389/fphar.2023.1109576] [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: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
Atherosclerosis (AS) is the pathology of atherosclerotic cardiovascular diseases (ASCVD), characterized by persistent chronic inflammation in the vessel wall, in which monocytes/macrophages play a key role. It has been reported that innate immune system cells can assume a persistent proinflammatory state after short stimulation with endogenous atherogenic stimuli. The pathogenesis of AS can be influenced by this persistent hyperactivation of the innate immune system, which is termed trained immunity. Trained immunity has also been implicated as a key pathological mechanism, leading to persistent chronic inflammation in AS. Trained immunity is mediated via epigenetic and metabolic reprogramming and occurs in mature innate immune cells and their bone marrow progenitors. Natural products are promising candidates for novel pharmacological agents that can be used to prevent or treat cardiovascular diseases (CVD). A variety of natural products and agents exhibiting antiatherosclerotic abilities have been reported to potentially interfere with the pharmacological targets of trained immunity. This review describes in as much detail as possible the mechanisms involved in trained immunity and how phytochemicals of this process inhibit AS by affecting trained monocytes/macrophages.
Collapse
Affiliation(s)
- Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Yong-Mei Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| |
Collapse
|
19
|
Chikugo A, Irie Y, Tsukano C, Uchino A, Maki T, Kume T, Kawase T, Hirose K, Kageyama Y, Tooyama I, Irie K. Optimization of the Linker Length in the Dimer Model of E22P-Aβ40 Tethered at Position 38. ACS Chem Neurosci 2022; 13:2913-2923. [PMID: 36095282 DOI: 10.1021/acschemneuro.2c00436] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Since amyloid β (Aβ) oligomers are more cytotoxic than fibrils, various dimer models have been synthesized. We focused on the C-terminal region that could form a hydrophobic core in the aggregation process and identified a toxic conformer-restricted dimer model (E22P,G38DAP-Aβ40 dimer) with an l,l-2,6-diaminopimelic acid linker (n = 3) at position 38, which exhibited moderate cytotoxicity. We synthesized four additional linkers (n = 2, 4, 5, 7) to determine the most appropriate distance between the two Aβ40 monomers for a toxic dimer model. Each di-Fmoc-protected two-valent amino acid was synthesized from a corresponding dialdehyde or cycloalkene followed by ozonolysis, using a Horner-Wadsworth-Emmons reaction and asymmetric hydrogenation. Then, the corresponding Aβ40 dimer models with these linkers at position 38 were synthesized using the solid-phase Fmoc strategy. Their cytotoxicity toward SH-SY5Y cells suggested that the shorter the linker length, the stronger the cytotoxicity. Particularly, the E22P,G38DAA-Aβ40 dimer (n = 2) formed protofibrillar aggregates and exhibited the highest cytotoxicity, equivalent to E22P-Aβ42, the most cytotoxic analogue of Aβ42. Ion mobility-mass spectrometry (IM-MS) measurement indicated that all dimer models except the E22P,G38DAA-Aβ40 dimer existed as stable oligomers (12-24-mer). NativePAGE analysis supported the IM-MS data, but larger oligomers (30-150-mer) were also detected after a 24 h incubation. Moreover, E22P,G38DAA-Aβ40, E22P,G38DAP-Aβ40, and E22P,G38DAZ-Aβ40 (n = 5) dimers suppressed long-term potentiation (LTP). Overall, the ability to form fibrils with cross β-sheet structures was key to achieving cytotoxicity, and forming stable oligomers less than 150-mer did not correlate with cytotoxicity and LTP suppression.
Collapse
Affiliation(s)
- Ayaka Chikugo
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto606-8502, Japan
| | - Yumi Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto606-8502, Japan
| | - Chihiro Tsukano
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto606-8502, Japan
| | - Ayumi Uchino
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto606-8502, Japan
| | - Takahito Maki
- Department of Applied Pharmacology, Graduate School of Medical and Pharmaceutical Sciences, University of Toyama, Toyama930-0194, Japan
| | - Toshiaki Kume
- Department of Applied Pharmacology, Graduate School of Medical and Pharmaceutical Sciences, University of Toyama, Toyama930-0194, Japan
| | | | | | - Yusuke Kageyama
- Molecular Neuroscience Research Center, Shiga University of Medical Sciences, Shiga520-2192, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Sciences, Shiga520-2192, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto606-8502, Japan
| |
Collapse
|
20
|
Murakami K, Ono K. Interactions of amyloid coaggregates with biomolecules and its relevance to neurodegeneration. FASEB J 2022; 36:e22493. [PMID: 35971743 DOI: 10.1096/fj.202200235r] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 01/16/2023]
Abstract
The aggregation of amyloidogenic proteins is a pathological hallmark of various neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. In these diseases, oligomeric intermediates or toxic aggregates of amyloids cause neuronal damage and degeneration. Despite the substantial effort made over recent decades to implement therapeutic interventions, these neurodegenerative diseases are not yet understood at the molecular level. In many cases, multiple disease-causing amyloids overlap in a sole pathological feature or a sole disease-causing amyloid represents multiple pathological features. Various amyloid pathologies can coexist in the same brain with or without clinical presentation and may even occur in individuals without disease. From sparse data, speculation has arisen regarding the coaggregation of amyloids with disparate amyloid species and other biomolecules, which are the same characteristics that make diagnostics and drug development challenging. However, advances in research related to biomolecular condensates and structural analysis have been used to overcome some of these challenges. Considering the development of these resources and techniques, herein we review the cross-seeding of amyloidosis, for example, involving the amyloids amyloid β, tau, α-synuclein, and human islet amyloid polypeptide, and their cross-inhibition by transthyretin and BRICHOS. The interplay of nucleic acid-binding proteins, such as prions, TAR DNA-binding protein 43, fused in sarcoma/translated in liposarcoma, and fragile X mental retardation polyglycine, with nucleic acids in the pathology of neurodegeneration are also described, and we thereby highlight the potential clinical applications in central nervous system therapy.
Collapse
Affiliation(s)
- Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kenjiro Ono
- Department of Neurology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
21
|
Ohashi H, Tsuji M, Oguchi T, Momma Y, Nohara T, Ito N, Yamamoto K, Nagata M, Kimura AM, Kiuchi Y, Ono K. Combined Treatment with Curcumin and Ferulic Acid Suppressed the Aβ-Induced Neurotoxicity More than Curcumin and Ferulic Acid Alone. Int J Mol Sci 2022; 23:ijms23179685. [PMID: 36077082 PMCID: PMC9456505 DOI: 10.3390/ijms23179685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease that leads to progressive cognitive decline. Several effective natural components have been identified for the treatment of AD. However, it is difficult to obtain conclusive evidence on the safety and effectiveness of natural components, because a variety of factors are associated with the progression of AD pathology. We hypothesized that a therapeutic effect could be achieved by combining multiple ingredients with different efficacies. The purpose of this study was thus to evaluate a combination treatment of curcumin (Cur) and ferulic acid (FA) for amyloid-β (Aβ)-induced neuronal cytotoxicity. The effect of Cur or FA on Aβ aggregation using thioflavin T assay was confirmed to be inhibited in a concentration-dependent manner by Cur single or Cur + FA combination treatment. The effects of Cur + FA on the cytotoxicity of human neuroblastoma (SH-SY5Y) cells induced by Aβ exposure were an increase in cell viability, a decrease in ROS and mitochondrial ROS, and repair of membrane damage. Combination treatment showed an overall higher protective effect than treatment with Cur or FA alone. These results suggest that the combined action mechanisms of Cur and FA may be effective in preventing and suppressing the progression of AD.
Collapse
Affiliation(s)
- Hideaki Ohashi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
- Correspondence: (M.T.); (K.O.)
| | - Tatsunori Oguchi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Yutaro Momma
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Tetsuhito Nohara
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Naohito Ito
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Ken Yamamoto
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Miki Nagata
- Department of Hospital Pharmaceutics, School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | - Atsushi Michael Kimura
- Division of Neurology, Department of Internal Medicine, School of Medicine, Showa University, Tokyo 142-8555, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, School of Medicine, Showa University, Tokyo 142-8555, Japan
- Pharmacological Research Center, Showa University, Tokyo 142-8555, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa 920-8640, Japan
- Correspondence: (M.T.); (K.O.)
| |
Collapse
|
22
|
Uchino A, Irie Y, Tsukano C, Kawase T, Hirose K, Kageyama Y, Tooyama I, Yanagita RC, Irie K. Synthesis and Characterization of Propeller- and Parallel-Type Full-Length Amyloid β40 Trimer Models. ACS Chem Neurosci 2022; 13:2517-2528. [PMID: 35930616 DOI: 10.1021/acschemneuro.2c00363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Oligomers of the amyloid β (Aβ) protein play a critical role in the pathogenesis of Alzheimer's disease. However, their heterogeneity and lability deter the identification of their tertiary structures and mechanisms of action. Aβ trimers and Aβ dimers may represent the smallest aggregation unit with cytotoxicity. Although propeller-type trimer models of E22P-Aβ40 tethered by an aromatic linker have recently been synthesized, they unexpectedly exhibited little cytotoxicity. To increase the flexibility of trimeric propeller-type models, we designed and synthesized trimer models with an alkyl linker, tert-butyltris-l-alanine (tButA), at position 36 or 38. In addition, we synthesized two parallel-type trimer models tethered at position 38 using alkyl linkers of different lengths, α,α-di-l-norvalyl-l-glycine (di-nV-Gly) and α,α-di-l-homonorleucyl-l-glycine (di-hnL-Gly), based on the previously reported toxic dimer model. The propeller-type E22P,V36tButA-Aβ40 trimer (4), which was designed to mimic the C-terminal anti-parallel β-sheet structures proposed by the structural analysis of 150 kDa oligomers of Aβ42, and the parallel-type E22P,G38di-nV-Gly-Aβ40 trimer (6) showed significant cytotoxicity against SH-SY5Y cells and aggregative ability to form protofibrillar species. In contrast, the E22P,G38tButA-Aβ40 trimer (5) and E22P,G38di-hnL-Gly-Aβ40 trimer (7) exhibited weak cytotoxicity, though they formed quasi-stable oligomers observed by ion mobility-mass spectrometry and native polyacrylamide gel electrophoresis. These results suggest that 4 and 6 could have some phase of the structure of toxic Aβ oligomers with a C-terminal hydrophobic core and that the conformation and/or aggregation process rather than the formation of stable oligomers contribute to the induction of cytotoxicity.
Collapse
Affiliation(s)
- Ayumi Uchino
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Yumi Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Chihiro Tsukano
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | | | | | - Yusuke Kageyama
- Molecular Neuroscience Research Center, Shiga University of Medical Sciences, Shiga 520-2192, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Sciences, Shiga 520-2192, Japan
| | - Ryo C Yanagita
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa 761-0795, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
23
|
Irie Y, Matsushima Y, Kita A, Miki K, Segawa T, Maeda M, Yanagita RC, Irie K. Structural basis of the 24B3 antibody against the toxic conformer of amyloid β with a turn at positions 22 and 23. Biochem Biophys Res Commun 2022; 621:162-167. [PMID: 35839743 DOI: 10.1016/j.bbrc.2022.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/04/2022] [Indexed: 12/31/2022]
Abstract
Amyloid β-protein (Aβ) oligomers are involved in the early stages of Alzheimer's disease (AD) and antibodies against these toxic oligomers could be useful for accurate diagnosis of AD. We identified the toxic conformer of Aβ42 with a turn at positions 22/23, which has a propensity to form toxic oligomers. The antibody 24B3, developed by immunization of a toxic conformer surrogate E22P-Aβ9-35 in mice, was found to be useful for AD diagnosis using human cerebrospinal fluid (CSF). However, it is not known how 24B3 recognizes the toxic conformation of wild-type Aβ in CSF. Here, we report the crystal structure of 24B3 Fab complexed with E22P-Aβ11-34, whose residues 16-26 were observed in electron densities, suggesting that the residues comprising the toxic turn at positions 22/23 were recognized by 24B3. Since 24B3 bound only to Aβ42 aggregates, several conformationally restricted analogs of Aβ42 with an intramolecular disulfide bond to mimic the conformation of toxic Aβ42 aggregates were screened by enzyme immunoassay. As a result, only F19C,A30homoC-SS-Aβ42 (1) bound significantly to 24B3. These data provide a structural basis for its low affinity to the Aβ42 monomer and selectivity for its aggregate form.
Collapse
Affiliation(s)
- Yumi Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Sakyo-Ku, Kyoto, 606-8502, Japan
| | - Yuka Matsushima
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Sakyo-Ku, Kyoto, 606-8502, Japan
| | - Akiko Kita
- Institute for Integrated Radiation and Nuclear Science, Kyoto University, Sennan, Osaka, 590-0494, Japan
| | - Kunio Miki
- Department of Chemistry, Graduate School of Science, Kyoto University, Kyoto, 606-8502, Japan
| | - Tatsuya Segawa
- Immuno-Biological Laboratories Co, Ltd, Gunma, 375-0005, Japan
| | - Masahiro Maeda
- Immuno-Biological Laboratories Co, Ltd, Gunma, 375-0005, Japan
| | - Ryo C Yanagita
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, 761-0795, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Sakyo-Ku, Kyoto, 606-8502, Japan.
| |
Collapse
|
24
|
Abstract
A study by Kass et al.1 aims to understand the size distribution of amyloid β oligomers in postmortem brain tissues from individuals with Alzheimer's disease (AD) and AD mouse models. Moreover, they show a dose-dependent oligomer elimination by the RD2 compound.
Collapse
|
25
|
Ring J, Tadic J, Ristic S, Poglitsch M, Bergmann M, Radic N, Mossmann D, Liang Y, Maglione M, Jerkovic A, Hajiraissi R, Hanke M, Küttner V, Wolinski H, Zimmermann A, Domuz Trifunović L, Mikolasch L, Moretti DN, Broeskamp F, Westermayer J, Abraham C, Schauer S, Dammbrueck C, Hofer SJ, Abdellatif M, Grundmeier G, Kroemer G, Braun RJ, Hansen N, Sommer C, Ninkovic M, Seba S, Rockenfeller P, Vögtle F, Dengjel J, Meisinger C, Keller A, Sigrist SJ, Eisenberg T, Madeo F. The HSP40 chaperone Ydj1 drives amyloid beta 42 toxicity. EMBO Mol Med 2022; 14:e13952. [PMID: 35373908 PMCID: PMC9081910 DOI: 10.15252/emmm.202113952] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 01/22/2023] Open
Abstract
Amyloid beta 42 (Abeta42) is the principal trigger of neurodegeneration during Alzheimer's disease (AD). However, the etiology of its noxious cellular effects remains elusive. In a combinatory genetic and proteomic approach using a yeast model to study aspects of intracellular Abeta42 toxicity, we here identify the HSP40 family member Ydj1, the yeast orthologue of human DnaJA1, as a crucial factor in Abeta42-mediated cell death. We demonstrate that Ydj1/DnaJA1 physically interacts with Abeta42 (in yeast and mouse), stabilizes Abeta42 oligomers, and mediates their translocation to mitochondria. Consequently, deletion of YDJ1 strongly reduces co-purification of Abeta42 with mitochondria and prevents Abeta42-induced mitochondria-dependent cell death. Consistently, purified DnaJ chaperone delays Abeta42 fibrillization in vitro, and heterologous expression of human DnaJA1 induces formation of Abeta42 oligomers and their deleterious translocation to mitochondria in vivo. Finally, downregulation of the Ydj1 fly homologue, Droj2, improves stress resistance, mitochondrial morphology, and memory performance in a Drosophila melanogaster AD model. These data reveal an unexpected and detrimental role for specific HSP40s in promoting hallmarks of Abeta42 toxicity.
Collapse
|
26
|
Hamaguchi T, Ono K, Yamada M. Transmission of Cerebral β-Amyloidosis Among Individuals. Neurochem Res 2022; 47:2469-2477. [PMID: 35277809 DOI: 10.1007/s11064-022-03566-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 11/26/2022]
Abstract
Deposition of amyloid β protein (Aβ) in the brain (cerebral β-amyloidosis) is a hallmark of Alzheimer's disease (AD). So far, there have been increasing number of experimental studies using AD mouse model that cerebral β-amyloidosis could be transmitted among individuals as prion-like mechanism. Furthermore, several pathological studies using autopsied patients with iatrogenic Creutzfeldt-Jakob disease (CJD) showed that cerebral β-amyloidosis in addition to the CJD pathology could be transmitted among humans via medical procedures, such as human growth hormone derived from cadaver injection and cadaveric dura mater graft. In addition, although cerebral amyloid angiopathy (CAA), which is Aβ deposition in the cerebral vessels, related cerebral hemorrhage rarely develops in young people, several patients with CAA-related cerebral hemorrhage under the age of 55 with histories of neurosurgeries with and without dura mater graft in early childhood have been reported. These patients might show that Aβ pathology is often recognized as Aβ-CAA rather than parenchymal Aβ deposition in the transmission of cerebral β-amyloidosis in humans, and we proposed an emerging concept, "acquired CAA". Considering that there have been several patients with acquired CAA with an incubation period from neurosurgery and the onset of CAA related cerebral hemorrhage of longer than 40 years, the number of cases is likely to increase in the future, and detailed epidemiological investigation is required. It is necessary to continue to elucidate the pathomechanisms of acquired CAA and urgently establish a method for preventing the transmission of cerebral β-amyloidosis among individuals.
Collapse
Affiliation(s)
- Tsuyoshi Hamaguchi
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| | - Kenjiro Ono
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8640, Japan.
- Division of Neurology, Department of Internal Medicine, Kudanzaka Hospital, Tokyo, Japan.
| |
Collapse
|
27
|
Matsushima Y, Irie Y, Kageyama Y, Bellier JP, Tooyama I, Maki T, Kume T, Yanagita RC, Irie K. Structure optimization of the toxic conformation model of amyloid β42 by intramolecular disulfide bond formation. Chembiochem 2022; 23:e202200029. [PMID: 35165998 DOI: 10.1002/cbic.202200029] [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] [Received: 01/18/2022] [Revised: 02/14/2022] [Indexed: 11/07/2022]
Abstract
Amyloid β (Aβ) oligomers play a critical role in the pathology of Alzheimer's disease. Recently, we reported that a conformation-restricted Aβ42 with an intramolecular disulfide bond through cysteine residues at positions 17/28 formed stable oligomers with potent cytotoxicity. To further optimize this compound as a toxic conformer model, we synthesized three analogs with a combination of cysteine and homocysteine at positions 17/28. The analogs with Cys-Cys, Cys-homoCys, or homoCys-Cys, but not the homoCys-homoCys analog, exhibited potent cytotoxicity against SH-SY5Y and THP-1 cells even at 10 nM. In contrast, the cytotoxicity of conformation-restricted analogs at positions 16/29 or 18/27 was significantly weaker than that of wild-type Aβ42. Furthermore, a thioflavin-T assay, non-denaturing gel electrophoresis, and morphological study suggested that the majority of these conformation-restricted analogs existed in an oligomeric state in cell culture medium, indicating that the toxic conformation of Aβ42, rather than the oligomeric state, is essential to induce cytotoxicity.
Collapse
Affiliation(s)
- Yuka Matsushima
- Kyoto University Graduate School of Agriculture Faculty of Agriculture: Kyoto Daigaku Nogaku Kenkyuka Nogakubu, Division of Food Science and Biotechnology, JAPAN
| | - Yumi Irie
- Kyoto University Graduate School of Agriculture Faculty of Agriculture: Kyoto Daigaku Nogaku Kenkyuka Nogakubu, Division of Food Science and Biotechnology, JAPAN
| | - Yusuke Kageyama
- Shiga University of Medical Science: Shiga Ika Daigaku, Molecular Neuroscience Research Center, JAPAN
| | - Jean-Pierre Bellier
- Shiga University of Medical Science: Shiga Ika Daigaku, Molecular Neuroscience Research Center, JAPAN
| | - Ikuo Tooyama
- Shiga University of Medical Science: Shiga Ika Daigaku, Molecular Neuroscience Research Center, JAPAN
| | - Takahito Maki
- University of Toyama: Toyama Daigaku, Department of Applied Pharmacology, JAPAN
| | - Toshiaki Kume
- University of Toyama: Toyama Daigaku, Department of Applied Pharmacology, JAPAN
| | - Ryo C Yanagita
- Kagawa University Faculty of Agriculture Graduate School of Agriculture: Kagawa Daigaku Nogakubu Daigakuin Nogaku Kenkyuka, Department of Applied Biological Sciences, JAPAN
| | - Kazuhiro Irie
- Kyoto University Graduate School of Agriculture Faculty of Agriculture: Kyoto Daigaku Nogaku Kenkyuka Nogakubu, Division of Food Science and Biotechnology, Kitashirakawa Oiwake-cho, Sakyo-ku, 606-8502, Kyoto, JAPAN
| |
Collapse
|
28
|
Hanaki M, Murakami K, Gunji H, Irie K. Activity-differential search for amyloid-β aggregation inhibitors using LC-MS combined with principal component analysis. Bioorg Med Chem Lett 2022; 61:128613. [PMID: 35176471 DOI: 10.1016/j.bmcl.2022.128613] [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: 12/22/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 11/02/2022]
Abstract
Aggregation of amyloid β42 (Aβ42) is one of the hallmarks of Alzheimer's disease (AD). Inhibition of Aβ42 aggregation is thus a promising approach for AD therapy. Kampo medicine has been widely used to combat dementias such as AD. Crude drug known as Shoyaku is an ingredient of Kampo that could have potential as a natural source of novel drugs. However, given that a mixture of compounds, rather than singular compounds, could contribute to the biological functions of crude drug, there are very limited studies on the structure and mechanism of each constituent in crude drug which may have anti-Aβ42 aggregation properties. Herein we provide an efficient method, using LC-MS combined with principal component analysis (PCA), to search for activity-dependent compounds that inhibit Aβ42 aggregation from 46 crude drug extracts originating from 18 plants. Only 5 extracts (Kakou, Kayou, Gusetsu, Rensu, and Renbou) from lotus demonstrated differentially inhibitory activities depending on the part of the plant from which they are derived (e.g. petiole, leaf, root node, stamen, and receptacle, respectively). To compare the anti-aggregative properties of compounds of active crude drug with those of inactive crude drug, these extracts were subjected to LC-MS measurement, followed by PCA. From 12 candidate compounds identified from the analysis, glucuronized and glucosidized quercetin, as well as 6 flavonoids (datiscetin, kaempferol, morin, robinetin, quercetin, and myricitrin), including catechol or flatness moiety suppressed Aβ42 aggregation, whereas curcumol, a sesquiterpene, did not. In conclusion, this study offers a new activity-differential methodology to identify bioactive natural products in crude drugs that inhibit Aβ42 aggregation and that could be applied to future AD therapies.
Collapse
Affiliation(s)
- Mizuho Hanaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Hiroki Gunji
- Alps-Pharmaceutical Industry Co., Ltd., Gifu 509-4241, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
29
|
Beneficial Effects of Spirulina Consumption on Brain Health. Nutrients 2022; 14:nu14030676. [PMID: 35277035 PMCID: PMC8839264 DOI: 10.3390/nu14030676] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 12/10/2022] Open
Abstract
Spirulina is a microscopic, filamentous cyanobacterium that grows in alkaline water bodies. It is extensively utilized as a nutraceutical food supplement all over the world due to its high levels of functional compounds, such as phycocyanins, phenols and polysaccharides, with anti-inflammatory, antioxidant, immunomodulating properties both in vivo and in vitro. Several scientific publications have suggested its positive effects in various pathologies such as cardiovascular diseases, hypercholesterolemia, hyperglycemia, obesity, hypertension, tumors and inflammatory diseases. Lately, different studies have demonstrated the neuroprotective role of Spirulina on the development of the neural system, senility and a number of pathological conditions, including neurological and neurodegenerative diseases. This review focuses on the role of Spirulina in the brain, highlighting how it exerts its beneficial anti-inflammatory and antioxidant effects, acting on glial cell activation, and in the prevention and/or progression of neurodegenerative diseases, in particular Parkinson’s disease, Alzheimer’s disease and Multiple Sclerosis; due to these properties, Spirulina could be considered a potential natural drug.
Collapse
|
30
|
Administration of mucuna beans (Mucuna pruriences (L.) DC. var. utilis) improves cognition and neuropathology of 3 × Tg-AD mice. Sci Rep 2022; 12:996. [PMID: 35046433 PMCID: PMC8770455 DOI: 10.1038/s41598-022-04777-z] [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: 09/08/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the accumulation of extracellular amyloid-beta peptides (Aβ) resulting in senile plaques and intracellular hyperphosphorylated tau protein resulting in neurofibrillary tangles (NFTs). Mucuna beans (Mucuna pruriences (L.) DC. var. utilis) are unique plants containing 3–9% L-3,4-dihydroxyphenylalanine (L-DOPA). Here we investigated the effect of the administration of Mucuna beans on AD prevention by feeding triple-transgenic mice (3 × Tg-AD mice) with a diet containing Mucuna beans for 13 months. The levels of Aβ oligomers and detergent-insoluble phosphorylated tau decreased in the brain of mice fed with Mucuna beans (Mucuna group) compared to those of the Control group. Aβ accumulation and phosphorylated tau accumulation in the brain in the Mucuna group were also reduced. In addition, administration of Mucuna beans improved cognitive function. These results suggest that administration of Mucuna beans may have a preventive effect on AD development in 3 × Tg-AD mice.
Collapse
|
31
|
Singh K, Kaur A, Goyal D, Goyal B. Mechanistic insights into the mitigation of Aβ aggregation and protofibril destabilization by a D–enantiomeric decapeptide rk10. Phys Chem Chem Phys 2022; 24:21975-21994. [DOI: 10.1039/d2cp02601e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
According to clinical studies, the development of Alzheimer’s disease (AD) is linked to the abnormal aggregation of amyloid-β (Aβ) peptides into toxic soluble oligomers, protofibrils as well as mature fibrils....
Collapse
|
32
|
Kinoshita K, Nakabayashi S, Ishikura A, Fujihara K, Hirabayashi S, Koike S, Sasaki H, Ogasawara Y, Koyama K. Inhibition of Amyloid-β Aggregation by p-Terphenyls from the Mushroom Polyozellus multiplex and Their Neuroprotective Effects. HETEROCYCLES 2022. [DOI: 10.3987/com-22-14711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
George DE, Tepe JJ. Advances in Proteasome Enhancement by Small Molecules. Biomolecules 2021; 11:1789. [PMID: 34944433 PMCID: PMC8699248 DOI: 10.3390/biom11121789] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 01/11/2023] Open
Abstract
The proteasome system is a large and complex molecular machinery responsible for the degradation of misfolded, damaged, and redundant cellular proteins. When proteasome function is impaired, unwanted proteins accumulate, which can lead to several diseases including age-related and neurodegenerative diseases. Enhancing proteasome-mediated substrate degradation with small molecules may therefore be a valuable strategy for the treatment of various neurodegenerative diseases such as Parkinson's, Alzheimer's, and Huntington's diseases. In this review, we discuss the structure of proteasome and how proteasome's proteolytic activity is associated with aging and various neurodegenerative diseases. We also summarize various classes of compounds that are capable of enhancing, directly or indirectly, proteasome-mediated protein degradation.
Collapse
Affiliation(s)
| | - Jetze J. Tepe
- Department of Chemistry and Pharmacology & Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
34
|
Aggregation and structure of amyloid β-protein. Neurochem Int 2021; 151:105208. [PMID: 34655726 DOI: 10.1016/j.neuint.2021.105208] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder and is characterized by major pathological hallmarks in the brain, including plaques composed of amyloid β-protein (Aβ) and neurofibrillary tangles of tau protein. Genetic studies, biochemical data, and animal models have suggested that Aβ is a critical species in the pathogenesis of AD. Aβ molecules aggregate to form oligomers, protofibrils (PFs), and mature fibrils. Because of their instability and structural heterogeneity, the misfolding and aggregation of Aβ is a highly complex process, leading to a variety of aggregates with different structures and morphologies. However, the elucidation of Aβ molecules is essential because they are believed to play an important role in AD pathogenesis. Recent combination studies using nuclear magnetic resonance (NMR) and cryo-electron microscopy (cryo-EM) have primarily revealed more detailed information about their aggregation process, including fibril extension and secondary nucleation, and the structural polymorphism of the fibrils under a variety of some conditions, including the actual brain. This review attempts to summarize the existing information on the major properties of the structure and aggregation of Aβ.
Collapse
|
35
|
Murakami K, Horii S, Hanaki M, Irie K. Searching for Natural Products That Delay Nucleation Phase and Promote Elongation Phase of Amyloid β42 toward Alzheimer's Disease Therapeutics. ACS Chem Neurosci 2021; 12:3467-3476. [PMID: 34463471 DOI: 10.1021/acschemneuro.1c00489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Aggregation of amyloid β42 (Aβ42) is one of the hallmarks of Alzheimer's disease (AD). The mechanism of Aβ42 aggregation mainly consists of two phases, nucleation and elongation (including plateau region as a saturation phase). During the nucleation phase, the monomer gradually forms toxic oligomers. During the elongation phase, each nucleus acts as a template and associates with monomers to initiate less toxic fibrillization. We previously proposed a method of classifying compounds into nine groups based on their ability to modulate the nucleation and/or elongation phases. An orcein derivative (O4), which is a phenoxazine dye isolated from the lichen Roccella tinctoria and containing a 2,5-cyclohexadienone moiety, was reported to convert oligomers into relatively inert fibrils, resulting in the reduction of the neurotoxicity of Aβ42. Focusing on O4 in the pursuit of anti-AD drugs, we herein screened 480 natural products including NPDepo (RIKEN) for the compounds that delayed the nucleation phase and promoted the elongation phase. The signal intensities for Aβ42 treated with each of the 15 compounds that met these criteria were lowered in dot blotting using antioligomer antibody, and the fibril formation of Aβ42 in the presence of these compounds was observed in transmission electron microscopy. Among the 15 compounds, 12 compounds (80%) reduced the toxicity of Aβ42 against mouse neuroblastoma Neuro-2a cells. Some of these anticytotoxic compounds contain 2-pyrone and 4-pyrone that interacted with Aβ42, maybe by shifting the equilibrium of Aβ from toxic oligomer into inert fibrils.
Collapse
Affiliation(s)
- Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Shiori Horii
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Mizuho Hanaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
36
|
Kimura AM, Tsuji M, Yasumoto T, Mori Y, Oguchi T, Tsuji Y, Umino M, Umino A, Nishikawa T, Nakamura S, Inoue T, Kiuchi Y, Yamada M, Teplow DB, Ono K. Myricetin prevents high molecular weight Aβ 1-42 oligomer-induced neurotoxicity through antioxidant effects in cell membranes and mitochondria. Free Radic Biol Med 2021; 171:232-244. [PMID: 34015458 DOI: 10.1016/j.freeradbiomed.2021.05.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/27/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022]
Abstract
Excessive accumulation of amyloid β-protein (Aβ) is one of the primary mechanisms that leads to neuronal death with phosphorylated tau in the pathogenesis of Alzheimer's disease (AD). Protofibrils, one of the high-molecular-weight Aβ oligomers (HMW-Aβo), are implicated to be important targets of disease modifying therapy of AD. We previously reported that phenolic compounds such as myricetin inhibit Aβ1-40, Aβ1-42, and α-synuclein aggregations, including their oligomerizations, which may exert protective effects against AD and Parkinson's disease. The purpose of this study was to clarify the detailed mechanism of the protective effect of myricetin against the neurotoxicity of HMW-Aβo in SH-SY5Y cells. To assess the effect of myricetin on HMW-Aβo-induced oxidative stress, we systematically examined the level of membrane oxidative damage by measuring cell membrane lipid peroxidation, membrane fluidity, and cell membrane potential, and the mitochondrial oxidative damage was evaluated by mitochondrial permeability transition (MPT), mitochondrial reactive oxygen species (ROS), and manganese-superoxide dismutase (Mn-SOD), and adenosine triphosphate (ATP) assay in SH-SY5Y cells. Myricetin has been found to increased cell viability by suppression of HMW-Aβo-induced membrane disruption in SH-SY5Y cells, as shown in reducing membrane phospholipid peroxidation and increasing membrane fluidity and membrane resistance. Myricetin has also been found to suppress HMW-Aβo-induced mitochondria dysfunction, as demonstrated in decreasing MPT, Mn-SOD, and ATP generation, raising mitochondrial membrane potential, and increasing mitochondrial-ROS generation. These results suggest that myricetin preventing HMW-Aβo-induced neurotoxicity through multiple antioxidant functions may be developed as a disease-modifying agent against AD.
Collapse
Affiliation(s)
- Atsushi Michael Kimura
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan; Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan.
| | - Taro Yasumoto
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Yukiko Mori
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | - Tatsunori Oguchi
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Yuya Tsuji
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Masakazu Umino
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Asami Umino
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Toru Nishikawa
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Tomio Inoue
- Department of Oral Physiology, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Division of Medical Pharmacology, School of Medicine, Showa University, Tokyo, 142-8555, Japan; Pharmacological Research Center, Showa University, Tokyo, 142-8555, Japan
| | - Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, 920-8640, Japan
| | - David B Teplow
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Room 445, Los Angeles, CA, 90095, USA
| | - Kenjiro Ono
- Department of Internal Medicine, Division of Neurology, School of Medicine, Showa University, Tokyo, 142-8666, Japan.
| |
Collapse
|
37
|
Yu TW, Lane HY, Lin CH. Novel Therapeutic Approaches for Alzheimer's Disease: An Updated Review. Int J Mol Sci 2021; 22:ijms22158208. [PMID: 34360973 PMCID: PMC8348485 DOI: 10.3390/ijms22158208] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease and accounts for most cases of dementia. The prevalence of AD has increased in the current rapidly aging society and contributes to a heavy burden on families and society. Despite the profound impact of AD, current treatments are unable to achieve satisfactory therapeutic effects or stop the progression of the disease. Finding novel treatments for AD has become urgent. In this paper, we reviewed novel therapeutic approaches in five categories: anti-amyloid therapy, anti-tau therapy, anti-neuroinflammatory therapy, neuroprotective agents including N-methyl-D-aspartate (NMDA) receptor modulators, and brain stimulation. The trend of therapeutic development is shifting from a single pathological target to a more complex mechanism, such as the neuroinflammatory and neurodegenerative processes. While drug repositioning may accelerate pharmacological development, non-pharmacological interventions, especially repetitive transcranial magnetic stimulation (rTMS) and transcranial direct current stimulation (tDCS), also have the potential for clinical application. In the future, it is possible for physicians to choose appropriate interventions individually on the basis of precision medicine.
Collapse
Affiliation(s)
- Tien-Wei Yu
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Hsien-Yuan Lane
- Department of Psychiatry and Brain Disease Research Center, China Medical University Hospital, Taichung 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung 41354, Taiwan
- Correspondence: (H.-Y.L.); (C.-H.L.); Tel.: +886-921-067-260 (H.-Y.L.); +886-7-7317123 (ext. 8753) (C.-H.L.); Fax: +886-4-2236-1042 (H.-Y.L.); +886-7-7326817 (C.-H.L.)
| | - Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: (H.-Y.L.); (C.-H.L.); Tel.: +886-921-067-260 (H.-Y.L.); +886-7-7317123 (ext. 8753) (C.-H.L.); Fax: +886-4-2236-1042 (H.-Y.L.); +886-7-7326817 (C.-H.L.)
| |
Collapse
|
38
|
Wang D, Chen F, Han Z, Yin Z, Ge X, Lei P. Relationship Between Amyloid-β Deposition and Blood-Brain Barrier Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2021; 15:695479. [PMID: 34349624 PMCID: PMC8326917 DOI: 10.3389/fncel.2021.695479] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloid-β (Aβ) is the predominant pathologic protein in Alzheimer's disease (AD). The production and deposition of Aβ are important factors affecting AD progression and prognosis. The deposition of neurotoxic Aβ contributes to damage of the blood-brain barrier. However, the BBB is also crucial in maintaining the normal metabolism of Aβ, and dysfunction of the BBB aggravates Aβ deposition. This review characterizes Aβ deposition and BBB damage in AD, summarizes their interactions, and details their respective mechanisms.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | | | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Xintong Ge
- Tianjin Neurological Institute, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
39
|
Wang K, Na L, Duan M. The Pathogenesis Mechanism, Structure Properties, Potential Drugs and Therapeutic Nanoparticles against the Small Oligomers of Amyloid-β. Curr Top Med Chem 2021; 21:151-167. [PMID: 32938351 DOI: 10.2174/1568026620666200916123000] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/02/2020] [Accepted: 08/13/2020] [Indexed: 12/27/2022]
Abstract
Alzheimer's Disease (AD) is a devastating neurodegenerative disease that affects millions of people in the world. The abnormal aggregation of amyloid β protein (Aβ) is regarded as the key event in AD onset. Meanwhile, the Aβ oligomers are believed to be the most toxic species of Aβ. Recent studies show that the Aβ dimers, which are the smallest form of Aβ oligomers, also have the neurotoxicity in the absence of other oligomers in physiological conditions. In this review, we focus on the pathogenesis, structure and potential therapeutic molecules against small Aβ oligomers, as well as the nanoparticles (NPs) in the treatment of AD. In this review, we firstly focus on the pathogenic mechanism of Aβ oligomers, especially the Aβ dimers. The toxicity of Aβ dimer or oligomers, which attributes to the interactions with various receptors and the disruption of membrane or intracellular environments, were introduced. Then the structure properties of Aβ dimers and oligomers are summarized. Although some structural information such as the secondary structure content is characterized by experimental technologies, detailed structures are still absent. Following that, the small molecules targeting Aβ dimers or oligomers are collected; nevertheless, all of these ligands have failed to come into the market due to the rising controversy of the Aβ-related "amyloid cascade hypothesis". At last, the recent progress about the nanoparticles as the potential drugs or the drug delivery for the Aβ oligomers are present.
Collapse
Affiliation(s)
- Ke Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Liu Na
- School of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mojie Duan
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
40
|
Akbor MM, Kurosawa N, Tanaka M, Isobe M. Polymorphic SERPINA3-R124C reduces pathogenesis of its wild type by shortening the lifetime of oligomeric Aβ. Biosci Biotechnol Biochem 2021; 85:1861-1868. [PMID: 34077500 DOI: 10.1093/bbb/zbab101] [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] [Received: 04/15/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022]
Abstract
Amyloid beta (Aβ) 42 peptide accumulated in Alzheimer disease (AD) patients' brain, often colocalized with serine protease inhibitor family A member 3 (SERPINA3). Being a chaperon, SERPINA3 accelerated Aβ42 fibrillization. While analyzing chaperon activity of human SERPINA3 polymorphisms, we found SERPINA3-R124C played a role in protecting cells from Aβ42 cytotoxicity. SH-SY5Y cells exposed to Aβ42 preincubated with wild-type SERPINA3 (SERPINA3-WT) resulted in extended toxicity leading cell death whereas Aβ42 with SERPINA3-R124C resulted in less cytotoxicity. Transmission electron microscope and thioflavin T assay revealed that SERPINA3-R124C shortened lifetime of small soluble oligomer and maintained β-sheet rich protofibril-like aggregates for longer time compared to that of with SERPINA3-WT. Western blot assay confirmed that SERPINA3-R124C converted Aβ42 mostly into high molecular aggregates. Here, we demonstrate first time that polymorphic SERPINA3 acts as a benign chaperon by modulating the transition states of Aβ42, which may contribute to the reduction of AD risk.
Collapse
Affiliation(s)
- Maruf Mohammad Akbor
- Laboratory of Molecular and Cellular Biology, Department of Life Sciences and Bioengineering, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Nobuyuki Kurosawa
- Laboratory of Molecular and Cellular Biology, Department of Life Sciences and Bioengineering, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Masashi Tanaka
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaharu Isobe
- Laboratory of Molecular and Cellular Biology, Department of Life Sciences and Bioengineering, Faculty of Engineering, University of Toyama, Toyama, Japan
| |
Collapse
|
41
|
Mori Y, Tsuji M, Oguchi T, Kasuga K, Kimura A, Futamura A, Sugimoto A, Kasai H, Kuroda T, Yano S, Hieda S, Kiuchi Y, Ikeuchi T, Ono K. Serum BDNF as a Potential Biomarker of Alzheimer's Disease: Verification Through Assessment of Serum, Cerebrospinal Fluid, and Medial Temporal Lobe Atrophy. Front Neurol 2021; 12:653267. [PMID: 33967943 PMCID: PMC8102980 DOI: 10.3389/fneur.2021.653267] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/16/2021] [Indexed: 12/16/2022] Open
Abstract
There is an urgent need to establish blood biomarkers for Alzheimer's disease (AD). Although it has been speculated that brain-derived neurotrophic factor (BDNF) is associated with AD, whether it can be used as a blood biomarker has yet to be determined. We used serum, cerebrospinal fluid (CSF), and medial temporal lobe atrophy from patients with AD to evaluate the association of BDNF with AD and assess its severity. For the blood analysis, 66 participants [21 normal controls (NCs) with normal cognitive function, 22 patients with mild cognitive impairment (MCI) due to AD, and 23 patients with AD] were included. For the CSF analysis, 30 participants were included. Magnetic resonance imaging, including a voxel-based specific regional analysis system for AD, and a Mini Mental State Examination were performed. Serum levels of BDNF and CSF levels of amyloid-β42, total tau, and phosphorylated tau were measured using ELISA. Serum BDNF levels were significantly lower in the MCI due to AD group than in the NC group (p = 0.037). Although there was no significant difference in the AD group, there was a downward trend compared to the NC group. Serum BDNF levels were positively correlated with CSF Aβ42 levels (r = 0.49, p = 0.005). There was a significant correlation between serum BDNF levels and medial temporal lobe atrophy. Decreased serum BDNF can potentially be used as a biomarker for early AD detection. Early detection of AD with a less invasive blood test is very beneficial, as it allows for intervention before dementia progresses.
Collapse
Affiliation(s)
- Yukiko Mori
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Tatsunori Oguchi
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Atsushi Kimura
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Akinori Futamura
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Azusa Sugimoto
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hideyo Kasai
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takeshi Kuroda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Satoshi Yano
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Sotaro Hieda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, School of Medicine, Showa University, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenjiro Ono
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Boutté AM, Thangavelu B, Nemes J, LaValle CR, Egnoto M, Carr W, Kamimori GH. Neurotrauma Biomarker Levels and Adverse Symptoms Among Military and Law Enforcement Personnel Exposed to Occupational Overpressure Without Diagnosed Traumatic Brain Injury. JAMA Netw Open 2021; 4:e216445. [PMID: 33861330 PMCID: PMC8052592 DOI: 10.1001/jamanetworkopen.2021.6445] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IMPORTANCE There is a scientific and operational need to define objective measures of exposure to low-level overpressure (LLOP) and concussion-like symptoms among persons with specialized occupations. OBJECTIVE To evaluate serum levels of neurotrauma biomarkers and their association with concussion-like symptoms reported by LLOP-exposed military and law enforcement personnel who are outwardly healthy and cleared to perform duties. DESIGN, SETTING, AND PARTICIPANTS This retrospective cohort study, conducted from January 23, 2017, to October 21, 2019, used serum samples and survey data collected from healthy, male, active-duty military and law enforcement personnel assigned to operational training at 4 US Department of Defense and civilian law enforcement training sites. Personnel aged 18 years or older with prior LLOP exposure but no diagnosed traumatic brain injury or with acute blast exposure during sampling participated in the study. Serum samples from 30 control individuals were obtained from a commercial vendor. MAIN OUTCOMES AND MEASURES Serum levels of glial fibrillary acidic protein, ubiquitin carboxyl hydrolase (UCH)-L1, neurofilament light chain, tau, amyloid β (Aβ)-40, and Aβ-42 from a random sample (30 participants) of the LLOP-exposed cohort were compared with those of 30 age-matched controls. Associations between biomarker levels and self-reported symptoms or operational demographics in the remainder of the study cohort (76 participants) were assessed using generalized linear modeling or Spearman correlations with age as a covariate. RESULTS Among the 30 randomly sampled participants (mean [SD] age, 32 [7.75] years), serum levels of UCH-L1 (mean difference, 4.92; 95% CI, 0.71-9.14), tau (mean difference, 0.16; 95% CI, -0.06 to 0.39), Aβ-40 (mean difference, 138.44; 95% CI, 116.32-160.56), and Aβ-42 (mean difference, 4.97; 95% CI, 4.10-5.83) were elevated compared with those in controls. Among the remaining cohort of 76 participants (mean [SD] age, 34 [7.43] years), ear ringing was reported by 44 (58%) and memory or sleep problems were reported by 24 (32%) and 20 (26%), respectively. A total of 26 participants (34%) reported prior concussion. Amyloid β-42 levels were associated with ear ringing (F1,72 = 7.40; P = .008) and memory problems (F1,72 = 9.20; P = .003). CONCLUSIONS AND RELEVANCE The findings suggest that long-term LLOP exposure acquired during occupational training may be associated with serum levels of neurotrauma biomarkers. Assessment of biomarkers and concussion-like symptoms among personnel considered healthy at the time of sampling may be useful for military occupational medicine risk management.
Collapse
Affiliation(s)
- Angela M. Boutté
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Bharani Thangavelu
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Jeffrey Nemes
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Christina R. LaValle
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Mike Egnoto
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Walter Carr
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| | - Gary H. Kamimori
- Blast Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, Maryland
| |
Collapse
|
43
|
Insoluble Vascular Amyloid Deposits Trigger Disruption of the Neurovascular Unit in Alzheimer's Disease Brains. Int J Mol Sci 2021; 22:ijms22073654. [PMID: 33915754 PMCID: PMC8036769 DOI: 10.3390/ijms22073654] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, characterized histopathologically by intra-neuronal tau-related lesions and by the accumulation of amyloid β-peptide (Aβ) in the brain parenchyma and around cerebral blood vessels. According to the vascular hypothesis of AD, an alteration in the neurovascular unit (NVU) could lead to Aβ vascular accumulation and promote neuronal dysfunction, accelerating neurodegeneration and dementia. To date, the effects of insoluble vascular Aβ deposits on the NVU and the blood-brain barrier (BBB) are unknown. In this study, we analyze different Aβ species and their association with the cells that make up the NVU. We evaluated post-mortem AD brain tissue. Multiple immunofluorescence assays were performed against different species of Aβ and the main elements that constitute the NVU. Our results showed that there are insoluble vascular deposits of both full-length and truncated Aβ species. Besides, insoluble aggregates are associated with a decrease in the phenotype of the cellular components that constitute the NVU and with BBB disruption. This approach could help identify new therapeutic targets against key molecules and receptors in the NVU that can prevent the accumulation of vascular fibrillar Aβ in AD.
Collapse
|
44
|
Uchino A, Tsukano C, Imamoto T, Irie K. Synthesis of Alkyl Bridged‐Tris‐α‐Amino Acids as C
3
‐Symmetric and Linear Linkers. European J Org Chem 2021. [DOI: 10.1002/ejoc.202001592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Ayumi Uchino
- Division of Food Science and Biotechnology Graduate School of Agriculture Kyoto University Kitashirakawa, Sakyo-ku Kyoto 606-8502 Japan
| | - Chihiro Tsukano
- Division of Food Science and Biotechnology Graduate School of Agriculture Kyoto University Kitashirakawa, Sakyo-ku Kyoto 606-8502 Japan
| | - Tsuneo Imamoto
- Department of Chemistry Graduate School of Science Chiba University Yayoi-cho, Inage-ku, Chiba 263-8522 Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology Graduate School of Agriculture Kyoto University Kitashirakawa, Sakyo-ku Kyoto 606-8502 Japan
| |
Collapse
|
45
|
Akbor MM, Kurosawa N, Nakayama H, Nakatani A, Tomobe K, Chiba Y, Ueno M, Tanaka M, Nomura Y, Isobe M. Polymorphic SERPINA3 prolongs oligomeric state of amyloid beta. PLoS One 2021; 16:e0248027. [PMID: 33662018 PMCID: PMC7932536 DOI: 10.1371/journal.pone.0248027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Molecular chaperon SERPINA3 colocalizes with accumulated amyloid peptide in Alzheimer’s disease (AD) patient’s brain. From the QTL analysis, we narrowed down Serpina3 with two SNPs in senescence-accelerated mouse prone (SAMP) 8 strain. Our study showed SAMP8 type Serpina3 prolonged retention of oligomeric Aβ 42 for longer duration (72 hr) while observing under transmission electron microscope (TEM). From Western blot results, we confirmed presence of Aβ 42 oligomeric forms (trimers, tetramers) were maintained for longer duration only in the presences of SAMP8 type Serpina3. Using SH-SY5Y neuroblastoma cell line, we observed until 36 hr preincubated Aβ 42 with SAMP8 type Serpina3 caused neuronal cell death compared to 12 hr preincubated Aβ 42 with SAMR1 or JF1 type Serpina3 proteins. Similar results were found by extending this study to analyze the effect of polymorphism of SERPINA3 gene of the Japanese SNP database for geriatric research (JG-SNP). We observed that polymorphic SERPINA3 I308T (rs142398813) prolonged toxic oligomeric Aβ 42 forms till 48 hr in comparison to the presence wild type SERPINA3 protein, resulting neuronal cell death. From this study, we first clarified pathogenic regulatory role of polymorphic SERPINA3 in neurodegeneration.
Collapse
Affiliation(s)
- Maruf Mohammad Akbor
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Nobuyuki Kurosawa
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Hiroki Nakayama
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Ayumi Nakatani
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, University of Toyama, Toyama, Japan
| | - Koji Tomobe
- Department of Pathophysiology, Yokohama University of Pharmacy, Yokohama, Japan
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Miki-cho, Kita-gun, Kagawa, Japan
| | - Masashi Tanaka
- Department for Health and Longevity Research, National Institutes of Biomedical Innovation, Health and Nutrition, Shinjuku, Tokyo, Japan
| | - Yasuyuki Nomura
- Department of Pharmacology, School of Medicine, Kurume University, Kurume, Fukuoka, Japan
| | - Masaharu Isobe
- Department of Life Sciences and Bioengineering, Laboratory of Molecular and Cellular Biology, Faculty of Engineering, University of Toyama, Toyama, Japan
- * E-mail:
| |
Collapse
|
46
|
van Oostveen WM, de Lange ECM. Imaging Techniques in Alzheimer's Disease: A Review of Applications in Early Diagnosis and Longitudinal Monitoring. Int J Mol Sci 2021; 22:ijms22042110. [PMID: 33672696 PMCID: PMC7924338 DOI: 10.3390/ijms22042110] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting many individuals worldwide with no effective treatment to date. AD is characterized by the formation of senile plaques and neurofibrillary tangles, followed by neurodegeneration, which leads to cognitive decline and eventually death. INTRODUCTION In AD, pathological changes occur many years before disease onset. Since disease-modifying therapies may be the most beneficial in the early stages of AD, biomarkers for the early diagnosis and longitudinal monitoring of disease progression are essential. Multiple imaging techniques with associated biomarkers are used to identify and monitor AD. AIM In this review, we discuss the contemporary early diagnosis and longitudinal monitoring of AD with imaging techniques regarding their diagnostic utility, benefits and limitations. Additionally, novel techniques, applications and biomarkers for AD research are assessed. FINDINGS Reduced hippocampal volume is a biomarker for neurodegeneration, but atrophy is not an AD-specific measure. Hypometabolism in temporoparietal regions is seen as a biomarker for AD. However, glucose uptake reflects astrocyte function rather than neuronal function. Amyloid-β (Aβ) is the earliest hallmark of AD and can be measured with positron emission tomography (PET), but Aβ accumulation stagnates as disease progresses. Therefore, Aβ may not be a suitable biomarker for monitoring disease progression. The measurement of tau accumulation with PET radiotracers exhibited promising results in both early diagnosis and longitudinal monitoring, but large-scale validation of these radiotracers is required. The implementation of new processing techniques, applications of other imaging techniques and novel biomarkers can contribute to understanding AD and finding a cure. CONCLUSIONS Several biomarkers are proposed for the early diagnosis and longitudinal monitoring of AD with imaging techniques, but all these biomarkers have their limitations regarding specificity, reliability and sensitivity. Future perspectives. Future research should focus on expanding the employment of imaging techniques and identifying novel biomarkers that reflect AD pathology in the earliest stages.
Collapse
Affiliation(s)
- Wieke M. van Oostveen
- Faculty of Science, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands;
| | - Elizabeth C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
47
|
Futamura A, Hieda S, Mori Y, Kasuga K, Sugimoto A, Kasai H, Kuroda T, Yano S, Tsuji M, Ikeuchi T, Irie K, Ono K. Toxic Amyloid-β42 Conformer May Accelerate the Onset of Alzheimer's Disease in the Preclinical Stage. J Alzheimers Dis 2021; 80:639-646. [PMID: 33579852 DOI: 10.3233/jad-201407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Toxic amyloid-β protein (Aβ) conformers play an important role in the progression of Alzheimer's disease (AD). The ratio of toxic conformer to total Aβ42 in cerebrospinal fluid (CSF) was significantly high in AD and mild cognitive impairment (MCI) due to AD using an enzyme-linked immunosorbent assay kit with a 24B3 antibody. OBJECTIVE We compared the toxic Aβ42, conformer at different stages of AD to identify its contribution to AD pathogenesis. METHODS We compared 5 patients with preclinical AD, 11 patients with MCI due to AD, 21 patients with AD, and 5 healthy controls to measure CSF levels of total Aβ42, total tau, tau phosphorylated at threonine 181 (p-tau), and toxic Aβ conformers. All were classified using the Clinical Dementia Rating. Cognitive function was assessed using the Japanese version of the Mini-Mental State Examination (MMSE-J). RESULTS Toxic Aβ conformer level was insignificant between groups, but its ratio to Aβ42 was significantly higher in AD than in preclinical AD (p < 0.05). Toxic Aβ42 conformer correlated positively with p-tau (r = 0.67, p < 0.01) and p-tau correlated negatively with MMSE-J (r = -0.38, p < 0.05). CONCLUSION Toxic Aβ conformer triggers tau accumulation leading to neuronal impairment in AD pathogenesis.
Collapse
Affiliation(s)
- Akinori Futamura
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Sotaro Hieda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Yukiko Mori
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Chuo-ku, Niigata, Japan
| | - Azusa Sugimoto
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Hideyo Kasai
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Takeshi Kuroda
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Satoshi Yano
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Shinagawa-ku, Tokyo, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Chuo-ku, Niigata, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kenjiro Ono
- Division of Neurology, Department of Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
48
|
Abstract
Incorporation of heterocycles into drug molecules can enhance physical properties and biological activity. A variety of heterocyclic groups is available to medicinal chemists, many of which have been reviewed in detail elsewhere. Oxadiazoles are a class of heterocycle containing one oxygen and two nitrogen atoms, available in three isomeric forms. While the 1,2,4- and 1,3,4-oxadiazoles have seen widespread application in medicinal chemistry, 1,2,5-oxadiazoles (furazans) are less common. This Review provides a summary of the application of furazan-containing molecules in medicinal chemistry and drug development programs from analysis of both patent and academic literature. Emphasis is placed on programs that reached clinical or preclinical stages of development. The examples provided herein describe the pharmacology and biological activity of furazan derivatives with comparative data provided where possible for other heterocyclic groups and pharmacophores commonly used in medicinal chemistry.
Collapse
Affiliation(s)
| | | | - Donald F Weaver
- Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada.,Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Mark A Reed
- Treventis Corporation, Toronto, Ontario M5T 0S8, Canada.,Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
49
|
Matsushima Y, Yanagita RC, Irie K. Control of the toxic conformation of amyloid β42 by intramolecular disulfide bond formation. Chem Commun (Camb) 2020; 56:4118-4121. [PMID: 32163091 DOI: 10.1039/d0cc01053g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a method to fix the conformation of Aβ42 to the toxic or non-toxic form by intramolecular disulfide bonds. We found that an Aβ42 analog crosslinked within the molecule at the 17th and 28th amino acid residues exhibited high aggregative ability and potent neurotoxicity comparable to those of E22P-Aβ42. This analog would be useful in the research of Aβ42 oligomers and to develop reliable antibodies for early diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Yuka Matsushima
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| | - Ryo C Yanagita
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa 761-0795, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
50
|
Ding B, Lin C, Liu Q, He Y, Ruganzu JB, Jin H, Peng X, Ji S, Ma Y, Yang W. Tanshinone IIA attenuates neuroinflammation via inhibiting RAGE/NF-κB signaling pathway in vivo and in vitro. J Neuroinflammation 2020; 17:302. [PMID: 33054814 PMCID: PMC7559789 DOI: 10.1186/s12974-020-01981-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Background Glial activation and neuroinflammation play a crucial role in the pathogenesis and development of Alzheimer’s disease (AD). The receptor for advanced glycation end products (RAGE)-mediated signaling pathway is related to amyloid beta (Aβ)-induced neuroinflammation. This study aimed to investigate the neuroprotective effects of tanshinone IIA (tan IIA), a natural product isolated from traditional Chinese herbal Salvia miltiorrhiza Bunge, against Aβ-induced neuroinflammation, cognitive impairment, and neurotoxicity as well as the underlying mechanisms in vivo and in vitro. Methods Open-field test, Y-maze test, and Morris water maze test were conducted to assess the cognitive function in APP/PS1 mice. Immunohistochemistry, immunofluorescence, thioflavin S (Th-S) staining, enzyme-linked immunosorbent assay (ELISA), real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR), and western blotting were performed to explore Aβ deposition, synaptic and neuronal loss, microglial and astrocytic activation, RAGE-dependent signaling, and the production of pro-inflammatory cytokines in APP/PS1 mice and cultured BV2 and U87 cells. Results Tan IIA treatment prevented spatial learning and memory deficits in APP/PS1 mice. Additionally, tan IIA attenuated Aβ accumulation, synapse-associated proteins (Syn and PSD-95) and neuronal loss, as well as peri-plaque microgliosis and astrocytosis in the cortex and hippocampus of APP/PS1 mice. Furthermore, tan IIA significantly suppressed RAGE/nuclear factor-κB (NF-κB) signaling pathway and the production of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) in APP/PS1 mice and cultured BV2 and U87 cells. Conclusions Taken together, the present results indicated that tan IIA improves cognitive decline and neuroinflammation partly via inhibiting RAGE/NF-κB signaling pathway in vivo and in vitro. Thus, tan IIA might be a promising therapeutic drug for halting and preventing AD progression.
Collapse
Affiliation(s)
- Bo Ding
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Chengheng Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Qian Liu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.,Medical Undergraduates of Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yanbing Ma
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|