1
|
Chen J, Ouyang X, Yu C, Xiang J. Functionalized pNIPAM-DNA Hydrogel Colorimetric Platform for Visual Detection of Low-Mass Soluble β-Amyloid Oligomers. Anal Chem 2025. [PMID: 40334134 DOI: 10.1021/acs.analchem.5c00949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Low-mass soluble β-amyloid oligomers (LSAβO) are critical Alzheimer's disease (AD) biomarkers with significant diagnostic and therapeutic potential. However, their application in early screening and detection is limited by the reliance on complex analytical instruments and procedures. To address this, we developed a visual sensing platform for LSAβO detection using a functionalized pNIPAM-DNA hydrogel. Exploiting the temperature-responsive nature of pNIPAM, the hydrogel selectively incorporates and enriches LSAβO from solution via temperature-induced expansion and contraction. LSAβO binding to aptamers on the hydrogel triggers the formation of G-quadruplex DNAzymes, which catalyze the oxidation of 3,3',5,5'-tetramethylbenzidine to generate a visible colorimetric signal. The hydrogel's small pore size further enhances selectivity by excluding larger oligomers during real sample analysis. This sensor exhibits a linear detection range of 0.1-7.5 nM for LSAβO and a detection limit of 50 pM. Combining LSAβO enrichment and size exclusion, this functionalized pNIPAM-DNA hydrogel platform provides a cost-effective, highly sensitive, selective, and high-throughput approach for preliminary LSAβO screening and detection.
Collapse
Affiliation(s)
- Jia Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Xuliang Ouyang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Chenxiao Yu
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Juan Xiang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| |
Collapse
|
2
|
MacAinsh M, Muhammedkutty FNK, Prasad R, Zhou HX. Membrane Association of Intrinsically Disordered Proteins. Annu Rev Biophys 2025; 54:275-302. [PMID: 39952269 PMCID: PMC12055482 DOI: 10.1146/annurev-biophys-070124-092816] [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] [Indexed: 02/17/2025]
Abstract
It is now clear that membrane association of intrinsically disordered proteins or intrinsically disordered regions regulates many cellular processes, such as membrane targeting of Src family kinases and ion channel gating. Residue-specific characterization by nuclear magnetic resonance spectroscopy, molecular dynamics simulations, and other techniques has shown that polybasic motifs and amphipathic helices are the main drivers of membrane association; sequence-based prediction of residue-specific membrane association propensity has become possible. Membrane association facilitates protein-protein interactions and protein aggregation-these effects are due to reduced dimensionality but are similar to those afforded by condensate formation via liquid-liquid phase separation (LLPS). LLPS at the membrane surface provides a powerful means for recruiting and clustering proteins, as well as for membrane remodeling.
Collapse
Affiliation(s)
- Matthew MacAinsh
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
| | | | - Ramesh Prasad
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois, Chicago, Illinois, USA;
- Department of Physics, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
3
|
Zhu J, Kreutzer AG, Liu Z, Li X, Richter SM, Pophristic V, Nowick JS. A β-hairpin peptide derived from Aβ forms different oligomers in the crystal state and in aqueous solution. Org Biomol Chem 2025; 23:3881-3893. [PMID: 40130612 PMCID: PMC12003087 DOI: 10.1039/d5ob00296f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
The supramolecular assembly of amyloid-β into soluble oligomers is critical Alzheimer's disease (AD) progression. Soluble Aβ oligomers have emerged as neurotoxic species involved in AD progression and some Aβ oligomers are thought to be composed of β-hairpins. In this work, we report the X-ray crystallographic and solution-phase assembly of a macrocyclic β-hairpin peptide that mimics a β-hairpin formed by Aβ16-36. In the crystal lattice, the peptide assembles into a symmetric hexamer composed of two identical triangular trimers. In aqueous solution, the peptide assembles to form an asymmetric hexamer. 1H NMR, TOCSY, and 1H,15N HSQC experiments establish that the asymmetric hexamer contains two different species, A and B. 15N-edited NOESY reveals that species A is a cylindrin-like trimer and species B is a triangular trimer that collectively constitute the asymmetric hexamer. Diffusion-ordered NMR spectroscopy (DOSY) suggests that two asymmetric hexamers further assemble to form a dodecamer. NMR-guided molecular mechanics and molecular dynamics studies provide a model for the asymmetric hexamer and suggest how two asymmetric hexamers can form a dodecamer. Solution-phase NMR studies of analogues show that intermolecular hydrogen bonding and the formation of a hydrophobic core help stabilize the asymmetric hexamer. These NMR and crystallographic studies illustrate how an Aβ β-hairpin peptide can assemble to form different well-defined oligomers in the crystal state and in aqueous solution, providing a deeper understanding of the heterogeneity of Aβ oligomers and new structural models of Aβ oligomers composed of Aβ β-hairpins.
Collapse
Affiliation(s)
- Jason Zhu
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA.
| | - Adam G Kreutzer
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA.
| | - Zhiwei Liu
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028-1701, USA
| | - Xingyue Li
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA.
| | - Sabrina M Richter
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA.
| | - Vojislava Pophristic
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, New Jersey 08028-1701, USA
| | - James S Nowick
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, California 92697-2025, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California 92697-2025, USA
| |
Collapse
|
4
|
Vosála O, Šmídová B, Novák J, Svoboda J, Petrásek T, Vojtěchová I, Macháček T. No evidence of Alzheimer's disease pathology in mice infected with Toxocara canis. Parasite 2025; 32:24. [PMID: 40214165 PMCID: PMC11987500 DOI: 10.1051/parasite/2025019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
The potential link between the infections and the development of Alzheimer's disease (AD) has led to speculations about the role of various pathogens in triggering amyloid-β (Aβ) overproduction, possibly leading to AD onset. The globally distributed dog roundworm Toxocara canis was suggested to be a suitable candidate due to neurotropism of the larvae and infection chronicity. This study investigated whether chronic T. canis infection induces AD-like pathology in mice and whether Aβ is toxic to T. canis. BALB/c and APP/PS1 transgenic mice, which overproduce Aβ, were infected with T. canis L3 larvae and monitored for larval burden, Aβ accumulation, and behavioral changes. In vitro tests of recombinant Aβ toxicity against the larvae were also performed. Despite the presence of T. canis larvae in the central nervous system 8 and 16 weeks post-infection, no significant increase in Aβ concentration or AD-related behavioral alterations were observed. Aβ was detected on the surface and within the intestines of T. canis larvae, but in vitro exposure to recombinant Aβ did not affect larval viability or morphology. Our findings suggest that T. canis infection does not trigger AD-like pathology in mice, and Aβ does not act as an antiparasitic agent. This challenges the emerging hypothesis that chronic neurotoxocarosis infections may contribute to AD development.
Collapse
Affiliation(s)
- Ondřej Vosála
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 Hradec Králové 50005 Czechia
| | - Barbora Šmídová
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
| | - Jan Novák
- Institute of Medical Microbiology, First Faculty of Medicine, Charles University and General University Hospital in Prague Studničkova 7 Prague 2 12800 Czechia
| | - Jan Svoboda
- Laboratory of Neurophysiology of Memory, Institute of Physiology, Czech Academy of Sciences Vídeňská 1083 Prague 4 14200 Czechia
| | - Tomáš Petrásek
- Sleep and Chronobiology Research Centre, National Institute of Mental Health Topolová 748 Klecany 25067 Czechia
| | - Iveta Vojtěchová
- Sleep and Chronobiology Research Centre, National Institute of Mental Health Topolová 748 Klecany 25067 Czechia
| | - Tomáš Macháček
- Department of Parasitology, Faculty of Science, Charles University Viničná 7 Prague 2 12844 Czechia
| |
Collapse
|
5
|
Han YL, Yin HH, Li C, Du J, He Y, Guan YX. Discovery of New Pentapeptide Inhibitors Against Amyloid-β Aggregation Using Word2Vec and Molecular Simulation. ACS Chem Neurosci 2025; 16:1055-1065. [PMID: 39999409 DOI: 10.1021/acschemneuro.4c00661] [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: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by the aggregation of amyloid-β (Aβ) peptides into toxic oligomers and fibrils. The efficacy of existing peptide inhibitors based on the central hydrophobic core (CHC) sequence of Aβ42 remains limited due to self-aggregation or poor inhibition. This study aimed to identify novel pentapeptide inhibitors with high similarity and low binding energy to the CHC region LVFFA using a new computational screening workflow based on Word2Vec and molecular simulation. The antimicrobial peptides and human brain protein sequences were used for training the Word2Vec model. After tuning the parameters of the Word2Vec model, 1017 pentapeptides with high similarity to LVFFA were identified. Molecular docking was employed to estimate the affinity of the pentapeptides for the target of Aβ14-42 pentamer, and 103 peptides with favorable docking scores were obtained. Finally, five pentapeptides with a low binding energy and high binding stability via molecular dynamics simulation were experimentally validated using thioflavin T assays. Surprisingly, one pentapeptide, i.e., PALIR, exhibited significant inhibition surpassing the positive control LPFFN. This study demonstrates an effective combinatorial strategy to discover new peptide inhibitors. With PALIR representing a promising lead candidate, further optimization of PALIR could aid in the development of improved therapies to prevent amyloid toxicity in AD.
Collapse
Affiliation(s)
- Yin-Lei Han
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Huan-Huan Yin
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Chen Li
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jiangyue Du
- Department of General Practice, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310020, China
| | - Yi He
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yi-Xin Guan
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Ruzmetov T, Hung TI, Jonnalagedda SP, Chen SH, Fasihianifard P, Guo Z, Bhanu B, Chang CEA. Sampling Conformational Ensembles of Highly Dynamic Proteins via Generative Deep Learning. J Chem Inf Model 2025; 65:2487-2502. [PMID: 39984300 DOI: 10.1021/acs.jcim.4c01838] [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: 02/23/2025]
Abstract
Proteins are inherently dynamic, and their conformational ensembles play a crucial role in biological function. Large-scale motions may govern the protein structure-function relationship, and numerous transient but stable conformations of intrinsically disordered proteins (IDPs) can play a crucial role in biological function. Investigating conformational ensembles to understand regulations and disease-related aggregations of IDPs is challenging, both experimentally and computationally. In this paper, we first introduce a deep learning-based model, termed Internal Coordinate Net (ICoN), which learns the physical principles of conformational changes from molecular dynamics simulation data. Second, we selected data points through interpolation in the learned latent space to rapidly identify novel synthetic conformations with sophisticated and large-scale side chains and backbone arrangements. Third, with the highly dynamic amyloid-β1-42 (Aβ42) monomer, our deep learning model provided a comprehensive sampling of Aβ42's conformational landscape. Analysis of these synthetic conformations revealed conformational clusters that could be used to rationalize experimental findings. Additionally, the method can identify novel conformations with important interactions in atomistic details that are not included in the training data. New synthetic conformations showed distinct side chain rearrangements that are probed by our electron paramagnetic resonance and amino acid substitution studies. This approach is highly transferable and can be used for any available data for training. The work also demonstrated the ability of deep learning to utilize natural atomistic motions in protein conformation sampling.
Collapse
Affiliation(s)
- Talant Ruzmetov
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Ta I Hung
- Department of Chemistry, University of California, Riverside, California 92521, United States
- Department of Bioengineering, University of California, Riverside, California 92521, United States
| | - Saisri Padmaja Jonnalagedda
- Department of Electrical and Computer Engineering, University of California, Riverside, California 92521, United States
| | - Si-Han Chen
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Parisa Fasihianifard
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Zhefeng Guo
- Department of Neurology, Brain Research Institute, University of California, Los Angeles, California 90095, United States
| | - Bir Bhanu
- Department of Bioengineering, University of California, Riverside, California 92521, United States
- Department of Electrical and Computer Engineering, University of California, Riverside, California 92521, United States
| | - Chia-En A Chang
- Department of Chemistry, University of California, Riverside, California 92521, United States
- Department of Bioengineering, University of California, Riverside, California 92521, United States
| |
Collapse
|
7
|
Irizarry B, Davis J, Rajpoot J, Zhu X, Xu F, Smith SO, Van Nostrand WE. Aβ40 Fibril Assembly on Human Cerebral Smooth Muscle Cells Impairs Cell Viability. Biochemistry 2025; 64:1065-1078. [PMID: 39763333 DOI: 10.1021/acs.biochem.4c00478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cerebral vascular deposition of the amyloid-β (Aβ) peptide, a condition known as cerebral amyloid angiopathy (CAA), is associated with intracerebral hemorrhaging and contributes to disease progression in Alzheimer's disease (AD) and vascular cognitive impairment and dementia (VCID). Familial mutations at positions 22 and 23 within the Aβ peptide lead to early onset and severe CAA pathology. Here, we evaluate the effects of fibrillar Aβ peptides on the viability of primary-cultured human cerebral smooth muscle (HCSM) cells, which are the major site of amyloid deposition in cerebral blood vessel walls. Comparisons are made of the familial E22Q (Dutch) mutant of Aβ40 with wild-type Aβ40 and Aβ42. In agreement with previous studies, we find that there is a significant reduction in cell viability when Aβ40-Dutch or Aβ42-WT peptides are added to HCSM cell cultures as monomeric Aβ, whereas Aβ40-WT is relatively nontoxic. The binding of Aβ fibrils derived from sporadic CAA or familial Dutch-type CAA brain tissue to the membrane surface of HCSM cells does not result in a significant loss of cell viability. In contrast, when Aβ40-WT monomers and sporadic CAA fibrils are coincubated in HCSM cell cultures, there is a significant reduction in HCSM cell viability that is accompanied by an increase in cell surface fibril formation. Lastly, intrathecal administration of Aβ40-Dutch fibrillar seeds promotes fibrillar amyloid accumulation in the smooth muscle of meningeal vessels in the rTg-D transgenic rat model of CAA. Together, the present findings suggest that fibrillar Aβ seeds propagate the expansion of new amyloid fibrils on cerebral vascular smooth muscle, leading to membrane disruption and cell death.
Collapse
Affiliation(s)
- Brandon Irizarry
- Center for Structural Biology, Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Judianne Davis
- George and Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmacological Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Jitika Rajpoot
- Center for Structural Biology, Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - Xiaoyue Zhu
- George and Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmacological Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Feng Xu
- George and Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmacological Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| | - Steven O Smith
- Center for Structural Biology, Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, United States
| | - William E Van Nostrand
- George and Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmacological Sciences, University of Rhode Island, Kingston, Rhode Island 02881, United States
| |
Collapse
|
8
|
Li CY, Fan LY, Lin CH, Hu CJ, Chiu MJ. Ultrasensitive Assays Detect Different Conformations of Plasma β Amyloids. ACS OMEGA 2025; 10:7256-7263. [PMID: 40028141 PMCID: PMC11865983 DOI: 10.1021/acsomega.4c10879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
With the developments of ultrasensitive technologies such as immunomagnetic reduction (IMR) assay, single molecule array (SIMOA) assay, electrochemiluminescence immunoassay (ECLIA), the assay of blood-based amyloid 1-42 (Aβ1-42) becomes possible. However, the changes in measured plasma Aβ1-42 concentrations in Alzheimer's disease (AD) compared to cognitively unimpaired subjects (CU) are inconsistent. A possible reason for the inconsistency regarding various conformations of Aβ1-42 in plasma is explored in this study. Three samples with equal amounts of Aβ1-42 but different proportions of monomers and oligomers of Aβ1-42 were prepared. The Aβ1-42 composition of monomers and oligomers in samples was analyzed with Western blot. Identically diluted versions of these three samples were assayed with IMR and SIMOA for Aβ1-42 concentrations. The three diluted samples showed similar levels of Aβ1-42 assayed with IMR, whereas much lower levels for samples with more oligomers assayed with SIOMA. The results imply that IMR detects both monomers and oligomers of Aβ1-42. The measured levels of Aβ1-42 are independent of the proportions of monomer or oligomer Aβ1-42 but depend on the total amounts of Aβ1-42. In the case of SIMOA, monomers of Aβ1-42 are the primary target measured. By comparing Aβ1-42 concentrations of the plasma using IMR and SIMOA, the significant difference in plasma Aβ1-42 levels using IMR in AD compared to CU is mainly due to the formations of oligomeric Aβ1-42. Therefore, if the target molecules are monomers of Aβ1-42, SIMOA is the method of choice. Still, if the target molecules should include monomers, small and large oligomers, IMR would be an optimal consideration. In the future, the clinical implications of the proportion of oligomeric Aβ1-42 need to be elucidated.
Collapse
Affiliation(s)
- Chia-Yu Li
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ling-Yun Fan
- Departments
of Neurology, National Taiwan University
Hospital Bei-Hu Branch, Taipei 108, Taiwan
| | - Chin-Hsien Lin
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute
of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department
of Biomedical Engineering, National Taiwan
University, Taipei 106, Taiwan
| | - Chaur-Jong Hu
- Taipei
Neuroscience Institute, Taipei Medical University, New Taipei City, 235 Taiwan
- Department
of Neurology and Dementia Center, Taipei
Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department
of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Jang Chiu
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
9
|
Muhammedkutty FNK, Zhou HX. Membrane-assisted Aβ40 aggregation pathways. CELL REPORTS. PHYSICAL SCIENCE 2025; 6:102436. [PMID: 40083905 PMCID: PMC11905421 DOI: 10.1016/j.xcrp.2025.102436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease (AD) is caused by the assembly of amyloid-beta (Aβ) peptides into oligomers and fibrils. Endogenous Aβ aggregation may be assisted by cell membranes, which can accelerate the nucleation step enormously, but knowledge of membrane-assisted aggregation is still very limited. Here, we used extensive molecular dynamics (MD) simulations to structurally and energetically characterize key intermediates along the membrane-assisted aggregation pathways of Aβ40. Reinforcing experimental observations, the simulations reveal unique roles of GM1 ganglioside and cholesterol in stabilizing membrane-embedded β sheets and of Y10 and K28 in the ordered release of a small oligomeric seed into solution. The same seed leads to either an open-shaped or R-shaped fibril, with significant stabilization provided by inter- or intra-subunit interfaces between a straight β sheet (residues Q15-D23) and a bent β sheet (residues A30-V36). This work presents a comprehensive picture of membrane-assisted aggregation of Aβ40, with broad implications for developing AD therapies and rationalizing disease-specific polymorphisms of amyloidogenic proteins.
Collapse
Affiliation(s)
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
- Department of Physics, University of Illinois Chicago, Chicago, IL, USA
- Lead contact
| |
Collapse
|
10
|
Iida-Adachi A, Nabika H. Changes in Adsorption, Aggregation, and Diffusion Nature of Amyloid β on a Lipid Membrane in an Open System. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3121-3129. [PMID: 39883914 DOI: 10.1021/acs.langmuir.4c03663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The aggregation and accumulation of amyloid β 42 (Aβ42) peptides on the surface of brain cells is associated with Alzheimer's disease (AD); however, the underlying molecular mechanisms remain unclear. Herein, we used a unique brain-mimetic open system that continuously flows Aβ42 solution to analyze the initial aggregation and adsorptive nature of Aβ42 at physiological concentrations on the lipid membrane. The open system accelerated the adsorption and dimerization kinetics. Upon the addition of Aβ42, monomeric Aβ42 was dominant on the lipid bilayer surface in the closed system with no flow, whereas dimers and high-order oligomers were dominant in the open system. Closed and open systems exhibited different oligomerization kinetics, lipid-Aβ42 interactions, and diffusive properties of monomers and oligomers. These results indicate the specific adsorptive and diffusive nature of Aβ42 on the cell membrane. The open system may help in elucidating the molecular mechanisms underlying AD progression.
Collapse
Affiliation(s)
- Akane Iida-Adachi
- Department of Science, Graduate School of Science and Engineering, Yamagata University, 1-4-12, Kojirakawa, Yamagata 990-8560, Japan
| | - Hideki Nabika
- Faculty of Science, Yamagata University, 1-4-12, Kojirakawa, Yamagata 990-8560, Japan
| |
Collapse
|
11
|
Ni Z, Tan J, Luo Y, Ye S. Dynamic protein hydration water mediates the aggregation kinetics of amyloid β peptides at interfaces. J Colloid Interface Sci 2025; 679:539-546. [PMID: 39467365 DOI: 10.1016/j.jcis.2024.10.105] [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: 07/12/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Protein hydration water is essential for protein misfolding and amyloid formation, but how it directs the course of amyloid formation has yet to be elucidated. Here, we experimentally demonstrated that femtosecond sum frequency generation vibrational spectroscopy (SFG-VS) and the femtosecond IR pump-SFG probe technique can serve as powerful tools for addressing this issue. Using amyloid β(1-42) peptide as a model, we determined the transient misfolding intermediates by probing the amide band spectral features and the local hydration water changes by measuring the ultrafast vibrational dynamics of the amide I band. For the first time, we established a correlation between the dynamic change in protein hydration water and aggregation propensity. The aggregation propensity depends on the dynamic change in the hydration water, rather than the static hydration water content of the initial protein state. Water expulsion enhances the aggregation propensity and promotes amyloid formation, while protein hydration attenuates the aggregation propensity and inhibits amyloid formation. The suppression of water expulsion and protein hydration can prevent protein aggregation and stabilize proteins. These findings contribute to a better understanding of the underlying effect of hydration water on amyloid formation and protein structural stability and provide a strategy for maintaining long-term stabilization of biomolecules.
Collapse
Affiliation(s)
- Zijian Ni
- Hefei National Research Center for Physical Sciences at the Microscale, and Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China
| | - Junjun Tan
- Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China
| | - Yi Luo
- Hefei National Research Center for Physical Sciences at the Microscale, and Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China; Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China.
| | - Shuji Ye
- Hefei National Research Center for Physical Sciences at the Microscale, and Department of Chemical Physics, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230026, China; Hefei National Laboratory, University of Science and Technology of China, Hefei, Anhui 230088, China.
| |
Collapse
|
12
|
Sant V, Matthes D, Mazal H, Antonschmidt L, Wieser F, Movellan KT, Xue K, Nimerovsky E, Stampolaki M, Nathan M, Riedel D, Becker S, Sandoghdar V, de Groot BL, Griesinger C, Andreas LB. Lipidic folding pathway of α-Synuclein via a toxic oligomer. Nat Commun 2025; 16:760. [PMID: 39824800 PMCID: PMC11742675 DOI: 10.1038/s41467-025-55849-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Aggregation intermediates play a pivotal role in the assembly of amyloid fibrils, which are central to the pathogenesis of neurodegenerative diseases. The structures of filamentous intermediates and mature fibrils are now efficiently determined by single-particle cryo-electron microscopy. By contrast, smaller pre-fibrillar α-Synuclein (αS) oligomers, crucial for initiating amyloidogenesis, remain largely uncharacterized. We report an atomic-resolution structural characterization of a toxic pre-fibrillar aggregation intermediate (I1) on pathway to the formation of lipidic fibrils, which incorporate lipid molecules on protofilament surfaces during fibril growth on membranes. Super-resolution microscopy reveals a tetrameric state, providing insights into the early oligomeric assembly. Time resolved nuclear magnetic resonance (NMR) measurements uncover a structural reorganization essential for the transition of I1 to mature lipidic L2 fibrils. The reorganization involves the transformation of anti-parallel β-strands during the pre-fibrillar I1 state into a β-arc characteristic of amyloid fibrils. This structural reconfiguration occurs in a conserved structural kernel shared by a vast number of αS-fibril polymorphs including extracted fibrils from Parkinson's and Lewy Body Dementia patients. Consistent with reports of anti-parallel β-strands being a defining feature of toxic αS pre-fibrillar intermediates, I1 impacts viability of neuroblasts and disrupts cell membranes, resulting in an increased calcium influx. Our results integrate the occurrence of anti-parallel β-strands as salient features of toxic oligomers with their significant role in the amyloid fibril assembly pathway. These structural insights have implications for the development of therapies and biomarkers.
Collapse
Affiliation(s)
- Vrinda Sant
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dirk Matthes
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hisham Mazal
- Max Planck Institute for Science of Light, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Leif Antonschmidt
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Franz Wieser
- Max Planck Institute for Science of Light, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Kumar T Movellan
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Brown Laboratory Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, USA
| | - Kai Xue
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Center of High Field Imaging, Nanyang Technological University, Singapore, Singapore
| | - Evgeny Nimerovsky
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Marianna Stampolaki
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Magdeline Nathan
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dietmar Riedel
- Facility for Electron Microscopy, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan Becker
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vahid Sandoghdar
- Max Planck Institute for Science of Light, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Bert L de Groot
- Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Christian Griesinger
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Loren B Andreas
- NMR Based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
13
|
Samdin TD, Kreutzer AG, Sahrai V, Wierzbicki M, Nowick JS. α-Methylation Enables the X-ray Crystallographic Observation of Oligomeric Assemblies Formed by a β-Hairpin Peptide Derived from Aβ. J Org Chem 2025; 90:394-400. [PMID: 39689228 PMCID: PMC11731301 DOI: 10.1021/acs.joc.4c02344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024]
Abstract
The assembly of the β-amyloid peptide Aβ into toxic oligomers plays a significant role in the neurodegeneration associated with the pathogenesis of Alzheimer's disease. Our laboratory has developed N-methylation as a tool to enable X-ray crystallographic studies of oligomers formed by macrocyclic β-hairpin peptides derived from Aβ. In this investigation, we set out to determine whether α-methylation could be used as an alternative to N-methylation in studying the oligomerization of a β-hairpin peptide derived from Aβ. α-Methylation permits the crystallographic assembly of a triangular trimer and ball-shaped dodecamer, resembling assemblies formed by the N-methylated homolog. Subtle differences are observed in the conformation of the α-methylated peptide when compared to the N-methylated homolog. Notably, α-methylation appears to promote a flatter and more extended β-sheet conformation than that of N-methylated β-sheets or a typical unmodified β-sheet. α-Methylation provides an alternative to N-methylation in X-ray crystallographic studies of oligomers formed by peptides derived from Aβ, with the attractive feature of preserving NH hydrogen-bond donors along the peptide backbone.
Collapse
Affiliation(s)
- Tuan D. Samdin
- Department
of Chemistry, University of California, Irvine, California 92697, United States
| | - Adam G. Kreutzer
- Department
of Chemistry, University of California, Irvine, California 92697, United States
| | - Victoria Sahrai
- Department
of Chemistry, University of California, Irvine, California 92697, United States
| | - Michał Wierzbicki
- Department
of Chemistry, University of California, Irvine, California 92697, United States
| | - James S. Nowick
- Department
of Chemistry, University of California, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California, Irvine, California 92697, United States
| |
Collapse
|
14
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
15
|
Ruzmetov T, Hung TI, Jonnalagedda SP, Chen SH, Fasihianifard P, Guo Z, Bhanu B, Chang CEA. Sampling Conformational Ensembles of Highly Dynamic Proteins via Generative Deep Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592587. [PMID: 38979147 PMCID: PMC11230202 DOI: 10.1101/2024.05.05.592587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Proteins are inherently dynamic, and their conformational ensembles are functionally important in biology. Large-scale motions may govern protein structure-function relationship, and numerous transient but stable conformations of Intrinsically Disordered Proteins (IDPs) can play a crucial role in biological function. Investigating conformational ensembles to understand regulations and disease-related aggregations of IDPs is challenging both experimentally and computationally. In this paper we first introduce a deep learning-based model, termed Internal Coordinate Net (ICoN), which learns the physical principles of conformational changes from Molecular Dynamics (MD) simulation data. Second, we selected interpolating data points in the learned latent space that rapidly identify novel synthetic conformations with sophisticated and large-scale sidechains and backbone arrangements. Third, with the highly dynamic amyloid-β 1-42 (Aβ42) monomer, our deep learning model provided a comprehensive sampling of Aβ42's conformational landscape. Analysis of these synthetic conformations revealed conformational clusters that can be used to rationalize experimental findings. Additionally, the method can identify novel conformations with important interactions in atomistic details that are not included in the training data. New synthetic conformations showed distinct sidechain rearrangements that are probed by our EPR and amino acid substitution studies. This approach is highly transferable and can be used for any available data for training. The work also demonstrated the ability of deep learning to utilize learned natural atomistic motions in protein conformation sampling.
Collapse
|
16
|
Cehlar O, Njemoga S, Horvath M, Cizmazia E, Bednarikova Z, Barrera EE. Structures of Oligomeric States of Tau Protein, Amyloid-β, α-Synuclein and Prion Protein Implicated in Alzheimer's Disease, Parkinson's Disease and Prionopathies. Int J Mol Sci 2024; 25:13049. [PMID: 39684761 DOI: 10.3390/ijms252313049] [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: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
In this review, we focus on the biophysical and structural aspects of the oligomeric states of physiologically intrinsically disordered proteins and peptides tau, amyloid-β and α-synuclein and partly disordered prion protein and their isolations from animal models and human brains. These protein states may be the most toxic agents in the pathogenesis of Alzheimer's and Parkinson's disease. It was shown that oligomers are important players in the aggregation cascade of these proteins. The structural information about these structural states has been provided by methods such as solution and solid-state NMR, cryo-EM, crosslinking mass spectrometry, AFM, TEM, etc., as well as from hybrid structural biology approaches combining experiments with computational modelling and simulations. The reliable structural models of these protein states may provide valuable information for future drug design and therapies.
Collapse
Affiliation(s)
- Ondrej Cehlar
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Stefana Njemoga
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Marian Horvath
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Erik Cizmazia
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Zuzana Bednarikova
- Institute of Experimental Physics, Slovak Academy of Sciences, 04001 Kosice, Slovakia
| | - Exequiel E Barrera
- Instituto de Histología y Embriología (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CC56, Universidad Nacional de Cuyo, Mendoza M5502JMA, Argentina
| |
Collapse
|
17
|
Tolstova AP, Adzhubei AA, Strelkova MA, Makarov AA, Mitkevich VA. Survey of the Aβ-peptide structural diversity: molecular dynamics approaches. Biophys Rev 2024; 16:701-722. [PMID: 39830132 PMCID: PMC11735825 DOI: 10.1007/s12551-024-01253-y] [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: 10/08/2024] [Accepted: 11/04/2024] [Indexed: 01/22/2025] Open
Abstract
The review deals with the application of Molecular Dynamics (MD) to the structure modeling of beta-amyloids (Aβ), currently classified as intrinsically disordered proteins (IDPs). In this review, we strive to relate the main advances in this area but specifically focus on the approaches and methodology. All relevant papers on the Aβ modeling are cited in the Tables in Supplementary Data, including a concise description of the applied approaches, sorted according to the types of the studied systems: modeling of the monomeric Aβ and Aβ aggregates. Similar sections focused according to the type of modeled object are present in the review. In the final part of the review, novel methods of general IDP modeling not confined to Aβ are described. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-024-01253-y.
Collapse
Affiliation(s)
- Anna P. Tolstova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Alexei A. Adzhubei
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
- Washington University School of Medicine and Health Sciences, Washington, DC USA
| | - Maria A. Strelkova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str. 32, 119991 Moscow, Russia
| |
Collapse
|
18
|
Özdemir AY, Hofbauerová K, Kopecký V, Novotný J, Rudajev V. Different amyloid β42 preparations induce different cell death pathways in the model of SH-SY5Y neuroblastoma cells. Cell Mol Biol Lett 2024; 29:143. [PMID: 39551742 PMCID: PMC11572474 DOI: 10.1186/s11658-024-00657-8] [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: 07/19/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024] Open
Abstract
Amyloid β42 (Aβ42) plays a decisive role in the pathology of Alzheimer's disease. The Aβ42 peptide can aggregate into various supramolecular structures, with oligomers being the most toxic form. However, different Aβ species that cause different effects have been described. Many cell death pathways can be activated in connection with Aβ action, including apoptosis, necroptosis, pyroptosis, oxidative stress, ferroptosis, alterations in mitophagy, autophagy, and endo/lysosomal functions. In this study, we used a model of differentiated SH-SY5Y cells and applied two different Aβ42 preparations for 2 and 4 days. Although we found no difference in the shape and size of Aβ species prepared by two different methods (NaOH or NH4OH for Aβ solubilization), we observed strong differences in their effects. Treatment of cells with NaOH-Aβ42 mainly resulted in damage of mitochondrial function and increased production of reactive oxygen species, whereas application of NH4OH-Aβ42 induced necroptosis and first steps of apoptosis, but also caused an increase in protective Hsp27. Moreover, the two Aβ42 preparations differed in the mechanism of interaction with the cells, with the effect of NaOH-Aβ42 being dependent on monosialotetrahexosylganglioside (GM1) content, whereas the effect of NH4OH-Aβ42 was independent of GM1. This suggests that, although both preparations were similar in size, minor differences in secondary/tertiary structure are likely to strongly influence the resulting processes. Our work reveals, at least in part, one of the possible causes of the inconsistency in the data observed in different studies on Aβ-toxicity pathways.
Collapse
Affiliation(s)
- Alp Yigit Özdemir
- Department of Physiology, Faculty of Sciences, Charles University, Viničná 7, 12844, Prague 2, Czech Republic
| | - Kateřina Hofbauerová
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, Ke Karlovu 5, 12116, Prague 2, Czech Republic
| | - Vladimír Kopecký
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, Ke Karlovu 5, 12116, Prague 2, Czech Republic
| | - Jiří Novotný
- Department of Physiology, Faculty of Sciences, Charles University, Viničná 7, 12844, Prague 2, Czech Republic
| | - Vladimír Rudajev
- Department of Physiology, Faculty of Sciences, Charles University, Viničná 7, 12844, Prague 2, Czech Republic.
| |
Collapse
|
19
|
Seychell RM, El Saghir A, Vassallo N. Modulation of Biological Membranes Using Small-Molecule Compounds to Counter Toxicity Caused by Amyloidogenic Proteins. MEMBRANES 2024; 14:231. [PMID: 39590617 PMCID: PMC11596372 DOI: 10.3390/membranes14110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
The transition of peptides or proteins along a misfolding continuum from soluble functional states to pathological aggregates, to ultimately deposit as amyloid fibrils, is a process that underlies an expanding group of human diseases-collectively known as protein-misfolding disorders (PMDs). These include common and debilitating conditions, such as Alzheimer's disease, Parkinson's disease, and type-2 diabetes. Compelling evidence has emerged that the complex interplay between the misfolded proteins and biological membranes is a key determinant of the pathogenic mechanisms by which harmful amyloid entities are formed and exert their cytotoxicity. Most efforts thus far to develop disease-modifying treatments for PMDs have largely focused on anti-aggregation strategies: to neutralise, or prevent the formation of, toxic amyloid species. Herein, we review the critical role of the phospholipid membrane in mediating and enabling amyloid pathogenicity. We consequently propose that the development of small molecules, which have the potential to uniquely modify the physicochemical properties of the membrane and make it more resilient against damage by misfolded proteins, could provide a novel therapeutic approach in PMDs. By way of an example, natural compounds shown to intercalate into lipid bilayers and inhibit amyloid-lipid interactions, such as the aminosterols, squalamine and trodusquamine, cholesterol, ubiquinone, and select polyphenols, are discussed. Such a strategy would provide a novel approach to counter a wide range of toxic biomolecules implicit in numerous human amyloid pathologies.
Collapse
Affiliation(s)
- Raina Marie Seychell
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Adam El Saghir
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
20
|
Bulgart HR, Lopez Perez MA, Tucker A, Giarrano GN, Banford K, Miller O, Bonser SWG, Wold LE, Scharre D, Weisleder N. Plasma membrane repair defect in Alzheimer's disease neurons is driven by the reduced dysferlin expression. FASEB J 2024; 38:e70099. [PMID: 39400395 PMCID: PMC11486262 DOI: 10.1096/fj.202401731rr] [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: 07/27/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and a defect in neuronal plasma membrane repair could exacerbate neurotoxicity, neuronal death, and disease progression. In this study, application of AD patient cerebrospinal fluid (CSF) and recombinant human Aβ to otherwise healthy neurons induces defective neuronal plasma membrane repair in vitro and ex vivo. We identified Aβ as the biochemical component in patient CSF leading to compromised repair capacity and depleting Aβ rescued repair capacity. These elevated Aβ levels reduced expression of dysferlin, a protein that facilitates membrane repair, by altering autophagy and reducing dysferlin trafficking to sites of membrane injury. Overexpression of dysferlin and autophagy inhibition rescued membrane repair. Overall, these findings indicate an AD pathogenic mechanism where Aβ impairs neuronal membrane repair capacity and increases susceptibility to cell death. This suggests that membrane repair could be therapeutically targeted in AD to restore membrane integrity and reduce neurotoxicity and neuronal death.
Collapse
Affiliation(s)
- Hannah R. Bulgart
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Miguel A. Lopez Perez
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Alexis Tucker
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Gianni N. Giarrano
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Kassidy Banford
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Olivia Miller
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Sidney W. G. Bonser
- Department of Applied Statistics and Research MethodsUniversity of Northern ColoradoGreeleyColoradoUSA
| | - Loren E. Wold
- Division of Cardiac Surgery, Department of Surgery, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Douglas Scharre
- Department of NeurologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Noah Weisleder
- Department of Physiology & Cell Biology, Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Molecular and Cellular BiochemistryUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| |
Collapse
|
21
|
Gonçalves PB, Sodero ACR, Cordeiro Y. Natural products targeting amyloid-β oligomer neurotoxicity in Alzheimer's disease. Eur J Med Chem 2024; 276:116684. [PMID: 39032401 DOI: 10.1016/j.ejmech.2024.116684] [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: 06/06/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) constitutes a major global health issue, characterized by progressive neurodegeneration and cognitive impairment, for which no curative treatment is currently available. Current therapeutic approaches are focused on symptom management, highlighting the critical need for disease-modifying therapy. The hallmark pathology of AD involves the aggregation and accumulation of amyloid-β (Aβ) peptides in the brain. Consequently, drug discovery efforts in recent decades have centered on the Aβ aggregation cascade, which includes the transition of monomeric Aβ peptides into toxic oligomers and, ultimately, mature fibrils. Historically, anti-Aβ strategies focused on the clearance of amyloid fibrils using monoclonal antibodies. However, substantial evidence has highlighted the critical role of Aβ oligomers (AβOs) in AD pathogenesis. Soluble AβOs are now recognized as more toxic than fibrils, directly contributing to synaptic impairment, neuronal damage, and the onset of AD. Targeting AβOs has emerged as a promising therapeutic approach to mitigate cognitive decline in AD. Natural products (NPs) have demonstrated promise against AβO neurotoxicity through various mechanisms, including preventing AβO formation, enhancing clearance mechanisms, or converting AβOs into non-toxic species. Understanding the mechanisms by which anti-AβO NPs operate is useful for developing disease-modifying treatments for AD. In this review, we explore the role of NPs in mitigating AβO neurotoxicity for AD drug discovery, summarizing key evidence from biophysical methods, cellular assays, and animal models. By discussing how NPs modulate AβO neurotoxicity across various experimental systems, we aim to provide valuable insights into novel therapeutic strategies targeting AβOs in AD.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-900, Brazil
| |
Collapse
|
22
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
23
|
Deng JL, Huang LF, Bian ZY, Feng XY, Qi RY, Dong WX, Gao JM, Tang JJ. A new neuroprotective candidate TJ1 targeting amyloidogenesis in 5xFAD Alzheimer's disease mice. Int Immunopharmacol 2024; 138:112653. [PMID: 38996664 DOI: 10.1016/j.intimp.2024.112653] [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: 03/04/2024] [Revised: 06/27/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
As one of the main pathmechanisms of Alzheimer's disease (AD), amyloid-β (Aβ) is widely considered to be the prime target for the development of AD therapy. Recently, imidazolylacetophenone oxime ethers or esters (IOEs) have shown neuroprotective effects against neuronal cells damage, suggesting their potential use in the prevention and treatment of AD. Thirty IOEs compounds from our lab in-house library were constructed and screened for the inhibitory effects on Aβ42-induced cytotoxicity. Among them, TJ1, as a new IOEs hit, preliminarily showed the effect on inhibiting Aβ42-induced cytotoxicity. Furthermore, the inhibitory effects of TJ1 on Aβ42 aggregation were tested by ThT assays and TEM. The neuroprotective effects of TJ1 were evaluated in Aβ42-stimulated SH-SY5Y cells, LPS-stimulated BV-2 cells, and H2O2- and RSL3-stimulated PC12 cells. The cognitive improvement of TJ1 was assessed in 5xFAD (C57BL/6J) transgenic mouse. These results showed that TJ1 had strong neuroprotective effects and high blood-brain barrier (BBB) permeability without obvious cytotoxicity. TJ1 impeded the self-accumulation process of Aβ42 by acting on Aβ oligomerization and fibrilization. Besides, TJ1 reversed Aβ-, H2O2- and RSL3-induced neuronal cell damage and decreased neuroinflammation. In 5xFAD mice, TJ1 improved cognitive impairment, increased GSH level, reduced the level of Aβ42 and Aβ plaques, and attenuated the glia reactivation and inflammatory response in the brain,. Taken together, our results demonstrate that TJ1 improves cognitive impairments as a new neuroprotective candidate via targeting amyloidogenesis, which suggests the potential of TJ1 as a treatment for AD.
Collapse
Affiliation(s)
- Jia-Le Deng
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Lan-Fang Huang
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Zhao-Yuan Bian
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Xu-Yao Feng
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Ruo-Yu Qi
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Wei-Xuan Dong
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China
| | - Jiang-Jiang Tang
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
24
|
Muhammedkutty FNK, Zhou HX. Membrane-assisted Aβ40 aggregation pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611426. [PMID: 39282376 PMCID: PMC11398458 DOI: 10.1101/2024.09.05.611426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Alzheimer's disease (AD) is caused by the assembly of amyloid-beta (Aβ) peptides into oligomers and fibrils. Endogenous Aβ aggregation may be assisted by cell membranes, which can accelerate the nucleation step enormously, but knowledge of membrane-assisted aggregation is still very limited. Here we used extensive MD simulations to structurally and energetically characterize key intermediates along the membrane-assisted aggregation pathways of Aβ40. Reinforcing experimental observations, the simulations reveal unique roles of GM1 ganglioside and cholesterol in stabilizing membrane-embedded β-sheets and of Y10 and K28 in the ordered release of a small oligomeric seed into solution. The same seed leads to either an open-shaped or R-shaped fibril, with significant stabilization provided by inter- or intra-subunit interfaces between a straight β-sheet (residues Q15-D23) and a bent β-sheet (residues A30-V36). This work presents the first comprehensive picture of membrane-assisted aggregation of Aβ40, with broad implications for developing AD therapies and rationalizing disease-specific polymorphisms of amyloidogenic proteins.
Collapse
Affiliation(s)
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
- Department of Physics, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
25
|
Bhopatkar AA, Bhatt N, Haque MA, Xavier R, Fung L, Jerez C, Kayed R. MAPT mutations associated with familial tauopathies lead to formation of conformationally distinct oligomers that have cross-seeding ability. Protein Sci 2024; 33:e5099. [PMID: 39145409 PMCID: PMC11325167 DOI: 10.1002/pro.5099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 08/16/2024]
Abstract
The microtubule associated protein, tau, is implicated in a multitude of neurodegenerative disorders that are collectively termed as tauopathies. These disorders are characterized by the presence of tau aggregates within the brain of afflicted individuals. Mutations within the MAPT gene that encodes the tau protein form the genetic backdrop for familial forms of tauopathies, such as frontotemporal dementia (FTD), but the molecular consequences of such alterations and their pathological effects are unclear. We sought to investigate the conformational properties of the aggregates of three tau mutants: A152T, P301L, and R406W, all implicated within FTD, and compare them to those of the native form (WT-Tau 2N4R). Our immunochemical analysis reveals that mutants and WT tau oligomers exhibit similar affinity for conformation-specific antibodies but have distinct morphology and secondary structure. Additionally, these oligomers possess different dye-binding properties and varying sensitivity to proteolytic processing. These results point to conformational variety among them. We then tested the ability of the mutant oligomers to cross-seed the aggregation of WT tau monomer. Using similar array of experiments, we found that cross-seeding with mutant aggregates leads to the formation of conformationally unique WT oligomers. The results discussed in this paper provide a novel perspective on the structural properties of oligomeric forms of WT tau 2N4R and its mutant, along with shedding some light on their cross-seeding behavior.
Collapse
Affiliation(s)
- Anukool A. Bhopatkar
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Nemil Bhatt
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Md Anzarul Haque
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rhea Xavier
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Leiana Fung
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
- Present address:
Neuroscience Graduate Program, UT Southwestern Medical CenterDallasTexasUSA
| | - Cynthia Jerez
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
- Departments of Neurology, Neuroscience and Cell BiologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative DiseasesUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
26
|
Niu Z, Gui X, Feng S, Reif B. Aggregation Mechanisms and Molecular Structures of Amyloid-β in Alzheimer's Disease. Chemistry 2024; 30:e202400277. [PMID: 38888453 DOI: 10.1002/chem.202400277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
Amyloid plaques are a major pathological hallmark involved in Alzheimer's disease and consist of deposits of the amyloid-β peptide (Aβ). The aggregation process of Aβ is highly complex, which leads to polymorphous aggregates with different structures. In addition to aberrant aggregation, Aβ oligomers can undergo liquid-liquid phase separation (LLPS) and form dynamic condensates. It has been hypothesized that these amyloid liquid droplets affect and modulate amyloid fibril formation. In this review, we briefly introduce the relationship between stress granules and amyloid protein aggregation that is associated with neurodegenerative diseases. Then we highlight the regulatory role of LLPS in Aβ aggregation and discuss the potential relationship between Aβ phase transition and aggregation. Furthermore, we summarize the current structures of Aβ oligomers and amyloid fibrils, which have been determined using nuclear magnetic resonance (NMR) and cryo-electron microscopy (cryo-EM). The structural variations of Aβ aggregates provide an explanation for the different levels of toxicity, shed light on the aggregation mechanism and may pave the way towards structure-based drug design for both clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Zheng Niu
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Xinrui Gui
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shuang Feng
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Bernd Reif
- Bavarian NMR Center (B NMRZ), Department of Bioscience, TUM School of Natural Sciences, Technische Universität München (TUM), Garching, 85747, Germany
- Institute of Structural Biology (STB), Helmholtz-Zentrum, München (HMGU), Neuherberg, 85764, Germany
| |
Collapse
|
27
|
Huang Z, Chen Q, Mu X, An Z, Xu Y. Elucidating the Functional Roles of Long Non-Coding RNAs in Alzheimer's Disease. Int J Mol Sci 2024; 25:9211. [PMID: 39273160 PMCID: PMC11394787 DOI: 10.3390/ijms25179211] [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: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative disorder characterized by cognitive decline and neuronal loss, representing a most challenging health issue. We present a computational analysis of transcriptomic data of AD tissues vs. healthy controls, focused on the elucidation of functional roles played by long non-coding RNAs (lncRNAs) throughout the AD progression. We first assembled our own lncRNA transcripts from the raw RNA-Seq data generated from 527 samples of the dorsolateral prefrontal cortex, resulting in the identification of 31,574 novel lncRNA genes. Based on co-expression analyses between mRNAs and lncRNAs, a co-expression network was constructed. Maximal subnetworks with dense connections were identified as functional clusters. Pathway enrichment analyses were conducted over mRNAs and lncRNAs in each cluster, which served as the basis for the inference of functional roles played by lncRNAs involved in each of the key steps in an AD development model that we have previously built based on transcriptomic data of protein-encoding genes. Detailed information is presented about the functional roles of lncRNAs in activities related to stress response, reprogrammed metabolism, cell polarity, and development. Our analyses also revealed that lncRNAs have the discerning power to distinguish between AD samples of each stage and healthy controls. This study represents the first of its kind.
Collapse
Affiliation(s)
- Zhenyu Huang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China;
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.C.); (X.M.)
| | - Qiufen Chen
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.C.); (X.M.)
| | - Xuechen Mu
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.C.); (X.M.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Zheng An
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Ying Xu
- Systems Biology Lab for Metabolic Reprogramming, Department of Human Genetics and Cell Biology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.C.); (X.M.)
| |
Collapse
|
28
|
Samajdar R, Meigooni M, Yang H, Li J, Liu X, Jackson NE, Mosquera MA, Tajkhorshid E, Schroeder CM. Secondary structure determines electron transport in peptides. Proc Natl Acad Sci U S A 2024; 121:e2403324121. [PMID: 39052850 PMCID: PMC11317557 DOI: 10.1073/pnas.2403324121] [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: 02/21/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
Proteins play a key role in biological electron transport, but the structure-function relationships governing the electronic properties of peptides are not fully understood. Despite recent progress, understanding the link between peptide conformational flexibility, hierarchical structures, and electron transport pathways has been challenging. Here, we use single-molecule experiments, molecular dynamics (MD) simulations, nonequilibrium Green's function-density functional theory (NEGF-DFT), and unsupervised machine learning to understand the role of secondary structure on electron transport in peptides. Our results reveal a two-state molecular conductance behavior for peptides across several different amino acid sequences. MD simulations and Gaussian mixture modeling are used to show that this two-state molecular conductance behavior arises due to the conformational flexibility of peptide backbones, with a high-conductance state arising due to a more defined secondary structure (beta turn or 310 helices) and a low-conductance state occurring for extended peptide structures. These results highlight the importance of helical conformations on electron transport in peptides. Conformer selection for the peptide structures is rationalized using principal component analysis of intramolecular hydrogen bonding distances along peptide backbones. Molecular conformations from MD simulations are used to model charge transport in NEGF-DFT calculations, and the results are in reasonable qualitative agreement with experiments. Projected density of states calculations and molecular orbital visualizations are further used to understand the role of amino acid side chains on transport. Overall, our results show that secondary structure plays a key role in electron transport in peptides, which provides broad avenues for understanding the electronic properties of proteins.
Collapse
Affiliation(s)
- Rajarshi Samajdar
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Moeen Meigooni
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Hao Yang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Jialing Li
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Xiaolin Liu
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Nicholas E. Jackson
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Martín A. Mosquera
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT59717
| | - Emad Tajkhorshid
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| | - Charles M. Schroeder
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL61801
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL61801
| |
Collapse
|
29
|
Gonçalves PB, Cordeiro Y, Rennó Sodero AC. Understanding the mechanisms of green tea EGCG against amyloid β oligomer neurotoxicity through computational studies. RSC Adv 2024; 14:22525-22539. [PMID: 39015669 PMCID: PMC11251396 DOI: 10.1039/d4ra03343d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
Oligomeric species of amyloid β peptide (Aβ) are pivotal in Alzheimer's disease (AD) pathogenesis, making them valuable therapeutic targets. Currently, there is no cure or preventive therapy available for AD, with only a few therapeutics offering temporary alleviation of symptoms. Natural products (NPs) are now considered promising anti-amyloid agents. Green tea catechins have garnered considerable attention due to their ability to remodel the toxic amyloid β peptide oligomers (AβOs) into non-toxic assemblies. Nevertheless, the precise molecular mechanism underlying their effects on AβOs remains unclear. In this study, we employ a combination of binding site prediction, molecular docking, and dynamics simulations to gain mechanistic insights into the binding of the potent anti-amyloid epigallocatechin-3-gallate (EGCG) and the less effective catechin, epicatechin (EC), on the structure of pore-forming Aβ tetramers (PDB ID 6RHY). This recently elucidated structure represents AβO(1-42) with two faces of the hydrophobic β-sheet core and hydrophilic edges. Our simulations revealed three potential druggable binding sites within the AβO: two in hydrophilic edges and one in the β-sheet core. Although both catechins bind via hydrogen bond (H-bond) and aromatic interactions to the three potential binding sites, EGCG interacted with key residues more efficiently than EC. We propose that EGCG may remodel AβOs preventing pore formation by binding to the hydrophilic edge binding sites. Additionally, EGCG interacts with key residues in the oligomer's β-sheet core binding site, crucial for fibrillar aggregation. A better understanding of how anti-amyloid compounds remodelling AβOs could be valuable for the development of new therapeutic strategies targeting Aβ in AD. Further experimental validation using point mutations involving key residues could be useful to define whether the establishment of these interactions is crucial for the EGCG remodelling effect.
Collapse
Affiliation(s)
- Priscila Baltazar Gonçalves
- Faculdade de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro RJ 21941-902 Brazil
| | - Yraima Cordeiro
- Faculdade de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro RJ 21941-902 Brazil
| | - Ana Carolina Rennó Sodero
- Faculdade de Farmácia, Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio de Janeiro RJ 21941-902 Brazil
| |
Collapse
|
30
|
Arango AS, Park H, Tajkhorshid E. Topological Learning Approach to Characterizing Biological Membranes. J Chem Inf Model 2024; 64:5242-5252. [PMID: 38912752 PMCID: PMC12009557 DOI: 10.1021/acs.jcim.4c00552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Biological membranes play key roles in cellular compartmentalization, structure, and its signaling pathways. At varying temperatures, individual membrane lipids sample from different configurations, a process that frequently leads to higher-order phase behavior and phenomena. Here, we present a persistent homology (PH)-based method for quantifying the structural features of individual and bulk lipids, providing local and contextual information on lipid tail organization. Our method leverages the mathematical machinery of algebraic topology and machine learning to infer temperature-dependent structural information on lipids from static coordinates. To train our model, we generated multiple molecular dynamics trajectories of dipalmitoyl-phosphatidylcholine membranes at varying temperatures. A fingerprint was then constructed for each set of lipid coordinates by PH filtration, in which interaction spheres were grown around the lipid atoms while tracking their intersections. The sphere filtration formed a simplicial complex that captures enduring key topological features of the configuration landscape using homology, yielding persistence data. Following fingerprint extraction for physiologically relevant temperatures, the persistence data were used to train an attention-based neural network for assignment of effective temperature values to selected membrane regions. Our persistence homology-based method captures the local structural effects, via effective temperature, of lipids adjacent to other membrane constituents, e.g., sterols and proteins. This topological learning approach can predict lipid effective temperatures from static coordinates across multiple spatial resolutions. The tool, called MembTDA, can be accessed at https://github.com/hyunp2/Memb-TDA.
Collapse
Affiliation(s)
- Andres S Arango
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hyun Park
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
31
|
Ikeda K, Sugiura Y, Nakao H, Nakano M. Thermodynamics of oligomerization and Helix-to-sheet structural transition of amyloid β-protein on anionic phospholipid vesicles. Biophys Chem 2024; 310:107248. [PMID: 38653174 DOI: 10.1016/j.bpc.2024.107248] [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: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Understanding oligomerization and aggregation of the amyloid-β protein is important to elucidate the pathological mechanisms of Alzheimer's disease, and lipid membranes play critical roles in this process. In addition to studies reported by other groups, our group has also reported that the negatively-charged lipid bilayers with a high positive curvature induced α-helix-to-β-sheet conformational transitions of amyloid-β-(1-40) upon increase in protein density on the membrane surface and promoted amyloid fibril formation of the protein. Herein, we investigated detailed mechanisms of the conformational transition and oligomer formation of the amyloid-β protein on the membrane surface. Changes in the fractions of the three protein conformers (free monomer, membrane-bound α-helix-rich conformation, and β-sheet-rich conformation) were determined from the fluorescent spectral changes of the tryptophan probe in the protein. The helix-to-sheet structural transition on the surface was described by a thermodynamic model of octamer formation driven by entropic forces including hydrophobic interactions. These findings provide useful information for understanding the self-assembly of amyloidogenic proteins on lipid membrane surfaces.
Collapse
Affiliation(s)
- Keisuke Ikeda
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan.
| | - Yuuki Sugiura
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan
| | - Hiroyuki Nakao
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan
| | - Minoru Nakano
- Department of Biointerface Chemistry, Faculty of Pharmaceutical Sciences, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan
| |
Collapse
|
32
|
Dey A, Patil A, Arumugam S, Maiti S. Single-Molecule Maps of Membrane Insertion by Amyloid-β Oligomers Predict Their Toxicity. J Phys Chem Lett 2024; 15:6292-6298. [PMID: 38855822 DOI: 10.1021/acs.jpclett.4c01048] [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: 06/11/2024]
Abstract
The interaction of small Amyloid-β (Aβ) oligomers with the lipid membrane is an important component of the pathomechanism of Alzheimer's disease (AD). However, oligomers are heterogeneous in size. How each type of oligomer incorporates into the membrane, and how that relates to their toxicity, is unknown. Here, we employ a single molecule technique called Q-SLIP (Quencher-induced Step Length Increase in Photobleaching) to measure the membrane insertion of each monomeric unit of individual oligomers of Aβ42, Aβ40, and Aβ40-F19-Cyclohexyl alanine (Aβ40-F19Cha), and correlate it with their toxicity. We observe that the N-terminus of Aβ42 inserts close to the center of the bilayer, the less toxic Aβ40 inserts to a shallower depth, and the least toxic Aβ40-F19Cha has no specific distribution. This oligomer-specific map provides a mechanistic representation of membrane-mediated Aβ toxicity and should be a valuable tool for AD research.
Collapse
Affiliation(s)
- Arpan Dey
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| | - Abhishek Patil
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC 3800, Australia
| | - Senthil Arumugam
- Monash Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton/Melbourne, VIC 3800, Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, VIC 3800, Australia
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai 400005, India
| |
Collapse
|
33
|
Santos J, Pallarès I, Ventura S. A glimpse into the structural properties of α-synuclein oligomers. Biofactors 2024; 50:439-449. [PMID: 38063360 DOI: 10.1002/biof.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/03/2023] [Indexed: 06/15/2024]
Abstract
α-Synuclein (αS) aggregation is the main neurological hallmark of a group of debilitating neurodegenerative disorders, collectively referred to as synucleinopathies, of which Parkinson's disease is the most prevalent. αS oligomers formed during the initial stages of aggregation are considered key pathogenic drivers of disease onset and progression, standing as privileged targets for therapeutic intervention and diagnosis. However, the structure of αS oligomers and the mechanistic basis of oligomer to fibril conversion are yet poorly understood, thereby precluding the rational formulation of strategies aimed at targeting oligomeric species. In this review, we delve into the recent advances in the structural and mechanistic characterization of αS oligomers. We also discuss how these advances are transforming our understanding of these elusive species and paving the way for oligomer-targeting therapeutics and diagnosis.
Collapse
Affiliation(s)
- Jaime Santos
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irantzu Pallarès
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Ruttenberg SM, Nowick JS. A turn for the worse: Aβ β-hairpins in Alzheimer's disease. Bioorg Med Chem 2024; 105:117715. [PMID: 38615460 PMCID: PMC11876106 DOI: 10.1016/j.bmc.2024.117715] [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: 02/15/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
Amyloid-β (Aβ) oligomers are a cause of neurodegeneration in Alzheimer's disease (AD). These soluble aggregates of the Aβ peptide have proven difficult to study due to their inherent metastability and heterogeneity. Strategies to isolate and stabilize homogenous Aβ oligomer populations have emerged such as mutations, covalent cross-linking, and protein fusions. These strategies along with molecular dynamics simulations have provided a variety of proposed structures of Aβ oligomers, many of which consist of molecules of Aβ in β-hairpin conformations. β-Hairpins are intramolecular antiparallel β-sheets composed of two β-strands connected by a loop or turn. Three decades of research suggests that Aβ peptides form several different β-hairpin conformations, some of which are building blocks of toxic Aβ oligomers. The insights from these studies are currently being used to design anti-Aβ antibodies and vaccines to treat AD. Research suggests that antibody therapies designed to target oligomeric Aβ may be more successful at treating AD than antibodies designed to target linear epitopes of Aβ or fibrillar Aβ. Aβ β-hairpins are good epitopes to use in antibody development to selectively target oligomeric Aβ. This review summarizes the research on β-hairpins in Aβ peptides and discusses the relevance of this conformation in AD pathogenesis and drug development.
Collapse
Affiliation(s)
- Sarah M Ruttenberg
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697-2025, United States
| | - James S Nowick
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697-2025, United States.
| |
Collapse
|
35
|
Liang X, Andrikopoulos N, Tang H, Wang Y, Ding F, Ke PC. Nanoplastic Stimulates the Amyloidogenesis of Parkinson's Alpha-Synuclein NACore. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308753. [PMID: 37988678 PMCID: PMC10994764 DOI: 10.1002/smll.202308753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/04/2023] [Indexed: 11/23/2023]
Abstract
Environmental plastic wastes are potential health hazards due to their prevalence as well as their versatility in initiating physical, chemical, and biological interactions and transformations. Indeed, recent research has implicated the adverse effects of micro- and nano-plastics, including their neurotoxicity, yet how plastic particulates may impact the aggregation pathway and toxicity of amyloid proteins pertinent to the pathologies of neurological diseases remains unknown. Here, electrospray ionization time-of-flight mass spectrometry (ESI-TOF-MS) is employed to reveal the polymorphic oligomerization of NACore, a surrogate of alpha-synuclein that is associated with the pathogenesis of Parkinson's disease. These data indicate that the production rate and population of the NACore oligomers are modulated by their exposure to a polystyrene nanoplastic, and these cellular assays further reveal an elevated NACore toxicity in microglial cells elicited by the nanoplastic. These simulations confirm that the nanoplastic-NACore association is promoted by their hydrophobic interactions. These findings are corroborated by an impairment in zebrafish hatching, survival, and development in vivo upon their embryonic exposure to the nanoplastic. Together, this study has uncovered the dynamics and mechanism of amyloidogenesis elevated by a nanoplastic trigger, shedding a new light on the neurological burden of plastic pollution.
Collapse
Affiliation(s)
- Xiufang Liang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Nicholas Andrikopoulos
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Huayuan Tang
- College of Mechanics and Materials, Hohai University, Nanjing, 211100, China
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - Yue Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - Pu Chun Ke
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| |
Collapse
|
36
|
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
|
37
|
Karkisaval AG, Hassan R, Nguyen A, Balster B, Abedin F, Lal R, Tatulian SA. The structure of tyrosine-10 favors ionic conductance of Alzheimer's disease-associated full-length amyloid-β channels. Nat Commun 2024; 15:1296. [PMID: 38351257 PMCID: PMC10864385 DOI: 10.1038/s41467-023-43821-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 11/21/2023] [Indexed: 02/16/2024] Open
Abstract
Amyloid β (Aβ) ion channels destabilize cellular ionic homeostasis, which contributes to neurotoxicity in Alzheimer's disease. The relative roles of various Aβ isoforms are poorly understood. We use bilayer electrophysiology, AFM imaging, circular dichroism, FTIR and fluorescence spectroscopy to characterize channel activities of four most prevalent Aβ peptides, Aβ1-42, Aβ1-40, and their pyroglutamylated forms (AβpE3-42, AβpE3-40) and correlate them with the peptides' structural features. Solvent-induced fluorescence splitting of tyrosine-10 is discovered and used to assess the sequestration from the solvent and membrane insertion. Aβ1-42 effectively embeds in lipid membranes, contains large fraction of β-sheet in a β-barrel-like structure, forms multi-subunit pores in membranes, and displays well-defined ion channel features. In contrast, the other peptides are partially solvent-exposed, contain minimal β-sheet structure, form less-ordered assemblies, and produce irregular ionic currents. These findings illuminate the structural basis of Aβ neurotoxicity through membrane permeabilization and may help develop therapies that target Aβ-membrane interactions.
Collapse
Affiliation(s)
- Abhijith G Karkisaval
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA
| | - Rowan Hassan
- Department of Physics, University of Central Florida, Orlando, FL, USA
| | - Andrew Nguyen
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Benjamin Balster
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Faisal Abedin
- Department of Physics, University of Central Florida, Orlando, FL, USA
- Department of Biology, Xavier University of Louisiana, New Orleans, LA, USA
| | - Ratnesh Lal
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| | - Suren A Tatulian
- Department of Physics, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
38
|
Wang K, Cai W. Aggregation, structure and water permeability of membrane-embedded helical Aβ oligomers. Phys Chem Chem Phys 2024; 26:5128-5140. [PMID: 38259193 DOI: 10.1039/d3cp05317b] [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: 01/24/2024]
Abstract
It is widely recognized that membranes can facilitate the aggregation of amyloid-β (Aβ) peptides, while Aβ can in turn cause membrane damage. Many studies focus on the peptide-membrane interactions of Aβ oligomers with β-rich structures. However, the exact aggregation and toxicity mechanism of the membrane-embedded helical Aβ oligomers remain ambiguous. Herein, the molecular dynamics simulations were performed on membrane-embedded helical Aβ42 peptides. Initiated by eight Aβ42 monomers embedded in a lipid bilayer, the monomers aggregate into oligomers with stable transmembrane helix structures. With the aggregation of peptides, the membrane perturbations caused by Aβ aggregates decrease. The molecular architectures of oligomers were characterized and a helix-rich octamer stabilized by an annular network of hydrogen bonds was observed. The oligomers demonstrate the capability to assist transmembrane water transport. Our study may provide new insights for the investigation of transmembrane Aβ oligomers.
Collapse
Affiliation(s)
- Ke Wang
- Research Center for Analytical Sciences, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, China.
| | - Wensheng Cai
- Research Center for Analytical Sciences, College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, China.
| |
Collapse
|
39
|
Kreutzer AG, Parrocha CMT, Haerianardakani S, Guaglianone G, Nguyen JT, Diab MN, Yong W, Perez-Rosendahl M, Head E, Nowick JS. Antibodies Raised Against an Aβ Oligomer Mimic Recognize Pathological Features in Alzheimer's Disease and Associated Amyloid-Disease Brain Tissue. ACS CENTRAL SCIENCE 2024; 10:104-121. [PMID: 38292607 PMCID: PMC10823522 DOI: 10.1021/acscentsci.3c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Antibodies that target the β-amyloid peptide (Aβ) and its associated assemblies are important tools in Alzheimer's disease research and have emerged as promising Alzheimer's disease therapies. This paper reports the creation and characterization of a triangular Aβ trimer mimic composed of Aβ17-36 β-hairpins and the generation and study of polyclonal antibodies raised against the Aβ trimer mimic. The Aβ trimer mimic is covalently stabilized by three disulfide bonds at the corners of the triangular trimer to create a homogeneous oligomer. Structural, biophysical, and cell-based studies demonstrate that the Aβ trimer mimic shares characteristics with oligomers of full-length Aβ. X-ray crystallography elucidates the structure of the trimer and reveals that four copies of the trimer assemble to form a dodecamer. SDS-PAGE, size exclusion chromatography, and dynamic light scattering reveal that the trimer also forms higher-order assemblies in solution. Cell-based toxicity assays show that the trimer elicits LDH release, decreases ATP levels, and activates caspase-3/7 mediated apoptosis. Immunostaining studies on brain slices from people who lived with Alzheimer's disease and people who lived with Down syndrome reveal that the polyclonal antibodies raised against the Aβ trimer mimic recognize pathological features including different types of Aβ plaques and cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Adam G. Kreutzer
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Chelsea Marie T. Parrocha
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Sepehr Haerianardakani
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Gretchen Guaglianone
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - Jennifer T. Nguyen
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| | - Michelle N. Diab
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
| | - William Yong
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Mari Perez-Rosendahl
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - Elizabeth Head
- Department
of Pathology and Laboratory Medicine, University
of California Irvine, Irvine, California 92697, United States
| | - James S. Nowick
- Department
of Chemistry, University of California Irvine, Irvine, California 92697, United States
- Department
of Pharmaceutical Sciences, University of
California Irvine, Irvine, California 92697, United States
| |
Collapse
|
40
|
Ruttenberg S, Kreutzer AG, Truex NL, Nowick JS. β-Hairpin Alignment Alters Oligomer Formation in Aβ-Derived Peptides. Biochemistry 2024; 63:212-218. [PMID: 38163326 PMCID: PMC10795187 DOI: 10.1021/acs.biochem.3c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/14/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Amyloid-β (Aβ) forms heterogeneous oligomers, which are implicated in the pathogenesis of Alzheimer's disease (AD). Many Aβ oligomers consist of β-hairpin building blocks─Aβ peptides in β-hairpin conformations. β-Hairpins of Aβ can adopt a variety of alignments, but the role that β-hairpin alignment plays in the formation and heterogeneity of Aβ oligomers is poorly understood. To explore the effect of β-hairpin alignment on the oligomerization of Aβ peptides, we designed and studied two model peptides with two different β-hairpin alignments. Peptides Aβm17-36 and Aβm17-35 mimic two different β-hairpins that Aβ can form, the Aβ17-36 and Aβ17-35 β-hairpins, respectively. These hairpins are similar in composition but differ in hairpin alignment, altering the facial arrangements of the side chains of the residues that they contain. X-ray crystallography and SDS-PAGE demonstrate that the difference in facial arrangement between these peptides leads to distinct oligomer formation. In the crystal state, Aβm17-36 forms triangular trimers that further assemble to form hexamers, while Aβm17-35 forms tetrameric β-barrels. In SDS-PAGE, Aβm17-36 assembles to form a ladder of oligomers, while Aβm17-35 either assembles to form a dimer or does not assemble at all. The differences in the behavior of Aβm17-36 and Aβm17-35 suggest β-hairpin alignment as a source of the observed heterogeneity of Aβ oligomers.
Collapse
Affiliation(s)
- Sarah
M. Ruttenberg
- Department of Chemistry, University of California, Irvine Irvine, California 92697-2025, United
States
| | - Adam G. Kreutzer
- Department of Chemistry, University of California, Irvine Irvine, California 92697-2025, United
States
| | - Nicholas L. Truex
- Department of Chemistry, University of California, Irvine Irvine, California 92697-2025, United
States
| | - James S. Nowick
- Department of Chemistry, University of California, Irvine Irvine, California 92697-2025, United
States
| |
Collapse
|
41
|
Lv Y, Zhou Y, Dong H, Xu M, Zhang J, Yan M. Ultrasensitive electrochemical detection of amyloid-beta oligomers using double amplification strategy by MXene substrate and covalent organic framework-based probe. Talanta 2024; 266:125134. [PMID: 37659228 DOI: 10.1016/j.talanta.2023.125134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/02/2023] [Accepted: 08/26/2023] [Indexed: 09/04/2023]
Abstract
Most of the existing electrochemical systems failed to achieve satisfactory results in early diagnosis of Alzheimer's disease (AD) owing to a deficiency of effective signal transduction. A new method for the electrochemical detection of AD biomarkers (amyloid-beta oligomers, Aβ1-42 oligomers) was developed based on a double amplification strategy. Titanium carbide (Ti3C2) MXene decorated by gold nanoparticles (Au-MXene) as the electrode substrate not only gave rise to the electrochemical response due to its paradoxical surface area and conspicuous charge mobility, but also provided vast numbers of binding sites for aptamers (Apt) of Aβ1-42 oligomers. Meanwhile, AuNPs were incorporated into covalent organic frameworks (COFs), which were further modified by Apt and electron mediator (toluidine blue, TB). The Apt/TB-Au@COFs composite was utilized as a label because of their improvement of the electron-hole separation efficiency and optimization of the charge-carrier utilization. The proposed electrochemical assay established highly efficient platform for the detection of Aβ1-42 oligomers with a linear range from 0.01 pg mL-1 to 180 pg mL-1 and an ultralow detection limit of 4.27 fg mL-1 (S/N = 3). This biosensing platform had potential applications in molecular diagnostics of AD serum samples.
Collapse
Affiliation(s)
- Yubing Lv
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Yanli Zhou
- Henan Key Laboratory of Biomolecular Recognition and Sensing, College of Chemistry and Chemical Engineering, Shangqiu Normal University, Shangqiu, 476000, Henan, China.
| | - Hui Dong
- Henan Key Laboratory of Biomolecular Recognition and Sensing, College of Chemistry and Chemical Engineering, Shangqiu Normal University, Shangqiu, 476000, Henan, China
| | - Maotian Xu
- Henan Key Laboratory of Biomolecular Recognition and Sensing, College of Chemistry and Chemical Engineering, Shangqiu Normal University, Shangqiu, 476000, Henan, China
| | - Jing Zhang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China.
| | - Mei Yan
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, 250022, Shandong, China.
| |
Collapse
|
42
|
Huang Z. Evidence that Alzheimer's Disease Is a Disease of Competitive Synaptic Plasticity Gone Awry. J Alzheimers Dis 2024; 99:447-470. [PMID: 38669548 PMCID: PMC11119021 DOI: 10.3233/jad-240042] [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] [Indexed: 04/28/2024]
Abstract
Mounting evidence indicates that a physiological function of amyloid-β (Aβ) is to mediate neural activity-dependent homeostatic and competitive synaptic plasticity in the brain. I have previously summarized the lines of evidence supporting this hypothesis and highlighted the similarities between Aβ and anti-microbial peptides in mediating cell/synapse competition. In cell competition, anti-microbial peptides deploy a multitude of mechanisms to ensure both self-protection and competitor elimination. Here I review recent studies showing that similar mechanisms are at play in Aβ-mediated synapse competition and perturbations in these mechanisms underpin Alzheimer's disease (AD). Specifically, I discuss evidence that Aβ and ApoE, two crucial players in AD, co-operate in the regulation of synapse competition. Glial ApoE promotes self-protection by increasing the production of trophic monomeric Aβ and inhibiting its assembly into toxic oligomers. Conversely, Aβ oligomers, once assembled, promote the elimination of competitor synapses via direct toxic activity and amplification of "eat-me" signals promoting the elimination of weak synapses. I further summarize evidence that neuronal ApoE may be part of a gene regulatory network that normally promotes competitive plasticity, explaining the selective vulnerability of ApoE expressing neurons in AD brains. Lastly, I discuss evidence that sleep may be key to Aβ-orchestrated plasticity, in which sleep is not only induced by Aβ but is also required for Aβ-mediated plasticity, underlining the link between sleep and AD. Together, these results strongly argue that AD is a disease of competitive synaptic plasticity gone awry, a novel perspective that may promote AD research.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
43
|
Numaguchi Y, Tsukakoshi K, Takeuchi N, Suzuki Y, Ikebukuro K, Kawano R. Real-time monitoring of the amyloid β 1-42 monomer-to-oligomer channel transition using a lipid bilayer system. PNAS NEXUS 2024; 3:pgad437. [PMID: 38156289 PMCID: PMC10753159 DOI: 10.1093/pnasnexus/pgad437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/06/2023] [Indexed: 12/30/2023]
Abstract
This study describes the observation of the transformation of monomeric amyloid β1-42 (Aβ42) into oligomers in a lipid membrane utilizing a lipid bilayer system for electrophysiological measurement. The relevance of oligomers and protofibrils in Alzheimer's disease (AD) is underscored given their significant neurotoxicity. By closely monitoring the shift of Aβ42 from its monomeric state to forming oligomeric channels in phospholipid membranes, we noted that this transformation transpired within a 2-h frame. We manipulated the lipid membrane's constitution with components such as glycerophospholipid, porcine brain total lipid extract, sphingomyelin (SM), and cholesterol (Chol.) to effectively imitate nerve cell membranes. Interesting findings showcased Chol.'s ability to foster stable oligomeric channel formation in the lipid membrane, with SM and GM1 lipids potentially enhancing channel formation as well. Additionally, the study identified the potential of a catechin derivative, epigallocatechin gallate (EGCG), in obstructing oligomerization. With EGCG present in the outer solution of the Aβ42-infused membrane, a noteworthy reduction in channel current was observed, suggesting the successful inhibition of oligomerization. This conclusion held true in both, prior and subsequent, stages of oligomerization. Our findings shed light on the toxicity of oligomers, promising invaluable information for future advancements in AD treatment strategies.
Collapse
Affiliation(s)
- Yuri Numaguchi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-0011, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-0011, Japan
| | - Nanami Takeuchi
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-0011, Japan
| | - Yuki Suzuki
- Department of Chemistry for Materials, Graduate School of Engineering, Mie University, Mie 514-0102, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-0011, Japan
| | - Ryuji Kawano
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-0011, Japan
| |
Collapse
|
44
|
Sun J, Li L, Xiong L, Chen F, She L, Tang H, Zeng Y, Duan Y, Li L, Wang W, Li G, Zhao X, Liang G. Parthenolide alleviates cognitive dysfunction and neurotoxicity via regulation of AMPK/GSK3β(Ser9)/Nrf2 signaling pathway. Biomed Pharmacother 2023; 169:115909. [PMID: 37992573 DOI: 10.1016/j.biopha.2023.115909] [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: 09/14/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023] Open
Abstract
Alzheimer's disease (AD) stands as the predominant age-related neurodegenerative disorder, for which efficacious treatment remains elusive. An auspicious avenue for this disease lies in natural compounds sourced from tranditional medicine and plant origins. Parthenolide (PTN) is a natural product with multiple biological functionsand. Recent investigations have illuminated PTN's protective properties against neurological maladies; however, its potential therapeutic role against AD remains uncharted. This study aims to explore the role of PTN in treating AD. Our in vitro findings underscore PTN's bioactivity, as evidenced by its capacity to curtail apoptosis, reduce reactive oxygen species (ROS) production, and restore mitochondrial membrane potential in PC12 cells. Moreover, PTN treatment demonstrates a capacity to ameliorate deficits in spatial learning and memory in the 3 ×Tg-AD murine model. Notably, PTN's therapeutic efficacy surpasses that of a clinical agent, donepezil. Mechanistically, PTN's neuroprotective effects stem from its adept regulation of the AMPK/GSK3β(ser9)/Nrf2 signaling pathway and protection on neuronal cells from ROS-related apoptosis. Although the molecular target and the pre-clinical evaluations of PTN need to be further explored, this study indicates PTN as a potential agent or lead compound for the drug development against AD.
Collapse
Affiliation(s)
- Jinfeng Sun
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China
| | - Liwei Li
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Li Xiong
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Fan Chen
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Lingyu She
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hao Tang
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yuqing Zeng
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Ying Duan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China
| | - Luyao Li
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Wei Wang
- Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China
| | - Gao Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin 133002, China.
| | - Xia Zhao
- Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Affiliated Yongkang First People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Zhejiang TCM Key Laboratory of Pharmacology and Translational Research of Natural Products, School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
45
|
Samdin TD, Jones CR, Guaglianone G, Kreutzer AG, Freites JA, Wierzbicki M, Nowick JS. A β-barrel-like tetramer formed by a β-hairpin derived from Aβ. Chem Sci 2023; 15:285-297. [PMID: 38131075 PMCID: PMC10732006 DOI: 10.1039/d3sc05185d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023] Open
Abstract
β-Hairpins formed by the β-amyloid peptide Aβ are building blocks of Aβ oligomers. Three different alignments of β-hairpins have been observed in the structures of Aβ oligomers or fibrils. Differences in β-hairpin alignment likely contribute to the heterogeneity of Aβ oligomers and thus impede their study at high-resolution. Here, we designed, synthesized, and studied a series of β-hairpin peptides derived from Aβ12-40 in one of these three alignments and investigated their solution-phase assembly and folding. These assays reveal the formation of tetramers and octamers that are stabilized by intermolecular hydrogen bonding interactions between Aβ residues 12-14 and 38-40 as part of an extended β-hairpin conformation. X-ray crystallographic studies of one peptide from this series reveal the formation of β-barrel-like tetramers and octamers that are stabilized by edge-to-edge hydrogen bonding and hydrophobic packing. Dye-leakage and caspase 3/7 activation assays using tetramer and octamer forming peptides from this series reveal membrane-damaging and apoptotic properties. A molecular dynamics simulation of the β-barrel-like tetramer embedded in a lipid bilayer shows membrane disruption and water permeation. The tetramers and octamers described herein provide additional models of how Aβ may assemble into oligomers and supports the hypothesis that β-hairpin alignment and topology may contribute directly to oligomer heterogeneity.
Collapse
Affiliation(s)
- Tuan D Samdin
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - Chelsea R Jones
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - Gretchen Guaglianone
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - Adam G Kreutzer
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - J Alfredo Freites
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - Michał Wierzbicki
- Department of Chemistry, University of California Irvine California 92697-2025 USA
| | - James S Nowick
- Department of Chemistry, University of California Irvine California 92697-2025 USA
- Department of Pharmaceutical Sciences, University of California, Irvine Irvine California 92697-2025 USA
| |
Collapse
|
46
|
Arango AS, Park H, Tajkhorshid E. Topological Learning Approach to Characterizing Biological Membranes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.569053. [PMID: 38076911 PMCID: PMC10705453 DOI: 10.1101/2023.11.28.569053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Biological membranes play key roles in cellular compartmentalization, structure, and its signaling pathways. At varying temperatures, individual membrane lipids sample from different configurations, a process that frequently leads to higher-order phase behavior and phenomena. Here we present a persistent homology-based method for quantifying the structural features of individual and bulk lipids, providing local and contextual information on lipid tail organization. Our method leverages the mathematical machinery of algebraic topology and machine learning to infer temperature-dependent structural information of lipids from static coordinates. To train our model, we generated multiple molecular dynamics trajectories of DPPC membranes at varying temperatures. A fingerprint was then constructed for each set of lipid coordinates by a persistent homology filtration, in which interactions spheres were grown around the lipid atoms while tracking their intersections. The sphere filtration formed a simplicial complex that captures enduring key topological features of the configuration landscape, using homology, yielding persistence data. Following fingerprint extraction for physiologically relevant temperatures, the persistence data were used to train an attention-based neural network for assignment of effective temperature values to selected membrane regions. Our persistence homology-based method captures the local structural effects, via effective temperature, of lipids adjacent to other membrane constituents, e.g. sterols and proteins. This topological learning approach can predict lipid effective temperatures from static coordinates across multiple spatial resolutions. The tool, called MembTDA, can be accessed at https://github.com/hyunp2/Memb-TDA.
Collapse
Affiliation(s)
- Andres S Arango
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Hyun Park
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource Center for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
47
|
Muhammedkutty FNK, Prasad R, Gao Y, Sudarshan TR, Robang AS, Watzlawik JO, Rosenberry TL, Paravastu AK, Zhou HX. A common pathway for detergent-assisted oligomerization of Aβ42. Commun Biol 2023; 6:1184. [PMID: 37989804 PMCID: PMC10663524 DOI: 10.1038/s42003-023-05556-w] [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: 07/05/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
Amyloid beta (Aβ) aggregation is a slow process without seeding or assisted nucleation. Sodium dodecyl sulfate (SDS) micelles stabilize Aβ42 small oligomers (in the dimer to tetramer range); subsequent SDS removal leads to a 150-kD Aβ42 oligomer. Dodecylphosphorylcholine (DPC) micelles also stabilize an Aβ42 tetramer. Here we investigate the detergent-assisted oligomerization pathway by solid-state NMR spectroscopy and molecular dynamics simulations. SDS- and DPC-induced oligomers have the same structure, implying a common oligomerization pathway. An antiparallel β-sheet formed by the C-terminal region, the only stable structure in SDS and DPC micelles, is directly incorporated into the 150-kD oligomer. Three Gly residues (at positions 33, 37, and 38) create holes that are filled by the SDS and DPC hydrocarbon tails, thereby turning a potentially destabilizing feature into a stabilizing factor. These observations have implications for endogenous Aβ aggregation at cellular interfaces.
Collapse
Affiliation(s)
| | - Ramesh Prasad
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Yuan Gao
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Tarunya Rao Sudarshan
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Alicia S Robang
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Jens O Watzlawik
- Departments of Neuroscience and Pharmacology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Terrone L Rosenberry
- Departments of Neuroscience and Pharmacology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Anant K Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, GA, 30332, USA.
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, 60607, USA.
- Department of Physics, University of Illinois Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
48
|
Yu Z, Moshood Y, Wozniak MK, Patel S, Terpstra K, Llano DA, Dobrucki LW, Mirica LM. Amphiphilic Molecules Exhibiting Zwitterionic Excited-State Intramolecular Proton Transfer and Near-Infrared Emission for the Detection of Amyloid β Aggregates in Alzheimer's Disease. Chemistry 2023; 29:e202302408. [PMID: 37616059 PMCID: PMC10840928 DOI: 10.1002/chem.202302408] [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: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 08/25/2023]
Abstract
Chromophores with zwitterionic excited-state intramolecular proton transfer (ESIPT) have been shown to have larger Stock shifts and red-shifted emission wavelengths compared to the conventional π-delocalized ESIPT molecules. However, there is still a dearth of design strategies to expand the current library of zwitterionic ESIPT compounds. Herein, a novel zwitterionic excited-state intramolecular proton transfer system is reported, enabled by addition of 1,4,7-triazacyclononane (TACN) fragments on a dicyanomethylene-4H-pyran (DCM) scaffold. The solvent-dependent steady-state photophysical studies, pKa measurements, and computational analysis strongly support that the ESIPT process is more efficient with two TACN groups attached to the DCM scaffold and not affected by polar protic solvents. Impressively, compound DCM-OH-2-DT exhibits a near-infrared (NIR) emission at 740 nm along with an uncommonly large Stokes shift. Moreover, DCM-OH-2-DT shows high affinity towards soluble amyloid β (Aβ) oligomers in vitro and in 5xFAD mouse brain sections, and we have successfully applied DCM-OH-2-DT for the in vivo imaging of Aβ aggregates and demonstrated its potential use as an early diagnostic agent for AD. Overall, this study can provide a general molecular design strategy for developing new zwitterionic ESIPT compounds with NIR emission in vivo imaging applications.
Collapse
Affiliation(s)
- Zhengxin Yu
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Yusuff Moshood
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Marcin K. Wozniak
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Shrey Patel
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Karna Terpstra
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
| | - Daniel A. Llano
- Beckman Institute for Advanced Science and Technology, Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Lawrence W. Dobrucki
- Beckman Institute for Advanced Science and Technology, Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana IL 61801, United States
| | - Liviu M. Mirica
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, The Neuroscience Program, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, United States
| |
Collapse
|
49
|
Han YL, Yin HH, Xiao C, Bernards MT, He Y, Guan YX. Understanding the Molecular Mechanisms of Polyphenol Inhibition of Amyloid β Aggregation. ACS Chem Neurosci 2023; 14:4051-4061. [PMID: 37890131 DOI: 10.1021/acschemneuro.3c00586] [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: 10/29/2023] Open
Abstract
Alzheimer's disease (AD) is highly associated with self-aggregation of amyloid β (Aβ) proteins into fibrils. Inhibition of Aβ aggregation by polyphenols is one of the major therapeutic strategies for AD. Among them, four polyphenols (brazilin, resveratrol, hematoxylin, and rosmarinic acid) have been reported to be effective at inhibiting Aβ aggregation, but the inhibition mechanisms are still unclear. In this work, these four polyphenols were selected to explore their interactions with the Aβ17-42 pentamer by molecular dynamics simulation. All four polyphenols can bind to the pentamer tightly but prefer different binding sites. Conversion of the β-sheet to the random coil, fewer interchain hydrogen bonds, and weaker salt bridges were observed after binding. Interestingly, different Aβ17-42 pentamer destabilizing mechanisms for resveratrol and hematoxylin were found. Resveratrol inserts into the hydrophobic core of the pentamer by forming hydrogen bonds with Asp23 and Lys28, while hematoxylin prefers to bind beside chain A of the pentamer, which leads to β-sheet offset and dissociation of the β1 sheet of chain E. This work reveals the interactions between the Aβ17-42 pentamer and four polyphenols and discusses the relationship between inhibitor structures and their inhibition mechanisms, which also provides useful guidance for screening effective Aβ aggregation inhibitors and drug design against AD.
Collapse
Affiliation(s)
- Yin-Lei Han
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Huan-Huan Yin
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Chao Xiao
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Matthew T Bernards
- Department of Chemical and Biological Engineering, University of Idaho, Moscow 83844, Idaho, United States
| | - Yi He
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Chemical Engineering, University of Washington, Seattle 98195, Washington, United States
| | - Yi-Xin Guan
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
50
|
Meza U, Romero-Méndez C, Sánchez-Armáss S, Rodríguez-Menchaca AA. Role of rafts in neurological disorders. Neurologia 2023; 38:671-680. [PMID: 37858892 DOI: 10.1016/j.nrleng.2023.10.003] [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: 09/03/2020] [Accepted: 01/01/2021] [Indexed: 10/21/2023] Open
Abstract
INTRODUCTION Rafts are protein-lipid structural nanodomains involved in efficient signal transduction and the modulation of physiological processes of the cell plasma membrane. Raft disruption in the nervous system has been associated with a wide range of disorders. DEVELOPMENT We review the concept of rafts, the nervous system processes in which they are involved, and their role in diseases such as Parkinson's disease, Alzheimer disease, and Huntington disease. CONCLUSIONS Based on the available evidence, preservation and/or reconstitution of rafts is a promising treatment strategy for a wide range of neurological disorders.
Collapse
Affiliation(s)
- U Meza
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - C Romero-Méndez
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - S Sánchez-Armáss
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| | - A A Rodríguez-Menchaca
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México.
| |
Collapse
|