CoVAMPnet: Comparative Markov State Analysis for Studying Effects of Drug Candidates on Disordered BiomoleculesClick to copy article linkArticle link copied!
- Sérgio M. MarquesSérgio M. MarquesLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Sérgio M. Marques
- Petr KoubaPetr KoubaLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicCzech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech RepublicFaculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, Dejvice, Praha 6 166 27, Czech RepublicMore by Petr Kouba
- Anthony LegrandAnthony LegrandLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Anthony Legrand
- Jiri SedlarJiri SedlarCzech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech RepublicMore by Jiri Sedlar
- Lucas DissonLucas DissonCzech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech RepublicMore by Lucas Disson
- Joan Planas-IglesiasJoan Planas-IglesiasLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Joan Planas-Iglesias
- Zainab SanusiZainab SanusiLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Zainab Sanusi
- Antonin KunkaAntonin KunkaLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Antonin Kunka
- Jiri DamborskyJiri DamborskyLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Jiri Damborsky
- Tomas PajdlaTomas PajdlaCzech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech RepublicMore by Tomas Pajdla
- Zbynek ProkopZbynek ProkopLoschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Zbynek Prokop
- Stanislav Mazurenko*Stanislav Mazurenko*E-mail: [email protected]Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by Stanislav Mazurenko
- Josef Sivic*Josef Sivic*E-mail: [email protected]Czech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, Dejvice, Praha 6 160 00, Czech RepublicMore by Josef Sivic
- David Bednar*David Bednar*E-mail: [email protected]Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech RepublicInternational Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, Brno 656 91, Czech RepublicMore by David Bednar
Abstract
Computational study of the effect of drug candidates on intrinsically disordered biomolecules is challenging due to their vast and complex conformational space. Here, we developed a comparative Markov state analysis (CoVAMPnet) framework to quantify changes in the conformational distribution and dynamics of a disordered biomolecule in the presence and absence of small organic drug candidate molecules. First, molecular dynamics trajectories are generated using enhanced sampling, in the presence and absence of small molecule drug candidates, and ensembles of soft Markov state models (MSMs) are learned for each system using unsupervised machine learning. Second, these ensembles of learned MSMs are aligned across different systems based on a solution to an optimal transport problem. Third, the directional importance of inter-residue distances for the assignment to different conformational states is assessed by a discriminative analysis of aggregated neural network gradients. This final step provides interpretability and biophysical context to the learned MSMs. We applied this novel computational framework to assess the effects of ongoing phase 3 therapeutics tramiprosate (TMP) and its metabolite 3-sulfopropanoic acid (SPA) on the disordered Aβ42 peptide involved in Alzheimer’s disease. Based on adaptive sampling molecular dynamics and CoVAMPnet analysis, we observed that both TMP and SPA preserved more structured conformations of Aβ42 by interacting nonspecifically with charged residues. SPA impacted Aβ42 more than TMP, protecting α-helices and suppressing the formation of aggregation-prone β-strands. Experimental biophysical analyses showed only mild effects of TMP/SPA on Aβ42 and activity enhancement by the endogenous metabolization of TMP into SPA. Our data suggest that TMP/SPA may also target biomolecules other than Aβ peptides. The CoVAMPnet method is broadly applicable to study the effects of drug candidates on the conformational behavior of intrinsically disordered biomolecules.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Materials and Methods
Molecular Dynamics (MD) Simulations
System Preparation
MD Simulation Protocols
Analyses of Properties in Combined MD Ensembles
Comparative Markov State Model Analysis (CoVAMPnet)
Learning Markov State Models Using Neural Networks
Alignment of Learned States for Comparative Analysis
Aligning States within a Single System
Aligning Ensembles of Markov State Models Between Different Systems
Gradient-Based Characterization of Learned States
Estimation of the Free Energy Landscape
Experimental Validation
Results
Selection of the Computational Protocol for the Simulation of Aβ42
Secondary Structure Content in Simulations of Free Aβ42 and Aβ42 with Ligands
Effects of Ligands on the Evolution of Secondary Structure Elements Over Time
Conformational Analysis of Ligand Effects Using Markov State Models
Construction of Variational Markov State Models
Evaluation of the Effect of Using the Soft versus the Hard Assignment
Alignment of Learned States Across Systems with and without Ligands
Comparison of Learned States Across Systems with and without Ligands
Characterization of Learned Conformational States via Network Gradients
Molecular Interactions
Ligand–Peptide Interactions
Intramolecular Interactions of Aβ42
Experimental Validation
Discussion
Data Availability
The sampled stripped trajectories and intermediate data, including the trained neural network weights, are available at https://data.ciirc.cvut.cz/public/projects/2023CoVAMPnet/. The code and example data are available at https://github.com/KoubaPetr/CoVAMPnet.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacsau.4c00182.
Detailed materials and methods, complementary to the concise descriptions in this main text, Supplementary discussions (Notes 1–10), Supplementary figures: structure of Aβ42 (Figure S1), comparison of the Amber ff14SB and CHARMM36m force fields (Figures S2 and S3), comparison of different adaptive sampling protocols (Figures S4 and S5), temporal alignment and concatenation of the adaptive sampling and classical MDs (Figures S6–S9), conventional Markov state model analysis (Figures S10–S12), variational Markov state analysis using VAMPnets (Figures S13–S18), comparative Markov state model analysis (Figure S19), time-based evolution of the states (Figure S20), radius of gyration by state (Figure S21), characterization of learned conformational states via network gradients (Figures S22 and S23), interactions of Aβ42 with the small molecules (Figures S24–S26), experimental validation (Figures S27–S31), Supplementary tables: comparison of computational protocols for the simulation of Aβ42 (Table S1), analysis of classical MDs (Table S2), summary of the effects of small molecules (Table S3), and intramolecular interactions of Aβ42 (Table S4) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was supported by the Ministry of Education, Youth and Sports of the Czech Republic (grants ESFRI RECETOX RI LM2023069, e-INFRA CZ LM2018140, ESFRI ELIXIR CZ LM2023055, TEAMING CZ CZ.02.1.01/0.0/0.0/17_043/0009632), the European Regional Development Fund under the project IMPACT (reg. no. CZ.02.1.01/0.0/0.0/15_003/0000468), the National Institute for Neurology Research (EXCELES Neuro nr. LX22NPO5107 MEYS), and the European Union (Next Generation EU, SinFonia 814418, TEAMING 857560 and ERC FRONTIER 101097822). Views and opinions expressed are those of the author(s) only and do not necessarily reflect those of the European Union or the European Research Council. Neither the European Union nor the granting authority can be held responsible for them. Petr Kouba is a holder of the Brno Ph.D. Talent scholarship funded by the Brno City Municipality and the JCMM.
References
This article references 96 other publications.
- 1Gustavsson, A.; Norton, N.; Fast, T.; Frölich, L.; Georges, J.; Holzapfel, D.; Kirabali, T.; Krolak-Salmon, P.; Rossini, P. M.; Ferretti, M. T. Global Estimates on the Number of Persons across the Alzheimer’s Disease Continuum. Alzheimer’s Dementia 2022, 19, 658– 670, DOI: 10.1002/alz.12694Google ScholarThere is no corresponding record for this reference.
- 2Benilova, I.; Karran, E.; De Strooper, B. The Toxic Aβ Oligomer and Alzheimer’s Disease: An Emperor in Need of Clothes. Nat. Neurosci. 2012, 15 (3), 349– 357, DOI: 10.1038/nn.3028Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlOltrc%253D&md5=071f3c9b1e0fe9b2e66363c44d87e381The toxic Aβ oligomer and Alzheimer's disease: an emperor in need of clothesBenilova, Iryna; Karran, Eric; De Strooper, BartNature Neuroscience (2012), 15 (3), 349-357CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)A review. The 'toxic Aβ oligomer' hypothesis has attracted considerable attention among Alzheimer's disease researchers as a way of resolving the lack of correlation between deposited amyloid-β (Aβ) in amyloid plaques-in terms of both amt. and location-and cognitive impairment or neurodegeneration. However, the lack of a common, agreed-upon exptl. description of the toxic Aβ oligomer makes interpretation and direct comparison of data between different research groups impossible. Here we critically review the evidence supporting toxic Aβ oligomers as drivers of neurodegeneration and make some suggestions that might facilitate progress in this complex field.
- 3Karran, E.; De Strooper, B. The Amyloid Hypothesis in Alzheimer Disease: New Insights from New Therapeutics. Nat. Rev. Drug Discov. 2022, 21 (4), 306– 318, DOI: 10.1038/s41573-022-00391-wGoogle Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xkt1aru7s%253D&md5=44187b2d3c8e4aae3a27a9c18c80c6c3The amyloid hypothesis in Alzheimer disease: new insights from new therapeuticsKarran, Eric; De Strooper, BartNature Reviews Drug Discovery (2022), 21 (4), 306-318CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)Many drugs that target amyloid-beta (Abeta) in Alzheimer disease (AD) have failed to demonstrate clin. efficacy. However, four anti-Abeta antibodies have been shown to mediate the removal of amyloid plaque from brains of patients with AD, and the FDA has recently granted accelerated approval to one of these, aducanumab, using redn. of amyloid plaque as a surrogate end point. The rationale for approval and the extent of the clin. benefit from these antibodies are under intense debate. With the aim of informing this debate, we review clin. trial data for drugs that target Abeta from the perspective of the temporal interplay between the two pathognomonic protein aggregates in AD - Abeta plaques and tau neurofibrillary tangles - and their relationship to cognitive impairment, highlighting differences in drug properties that could affect their clin. performance. On this basis, we propose that Abeta pathol. drives tau pathol., that amyloid plaque would need to be reduced to a low level (®20 centiloids) to reveal significant clin. benefit and that there will be a lag between the removal of amyloid and the potential to observe a clin. benefit. We conclude that the speed of amyloid removal from the brain by a potential therapy will be important in demonstrating clin. benefit in the context of a clin. trial.
- 4Castellani, R. J.; Plascencia-Villa, G.; Perry, G. The Amyloid Cascade and Alzheimer’s Disease Therapeutics: Theory versus Observation. Lab. Invest. 2019, 99 (7), 958– 970, DOI: 10.1038/s41374-019-0231-zGoogle ScholarThere is no corresponding record for this reference.
- 5Matiiv, A. B.; Trubitsina, N. P.; Matveenko, A. G.; Barbitoff, Y. A.; Zhouravleva, G. A.; Bondarev, S. A. Amyloid and Amyloid-Like Aggregates: Diversity and the Term Crisis. Biochemistry (Moscow) 2020, 85 (9), 1011– 1034, DOI: 10.1134/S0006297920090035Google ScholarThere is no corresponding record for this reference.
- 6Bhattacharya, S.; Lin, X. Recent Advances in Computational Protocols Addressing Intrinsically Disordered Proteins. Biomolecules 2019, 9 (4), 146, DOI: 10.3390/biom9040146Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXovFCgsb4%253D&md5=42bb3479001b582aeaa37b9e25c8db57Recent advances in computational protocols addressing intrinsically disordered proteinsBhattacharya, Supriyo; Lin, XingchengBiomolecules (2019), 9 (4), 146CODEN: BIOMHC; ISSN:2218-273X. (MDPI AG)A review. Intrinsically disordered proteins (IDP) are abundant in the human genome and have recently emerged as major therapeutic targets for various diseases. Unlike traditional proteins that adopt a definitive structure, IDPs in free soln. are disordered and exist as an ensemble of conformations. This enables the IDPs to signal through multiple signaling pathways and serve as scaffolds for multi-protein complexes. The challenge in studying IDPs exptl. stems from their disordered nature. NMR (NMR), CD, small angle X-ray scattering, and single mol. Forster resonance energy transfer (FRET) can give the local structural information and overall dimension of IDPs, but seldom provide a unified picture of the whole protein. To understand the conformational dynamics of IDPs and how their structural ensembles recognize multiple binding partners and small mol. inhibitors, knowledge-based and physics-based sampling techniques are utilized in-silico, guided by exptl. structural data. However, efficient sampling of the IDP conformational ensemble requires traversing the numerous degrees of freedom in the IDP energy landscape, as well as force-fields that accurately model the protein and solvent interactions. In this review, we have provided an overview of the current state of computational methods for studying IDP structure and dynamics and discussed the major challenges faced in this field.
- 7Saurabh, S.; Nadendla, K.; Purohit, S. S.; Sivakumar, P. M.; Cetinel, S. Fuzzy Drug Targets: Disordered Proteins in the Drug-Discovery Realm. ACS Omega 2023, 8 (11), 9729– 9747, DOI: 10.1021/acsomega.2c07708Google ScholarThere is no corresponding record for this reference.
- 8Paul, A.; Samantray, S.; Anteghini, M.; Khaled, M.; Strodel, B. Thermodynamics and Kinetics of the Amyloid-β Peptide Revealed by Markov State Models Based on MD Data in Agreement with Experiment. Chem. Sci. 2021, 12 (19), 6652– 6669, DOI: 10.1039/D0SC04657DGoogle Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXovVaqtrk%253D&md5=53401b7f733acd3039b84361fe413cbcThermodynamics and kinetics of the amyloid-β peptide revealed by Markov state models based on MD data in agreement with experimentPaul, Arghadwip; Samantray, Suman; Anteghini, Marco; Khaled, Mohammed; Strodel, BirgitChemical Science (2021), 12 (19), 6652-6669CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The amlyoid-β peptide (Aβ) is closely linked to the development of Alzheimer's disease. Mol. dynamics (MD) simulations have become an indispensable tool for studying the behavior of this peptide at the atomistic level. General key aspects of MD simulations are the force field used for modeling the peptide and its environment, which is important for accurate modeling of the system of interest, and the length of the simulations, which dets. whether or not equil. is reached. In this study we address these points by analyzing 30-μs MD simulations acquired for Aβ40 using seven different force fields. We assess the convergence of these simulations based on the convergence of various structural properties and of NMR and fluorescence spectroscopic observables. Moreover, we calc Markov state models for the different MD simulations, which provide an unprecedented view of the thermodn. and kinetics of the amyloid-β peptide. This further allows us to provide answers for pertinent questions, like: which force fields are suitable for modeling Aβ (a99SB-UCB and a99SB-ILDN/TIP4P-D); what does Aβ peptide really look like (mostly extended and disordered) and; how long does it take MD simulations of Aβ to attain equil. (at least 20-30μs). We believe the analyses presented in this study will provide a useful ref. guide for important questions relating to the structure and dynamics of Aβ in particular, and by extension other similar disordered proteins.
- 9McGibbon, R. T.; Pande, V. S. Variational Cross-Validation of Slow Dynamical Modes in Molecular Kinetics. J. Chem. Phys. 2015, 142 (12), 124105, DOI: 10.1063/1.4916292Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsVWnur0%253D&md5=dc34e6f64a11721767439e91e68e778aVariational cross-validation of slow dynamical modes in molecular kineticsMcGibbon, Robert T.; Pande, Vijay S.Journal of Chemical Physics (2015), 142 (12), 124105/1-124105/12CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Markov state models are a widely used method for approximating the eigenspectrum of the mol. dynamics propagator, yielding insight into the long-timescale statistical kinetics and slow dynamical modes of biomol. systems. However, the lack of a unified theor. framework for choosing between alternative models has hampered progress, esp. for non-experts applying these methods to novel biol. systems. Here, we consider cross-validation with a new objective function for estimators of these slow dynamical modes, a generalized matrix Rayleigh quotient (GMRQ), which measures the ability of a rank-m projection operator to capture the slow subspace of the system. It is shown that a variational theorem bounds the GMRQ from above by the sum of the first m eigenvalues of the system's propagator, but that this bound can be violated when the requisite matrix elements are estd. subject to statistical uncertainty. This overfitting can be detected and avoided through cross-validation. These result make it possible to construct Markov state models for protein dynamics in a way that appropriately captures the tradeoff between systematic and statistical errors. (c) 2015 American Institute of Physics.
- 10Spiriti, J.; Noé, F.; Wong, C. F. Simulation of Ligand Dissociation Kinetics from the Protein Kinase PYK2. J. Comput. Chem. 2022, 43 (28), 1911– 1922, DOI: 10.1002/jcc.26991Google ScholarThere is no corresponding record for this reference.
- 11Dominic, A. J. I.; Cao, S.; Montoya-Castillo, A.; Huang, X. Memory Unlocks the Future of Biomolecular Dynamics: Transformative Tools to Uncover Physical Insights Accurately and Efficiently. J. Am. Chem. Soc. 2023, 145 (18), 9916– 9927, DOI: 10.1021/jacs.3c01095Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXosFyksr8%253D&md5=be056dee3d0e6c4987050f956bb70a2eMemory Unlocks the Future of Biomolecular Dynamics: Transformative Tools to Uncover Physical Insights Accurately and EfficientlyDominic, Anthony J.; Cao, Siqin; Montoya-Castillo, Andres; Huang, XuhuiJournal of the American Chemical Society (2023), 145 (18), 9916-9927CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A review. Conformational changes underpin function and encode complex biomol. mechanisms. Gaining at.-level detail of how such changes occur has the potential to reveal these mechanisms and is of crit. importance in identifying drug targets, facilitating rational drug design, and enabling bioengineering applications. While the past two decades have brought Markov state model techniques to the point where practitioners can regularly use them to glimpse the long-time dynamics of slow conformations in complex systems, many systems are still beyond their reach. In this Perspective, we discuss how including memory (i.e., non-Markovian effects) can reduce the computational cost to predict the long-time dynamics in these complex systems by orders of magnitude and with greater accuracy and resoln. than state-of-the-art Markov state models. We illustrate how memory lies at the heart of successful and promising techniques, ranging from the Fokker-Planck and generalized Langevin equations to deep-learning recurrent neural networks and generalized master equations. We delineate how these techniques work, identify insights that they can offer in biomol. systems, and discuss their advantages and disadvantages in practical settings. We show how generalized master equations can enable the investigation of, for example, the gate-opening process in RNA polymerase II and demonstrate how our recent advances tame the deleterious influence of statistical underconvergence of the mol. dynamics simulations used to parameterize these techniques. This represents a significant leap forward that will enable our memory-based techniques to interrogate systems that are currently beyond the reach of even the best Markov state models. We conclude by discussing some current challenges and future prospects for how exploiting memory will open the door to many exciting opportunities.
- 12Noé, F.; Wu, H.; Prinz, J.-H.; Plattner, N. Projected and Hidden Markov Models for Calculating Kinetics and Metastable States of Complex Molecules. J. Chem. Phys. 2013, 139 (18), 184114, DOI: 10.1063/1.4828816Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslKqt77P&md5=27bea55f9a13aae7f3231bb36ad2c1ceProjected and hidden Markov models for calculating kinetics and metastable states of complex moleculesNoe, Frank; Wu, Hao; Prinz, Jan-Hendrik; Plattner, NuriaJournal of Chemical Physics (2013), 139 (18), 184114/1-184114/17CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Markov state models (MSMs) have been successful in computing metastable states, slow relaxation timescales and assocd. structural changes, and stationary or kinetic exptl. observables of complex mols. from large amts. of mol. dynamics simulation data. However, MSMs approx. the true dynamics by assuming a Markov chain on a clusters discretization of the state space. This approxn. is difficult to make for high-dimensional biomol. systems, and the quality and reproducibility of MSMs has, therefore, been limited. Here, we discard the assumption that dynamics are Markovian on the discrete clusters. Instead, we only assume that the full phase-space mol. dynamics is Markovian, and a projection of this full dynamics is obsd. on the discrete states, leading to the concept of Projected Markov Models (PMMs). Robust estn. methods for PMMs are not yet available, but we derive a practically feasible approxn. via Hidden Markov Models (HMMs). It is shown how various mol. observables of interest that are often computed from MSMs can be computed from HMMs/PMMs. The new framework is applicable to both, simulation and single-mol. exptl. data. We demonstrate its versatility by applications to educative model systems, a 1 ms Anton MD simulation of the bovine pancreatic trypsin inhibitor protein, and an optical tweezer force probe trajectory of an RNA hairpin. (c) 2013 American Institute of Physics.
- 13Suárez, E.; Wiewiora, R. P.; Wehmeyer, C.; Noé, F.; Chodera, J. D.; Zuckerman, D. M. What Markov State Models Can and Cannot Do: Correlation versus Path-Based Observables in Protein-Folding Models. J. Chem. Theory Comput. 2021, 17 (5), 3119– 3133, DOI: 10.1021/acs.jctc.0c01154Google ScholarThere is no corresponding record for this reference.
- 14Dominic, A. J.; Sayer, T.; Cao, S.; Markland, T. E.; Huang, X.; Montoya-Castillo, A. Building Insightful, Memory-Enriched Models to Capture Long-Time Biochemical Processes from Short-Time Simulations. Proc. Natl. Acad. Sci. U. S. A. 2023, 120 (12), e2221048120 DOI: 10.1073/pnas.2221048120Google ScholarThere is no corresponding record for this reference.
- 15Wehmeyer, C.; Noé, F. Time-Lagged Autoencoders: Deep Learning of Slow Collective Variables for Molecular Kinetics. J. Chem. Phys. 2018, 148 (24), 241703, DOI: 10.1063/1.5011399Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslahsro%253D&md5=9e455103c2f05e0a6c0b16f93a9cf074Time-lagged autoencoders: Deep learning of slow collective variables for molecular kineticsWehmeyer, Christoph; Noe, FrankJournal of Chemical Physics (2018), 148 (24), 241703/1-241703/9CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Inspired by the success of deep learning techniques in the phys. and chem. sciences, we apply a modification of an autoencoder type deep neural network to the task of dimension redn. of mol. dynamics data. We can show that our time-lagged autoencoder reliably finds low-dimensional embeddings for high-dimensional feature spaces which capture the slow dynamics of the underlying stochastic processes - beyond the capabilities of linear dimension redn. techniques. (c) 2018 American Institute of Physics.
- 16Mardt, A.; Pasquali, L.; Wu, H.; Noé, F. VAMPnets for Deep Learning of Molecular Kinetics. Nat. Commun. 2018, 9 (1), 5, DOI: 10.1038/s41467-017-02388-1Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MzmvFGqtA%253D%253D&md5=a5871b9d4dc574f431de970e765e4262VAMPnets for deep learning of molecular kineticsMardt Andreas; Pasquali Luca; Wu Hao; Noe FrankNature communications (2018), 9 (1), 5 ISSN:.There is an increasing demand for computing the relevant structures, equilibria, and long-timescale kinetics of biomolecular processes, such as protein-drug binding, from high-throughput molecular dynamics simulations. Current methods employ transformation of simulated coordinates into structural features, dimension reduction, clustering the dimension-reduced data, and estimation of a Markov state model or related model of the interconversion rates between molecular structures. This handcrafted approach demands a substantial amount of modeling expertise, as poor decisions at any step will lead to large modeling errors. Here we employ the variational approach for Markov processes (VAMP) to develop a deep learning framework for molecular kinetics using neural networks, dubbed VAMPnets. A VAMPnet encodes the entire mapping from molecular coordinates to Markov states, thus combining the whole data processing pipeline in a single end-to-end framework. Our method performs equally or better than state-of-the-art Markov modeling methods and provides easily interpretable few-state kinetic models.
- 17Löhr, T.; Kohlhoff, K.; Heller, G. T.; Camilloni, C.; Vendruscolo, M. A Kinetic Ensemble of the Alzheimer’s Aβ Peptide. Nat. Comput. Sci. 2021, 1 (1), 71– 78, DOI: 10.1038/s43588-020-00003-wGoogle ScholarThere is no corresponding record for this reference.
- 18Ghorbani, M.; Prasad, S.; Klauda, J. B.; Brooks, B. R. GraphVAMPNet, using graph neural networks and variational approach to Markov processes for dynamical modeling of biomolecules. J. Chem. Phys. 2022, 156 (18), 184103, DOI: 10.1063/5.0085607Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xht1Oitr%252FL&md5=fd3c7fd1ee3f3af64afa1f53c257eff9GraphVAMPNet, using graph neural networks and variational approach to Markov processes for dynamical modeling of biomoleculesGhorbani, Mahdi; Prasad, Samarjeet; Klauda, Jeffery B.; Brooks, Bernard R.Journal of Chemical Physics (2022), 156 (18), 184103CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Finding a low dimensional representation of data from long-timescale trajectories of biomol. processes, such as protein folding or ligand-receptor binding, is of fundamental importance, and kinetic models, such as Markov modeling, have proven useful in describing the kinetics of these systems. Recently, an unsupervised machine learning technique called VAMPNet was introduced to learn the low dimensional representation and the linear dynamical model in an end-to-end manner. VAMPNet is based on the variational approach for Markov processes and relies on neural networks to learn the coarse-grained dynamics. In this paper, we combine VAMPNet and graph neural networks to generate an end-to-end framework to efficiently learn high-level dynamics and metastable states from the long-timescale mol. dynamics trajectories. This method bears the advantages of graph representation learning and uses graph message passing operations to generate an embedding for each datapoint, which is used in the VAMPNet to generate a coarse-grained dynamical model. This type of mol. representation results in a higher resoln. and a more interpretable Markov model than the std. VAMPNet, enabling a more detailed kinetic study of the biomol. processes. Our GraphVAMPNet approach is also enhanced with an attention mechanism to find the important residues for classification into different metastable states. (c) 2022 American Institute of Physics.
- 19Liu, B.; Xue, M.; Qiu, Y.; Konovalov, K. A.; O’Connor, M. S.; Huang, X. GraphVAMPnets for Uncovering Slow Collective Variables of Self-Assembly Dynamics. J. Chem. Phys. 2023, 159 (9), 094901, DOI: 10.1063/5.0158903Google ScholarThere is no corresponding record for this reference.
- 20Mardt, A.; Hempel, T.; Clementi, C.; Noé, F. Deep Learning to Decompose Macromolecules into Independent Markovian Domains. Nat. Commun. 2022, 13 (1), 7101, DOI: 10.1038/s41467-022-34603-zGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XivFOmsL7N&md5=eee8e2dd74b7a123c9f547e4a4b53eefDeep learning to decompose macromolecules into independent Markovian domainsMardt, Andreas; Hempel, Tim; Clementi, Cecilia; Noe, FrankNature Communications (2022), 13 (1), 7101CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)The increasing interest in modeling the dynamics of ever larger proteins has revealed a fundamental problem with models that describe the mol. system as being in a global configuration state. This notion limits our ability to gather sufficient statistics of state probabilities or state-to-state transitions because for large mol. systems the no. of metastable states grows exponentially with size. In this manuscript, we approach this challenge by introducing a method that combines our recent progress on independent Markov decompn. (IMD) with VAMPnets, a deep learning approach to Markov modeling. We establish a training objective that quantifies how well a given decompn. of the mol. system into independent subdomains with Markovian dynamics approximates the overall dynamics. By constructing an end-to-end learning framework, the decompn. into such subdomains and their individual Markov state models are simultaneously learned, providing a data-efficient and easily interpretable summary of the complex system dynamics. While learning the dynamical coupling between Markovian subdomains is still an open issue, the present results are a significant step towards learning Ising models of large mol. complexes from simulation data.
- 21Chen, W.; Sidky, H.; Ferguson, A. L. Nonlinear Discovery of Slow Molecular Modes Using State-Free Reversible VAMPnets. J. Chem. Phys. 2019, 150 (21), 214114, DOI: 10.1063/1.5092521Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFalurrM&md5=4b423a81430965b8d714eac2daba848bNonlinear discovery of slow molecular modes using state-free reversible VAMPnetsChen, Wei; Sidky, Hythem; Ferguson, Andrew L.Journal of Chemical Physics (2019), 150 (21), 214114/1-214114/16CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The success of enhanced sampling mol. simulations that accelerate along collective variables (CVs) is predicated on the availability of variables coincident with the slow collective motions governing the long-time conformational dynamics of a system. It is challenging to intuit these slow CVs for all but the simplest mol. systems, and their data-driven discovery directly from mol. simulation trajectories has been a central focus of the mol. simulation community to both unveil the important phys. mechanisms and drive enhanced sampling. In this work, we introduce state-free reversible VAMPnets (SRV) as a deep learning architecture that learns nonlinear CV approximants to the leading slow eigenfunctions of the spectral decompn. of the transfer operator that evolves equil.-scaled probability distributions through time. Orthogonality of the learned CVs is naturally imposed within network training without added regularization. The CVs are inherently explicit and differentiable functions of the input coordinates making them well-suited to use in enhanced sampling calcns. We demonstrate the utility of SRVs in capturing parsimonious nonlinear representations of complex system dynamics in applications to 1D and 2D toy systems where the true eigenfunctions are exactly calculable and to mol. dynamics simulations of alanine dipeptide and the WW domain protein. (c) 2019 American Institute of Physics.
- 22Kleiman, D. E.; Shukla, D. Active Learning of the Conformational Ensemble of Proteins Using Maximum Entropy VAMPNets. J. Chem. Theory Comput. 2023, 19 (14), 4377– 4388, DOI: 10.1021/acs.jctc.3c00040Google ScholarThere is no corresponding record for this reference.
- 23Mardt, A.; Noé, F. Progress in Deep Markov State Modeling: Coarse Graining and Experimental Data Restraints. J. Chem. Phys. 2021, 155 (21), 214106, DOI: 10.1063/5.0064668Google ScholarThere is no corresponding record for this reference.
- 24Tolar, M.; Abushakra, S.; Hey, J. A.; Porsteinsson, A.; Sabbagh, M. Aducanumab, Gantenerumab, BAN2401, and ALZ-801─the First Wave of Amyloid-Targeting Drugs for Alzheimer’s Disease with Potential for near Term Approval. Alzheimer’s Res. Ther. 2020, 12 (1), 95, DOI: 10.1186/s13195-020-00663-wGoogle ScholarThere is no corresponding record for this reference.
- 25Gervais, F.; Paquette, J.; Morissette, C.; Krzywkowski, P.; Yu, M.; Azzi, M.; Lacombe, D.; Kong, X.; Aman, A.; Laurin, J. Targeting Soluble Aβ Peptide with Tramiprosate for the Treatment of Brain Amyloidosis. Neurobiol. Aging 2007, 28 (4), 537– 547, DOI: 10.1016/j.neurobiolaging.2006.02.015Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXitFyks74%253D&md5=f871b841546682e832b666dc875a68efTargeting soluble Aβ peptide with Tramiprosate for the treatment of brain amyloidosisGervais, Francine; Paquette, Julie; Morissette, Celine; Krzywkowski, Pascale; Yu, Mathilde; Azzi, Mounia; Lacombe, Diane; Kong, Xianqi; Aman, Ahmed; Laurin, Julie; Szarek, Walter A.; Tremblay, PatrickNeurobiology of Aging (2007), 28 (4), 537-547CODEN: NEAGDO; ISSN:0197-4580. (Elsevier)Amyloid β-peptide (Aβ) is a major constituent of senile plaques in Alzheimer's disease (AD). Neurotoxicity results from the conformational transition of Aβ from random-coil to β-sheet and its oligomerization. Among a series of ionic compds. able to interact with sol. Aβ, Tramiprosate (3-amino-1-propanesulfonic acid; 3APS; Alzhemed) was found to maintain Aβ in a non-fibrillar form, to decrease Aβ42-induced cell death in neuronal cell cultures, and to inhibit amyloid deposition. Tramiprosate crosses the murine blood-brain barrier (BBB) to exert its activity. Treatment of TgCRND8 mice with Tramiprosate resulted in significant redn. (∼30%) in the brain amyloid plaque load and a significant decrease in the cerebral levels of sol. and insol. Aβ40 and Aβ42 (∼20-30%). A dose-dependent redn. (up to 60%) of plasma Aβ levels was also obsd., suggesting that Tramiprosate influences the central pool of Aβ, changing either its efflux or its metab. in the brain. We propose that Tramiprosate, which targets sol. Aβ, represents a new and promising therapeutic class of drugs for the treatment of AD.
- 26Caltagirone, C.; Ferrannini, L.; Marchionni, N.; Nappi, G.; Scapagnini, G.; Trabucchi, M. The Potential Protective Effect of Tramiprosate (Homotaurine) against Alzheimer’s Disease: A Review. Aging: Clin. Exp. Res. 2012, 24 (6), 580– 587, DOI: 10.1007/BF03654836Google ScholarThere is no corresponding record for this reference.
- 27Zou, X.; Himbert, S.; Dujardin, A.; Juhasz, J.; Ros, S.; Stöver, H. D. H.; Rheinstädter, M. C. Curcumin and Homotaurine Suppress Amyloid-Β25–35 Aggregation in Synthetic Brain Membranes. ACS Chem. Neurosci. 2021, 12 (8), 1395– 1405, DOI: 10.1021/acschemneuro.1c00057Google ScholarThere is no corresponding record for this reference.
- 28Abushakra, S.; Porsteinsson, A.; Vellas, B.; Cummings, J.; Gauthier, S.; Hey, J. A.; Power, A.; Hendrix, S.; Wang, P.; Shen, L.; Sampalis, J.; Tolar, M. Clinical Benefits of Tramiprosate in Alzheimer’s Disease Are Associated with Higher Number of APOE4 Alleles: The “APOE4 Gene-Dose Effect. J. Prev. Alzheimers Dis. 2016, 3 (4), 219– 228, DOI: 10.14283/jpad.2016.115Google ScholarThere is no corresponding record for this reference.
- 29Tian, J.; Dang, H.; Wallner, M.; Olsen, R.; Kaufman, D. L. Homotaurine, a Safe Blood-Brain Barrier Permeable GABAA-R-Specific Agonist, Ameliorates Disease in Mouse Models of Multiple Sclerosis. Sci. Rep. 2018, 8 (1), 16555, DOI: 10.1038/s41598-018-32733-3Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvotF2gtw%253D%253D&md5=08d91caa6e786493d66bb024fdd3c482Homotaurine, a safe blood-brain barrier permeable GABAA-R-specific agonist, ameliorates disease in mouse models of multiple sclerosisTian Jide; Dang Hoa; Wallner Martin; Olsen Richard; Kaufman Daniel LScientific reports (2018), 8 (1), 16555 ISSN:.There is a need for treatments that can safely promote regulatory lymphocyte responses. T cells express GABA receptors (GABAA-Rs) and GABA administration can inhibit Th1-mediated processes such as type 1 diabetes and rheumatoid arthritis in mouse models. Whether GABAA-R agonists can also inhibit Th17-driven processes such as experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS), is an open question. GABA does not pass through the blood-brain barrier (BBB) making it ill-suited to inhibit the spreading of autoreactivity within the CNS. Homotaurine is a BBB-permeable amino acid that antagonizes amyloid fibril formation and was found to be safe but ineffective in long-term Alzheimer's disease clinical trials. Homotaurine also acts as GABAA-R agonist with better pharmacokinetics than that of GABA. Working with both monophasic and relapsing-remitting mouse models of EAE, we show that oral administration of homotaurine can (1) enhance CD8(+)CD122(+)PD-1(+) and CD4(+)Foxp3(+) Treg, but not Breg, responses, (2) inhibit autoreactive Th17 and Th1 responses, and (3) effectively ameliorate ongoing disease. These observations demonstrate the potential of BBB-permeable GABAA-R agonists as a new class of treatment to enhance CD8(+) and CD4(+) Treg responses and limit Th17 and Th1-medaited inflammation in the CNS.
- 30Manzano, S.; Agüera, L.; Aguilar, M.; Olazarán, J. A Review on Tramiprosate (Homotaurine) in Alzheimer’s Disease and Other Neurocognitive Disorders. Front. Neurol. 2020, 11, 614, DOI: 10.3389/fneur.2020.00614Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jptlSjtA%253D%253D&md5=9b5ddd9c0751224da9c5ace8c3892ed1A Review on Tramiprosate (Homotaurine) in Alzheimer's Disease and Other Neurocognitive DisordersManzano Sagrario; Aguera Luis; Aguilar Miquel; Olazaran JavierFrontiers in neurology (2020), 11 (), 614 ISSN:1664-2295.Alzheimer's disease (AD) is the most prevalent neurodegenerative condition, especially among elderly people. The presence of cortical β-amyloid deposition, together with tau phosphorylation and intracellular accumulation of neurofibrillary tangles (NFT) is the main neuropathologic criteria for AD diagnosis. Additionally, a role of inflammatory, mitochondrial, and metabolic factors has been suggested. Tramiprosate binds to soluble amyloid, thus inhibiting its aggregation in the brain. It reduced oligomeric and fibrillar (plaque) amyloid, diminished hippocampal atrophy, improved cholinergic transmission, and stabilized cognition in preclinical and clinical studies. In this narrative review, current information on the efficacy and safety of tramiprosate, both in AD and in other neurocognitive disorders, is presented. Possible directions for future studies with tramiprosate are also discussed.
- 31Hey, J. A.; Yu, J. Y.; Versavel, M.; Abushakra, S.; Kocis, P.; Power, A.; Kaplan, P. L.; Amedio, J.; Tolar, M. Clinical Pharmacokinetics and Safety of ALZ-801, a Novel Prodrug of Tramiprosate in Development for the Treatment of Alzheimer’s Disease. Clin. Pharmacokinet. 2018, 57 (3), 315– 333, DOI: 10.1007/s40262-017-0608-3Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslWjurbP&md5=7fa4fd8ac6aa9ec857efbdd52aea7d59Clinical Pharmacokinetics and Safety of ALZ-801, a Novel Prodrug of Tramiprosate in Development for the Treatment of Alzheimer's DiseaseHey, John A.; Yu, Jeremy Y.; Versavel, Mark; Abushakra, Susan; Kocis, Petr; Power, Aidan; Kaplan, Paul L.; Amedio, John; Tolar, MartinClinical Pharmacokinetics (2018), 57 (3), 315-333CODEN: CPKNDH; ISSN:0312-5963. (Springer International Publishing AG)ALZ-801 is an orally available, valine-conjugated prodrug of tramiprosate. Tramiprosate, the active agent, is a small-mol. β-amyloid (Aβ) anti-oligomer and aggregation inhibitor that was evaluated extensively in preclin. and clin. investigations for the treatment of Alzheimer's disease (AD). Tramiprosate has been found to inhibit β-amyloid oligomer formation by a multi-ligand enveloping mechanism of action that stabilizes Aβ42 monomers, resulting in the inhibition of formation of oligomers and subsequent aggregation. Although promising as an AD treatment, tramiprosate exhibited two limiting deficiencies: high intersubject pharmacokinetic (PK) variability likely due to extensive gastrointestinal metab., and mild-to-moderate incidence of nausea and vomiting. To address these, we developed an optimized prodrug, ALZ-801, which retains the favorable efficacy attributes of tramiprosate while improving oral PK variability and gastrointestinal tolerability. In this study, we summarize the phase I bridging program to evaluate the safety, tolerability and PK for ALZ-801 after single and multiple rising dose administration in healthy volunteers. Randomized, placebo-controlled, phase I studies in 127 healthy male and female adult and elderly volunteers included [1] a single ascending dose (SAD) study; [2] a 14-day multiple ascending dose (MAD) study; and [3] a single-dose tablet food-effect study. This program was conducted with both a loose-filled capsule and an immediate-release tablet formulation, under both fasted and fed conditions. Safety and tolerability were assessed, and plasma and urine were collected for liq. chromatog.-mass spectrometry (LC-MS) detn. and non-compartmental PK anal. In addn., we defined the target dose of ALZ-801 that delivers a steady-state plasma area under the curve (AUC) exposure of tramiprosate equiv. to that studied in the tramiprosate phase III study. ALZ-801 was well tolerated and there were no severe or serious adverse events (AEs) or lab. findings. The most common AEs were transient mild nausea and some instances of vomiting, which were not dose-related and showed development of tolerance after continued use. ALZ-801 produced dose-dependent max. plasma concn. (Cmax) and AUC exposures of tramiprosate, which were equiv. to that after oral tramiprosate, but with a substantially reduced intersubject variability and a longer elimination half-life. Administration of ALZ-801 with food markedly reduced the incidence of gastrointestinal symptoms compared with the fasted state, without affecting plasma tramiprosate exposure. An immediate-release tablet formulation of ALZ-801 displayed plasma exposure and low variability similar to the loose-filled capsule. ALZ-801 also showed excellent dose-proportionality without accumulation or decrease in plasma exposure of tramiprosate over 14 days. Based on these data, 265 mg of ALZ-801 twice daily was found to achieve a steady-state AUC exposure of tramiprosate equiv. to 150 mg twice daily of oral tramiprosate in the previous phase III trials. ALZ-801, when administered in capsule and tablet forms, showed excellent oral safety and tolerability in healthy adults and elderly volunteers, with significantly improved PK characteristics over oral tramiprosate. A clin. dose of ALZ-801 (265 mg twice daily) was established that achieves the AUC exposure of 150 mg of tramiprosate twice daily, which showed pos. cognitive and functional improvements in apolipoprotein E4/4 homozygous AD patients. These bridging data support the phase III development of ALZ-801 in patients with AD.
- 32A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study of the Efficacy, Safety and Biomarker Effects of ALZ-801 in Subjects With Early Alzheimer’s Disease and APOE4/4 Genotype, ClinicalTrials.gov ID; Clinical trial registration NCT04770220; https://clinicaltrials.gov/ct2/show/NCT04770220 (accessed 2022–07–21).Google ScholarThere is no corresponding record for this reference.
- 33Kocis, P.; Tolar, M.; Yu, J.; Sinko, W.; Ray, S.; Blennow, K.; Fillit, H.; Hey, J. A. Elucidating the Aβ42 Anti-Aggregation Mechanism of Action of Tramiprosate in Alzheimer’s Disease: Integrating Molecular Analytical Methods, Pharmacokinetic and Clinical Data. CNS Drugs 2017, 31 (6), 495– 509, DOI: 10.1007/s40263-017-0434-zGoogle ScholarThere is no corresponding record for this reference.
- 34Hey, J. A.; Kocis, P.; Hort, J.; Abushakra, S.; Power, A.; Vyhnálek, M.; Yu, J. Y.; Tolar, M. Discovery and Identification of an Endogenous Metabolite of Tramiprosate and Its Prodrug ALZ-801 That Inhibits Beta Amyloid Oligomer Formation in the Human Brain. CNS Drugs 2018, 32 (9), 849– 861, DOI: 10.1007/s40263-018-0554-0Google ScholarThere is no corresponding record for this reference.
- 35Liang, C.; Savinov, S. N.; Fejzo, J.; Eyles, S. J.; Chen, J. Modulation of Amyloid-Β42 Conformation by Small Molecules Through Nonspecific Binding. J. Chem. Theory Comput. 2019, 15 (10), 5169– 5174, DOI: 10.1021/acs.jctc.9b00599Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12jt77L&md5=30ffad7b37865a67ad8ef17affc0df02Modulation of Amyloid-β42 Conformation by Small Molecules Through Nonspecific BindingLiang, Chungwen; Savinov, Sergey N.; Fejzo, Jasna; Eyles, Stephen J.; Chen, JianhanJournal of Chemical Theory and Computation (2019), 15 (10), 5169-5174CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Aggregation of amyloid-β (Aβ) peptides is a crucial step in the progression of Alzheimer's disease (AD). Identifying aggregation inhibitors against AD has been a great challenge. We report an atomistic simulation study of the inhibition mechanism of two small mols., homotaurine and scyllo-inositol, which are AD drug candidates currently under investigation. We show that both small mols. promote a conformational change of the Aβ42 monomer toward a more collapsed phase through a nonspecific binding mechanism. This finding provides atomistic-level insights into designing potential drug candidates for future AD treatments.
- 36Hanwell, M. D.; Curtis, D. E.; Lonie, D. C.; Vandermeersch, T.; Zurek, E.; Hutchison, G. R. Avogadro: An Advanced Semantic Chemical Editor, Visualization, and Analysis Platform. J. Cheminf. 2012, 4 (1), 17, DOI: 10.1186/1758-2946-4-17Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVGksLg%253D&md5=f10400f51db314afa780e99403ca748aAvogadro: an advanced semantic chemical editor, visualization, and analysis platformHanwell, Marcus D.; Curtis, Donald E.; Lonie, David C.; Vandermeersch, Tim; Zurek, Eva; Hutchison, Geoffrey R.Journal of Cheminformatics (2012), 4 (), 17CODEN: JCOHB3; ISSN:1758-2946. (Chemistry Central Ltd.)Background: The Avogadro project has developed an advanced mol. editor and visualizer designed for cross-platform use in computational chem., mol. modeling, bioinformatics, materials science, and related areas. It offers flexible, high quality rendering, and a powerful plugin architecture. Typical uses include building mol. structures, formatting input files, and analyzing output of a wide variety of computational chem. packages. By using the CML file format as its native document type, Avogadro seeks to enhance the semantic accessibility of chem. data types. Results: The work presented here details the Avogadro library, which is a framework providing a code library and application programming interface (API) with three-dimensional visualization capabilities; and has direct applications to research and education in the fields of chem., physics, materials science, and biol. The Avogadro application provides a rich graphical interface using dynamically loaded plugins through the library itself. The application and library can each be extended by implementing a plugin module in C++ or Python to explore different visualization techniques, build/manipulate mol. structures, and interact with other programs. We describe some example extensions, one which uses a genetic algorithm to find stable crystal structures, and one which interfaces with the PackMol program to create packed, solvated structures for mol. dynamics simulations. The 1.0 release series of Avogadro is the main focus of the results discussed here. Conclusions: Avogadro offers a semantic chem. builder and platform for visualization and anal. For users, it offers an easy-to-use builder, integrated support for downloading from common databases such as PubChem and the Protein Data Bank, extg. chem. data from a wide variety of formats, including computational chem. output, and native, semantic support for the CML file format. For developers, it can be easily extended via a powerful plugin mechanism to support new features in org. chem., inorg. complexes, drug design, materials, biomols., and simulations.
- 37Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Scalmani, G.; Barone, V.; Mennucci, B.; Petersson, G. A., Gaussian 09. In Revision E.01, Gaussian, Inc., 2009.Google ScholarThere is no corresponding record for this reference.
- 38Case, D. A.; Betz, R. M.; Cerutti, D. S.; Cheatham, III, T. E.; Darden, T. A.; Duke, R. E.; Giese, T. J.; Gohlke, H.; Goetz, A. W.; Homeyer, N., ; AMBER 16, University of California, San Francisco, 2016.Google ScholarThere is no corresponding record for this reference.
- 39Rose, P. W.; Bi, C.; Bluhm, W. F.; Christie, C. H.; Dimitropoulos, D.; Dutta, S.; Green, R. K.; Goodsell, D. S.; Prlić, A.; Quesada, M. The RCSB Protein Data Bank: New Resources for Research and Education. Nucleic Acids Res. 2012, 41 (D1), D475– D482, DOI: 10.1093/nar/gks1200Google ScholarThere is no corresponding record for this reference.
- 40Bas, D. C.; Rogers, D. M.; Jensen, J. H. Very fast prediction and rationalization of pKa values for protein–ligand complexes. Proteins: Struct., Funct., Bioinf. 2008, 73 (3), 765– 783, DOI: 10.1002/prot.22102Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCgsbjO&md5=34f63cf947000be5482b745675ff8a8aVery fast prediction and rationalization of pKa values for protein-ligand complexesBas, Delphine C.; Rogers, David M.; Jensen, Jan H.Proteins: Structure, Function, and Bioinformatics (2008), 73 (3), 765-783CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The PROPKA method for the prediction of the pKa values of ionizable residues in proteins is extended to include the effect of non-proteinaceous ligands on protein pKa values as well as predict the change in pKa values of ionizable groups on the ligand itself. This new version of PROPKA (PROPKA 2.0) is, as much as possible, developed by adapting the empirical rules underlying PROPKA 1.0 to ligand functional groups. Thus, the speed of PROPKA is retained, so that the pKa values of all ionizable groups are computed in a matter of seconds for most proteins. This adaptation is validated by comparing PROPKA 2.0 predictions to exptl. data for 26 protein-ligand complexes including trypsin, thrombin, three pepsins, HIV-1 protease, chymotrypsin, xylanase, hydroxynitrile lyase, and dihydrofolate reductase. For trypsin and thrombin, large protonation state changes (|n| > 0.5) have been obsd. exptl. for 4 out of 14 ligand complexes. PROPKA 2.0 and Klebe's PEOE approach both identify three of the four large protonation state changes. The protonation state changes due to plasmepsin II, cathepsin D and endothiapepsin binding to pepstatin are predicted to within 0.4 proton units at pH 6.5 and 7.0, resp. The PROPKA 2.0 results indicate that structural changes due to ligand binding contribute significantly to the proton uptake/release, as do residues far away from the binding site, primarily due to the change in the local environment of a particular residue and hence the change in the local hydrogen bonding network. Overall the results suggest that PROPKA 2.0 provides a good description of the protein-ligand interactions that have an important effect on the pKa values of titratable groups, thereby permitting fast and accurate detn. of the protonation states of key residues and ligand functional groups within the binding or active site of a protein.
- 41Doerr, S.; Harvey, M. J.; Noé, F.; De Fabritiis, G. HTMD: High-Throughput Molecular Dynamics for Molecular Discovery. J. Chem. Theory Comput. 2016, 12 (4), 1845– 1852, DOI: 10.1021/acs.jctc.6b00049Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs12hu7o%253D&md5=c7ce0d9a709642ad5e28e202655a9a9dHTMD: High-Throughput Molecular Dynamics for Molecular DiscoveryDoerr, S.; Harvey, M. J.; Noe, Frank; De Fabritiis, G.Journal of Chemical Theory and Computation (2016), 12 (4), 1845-1852CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Recent advances in mol. simulations have allowed scientists to investigate slower biol. processes than ever before. Together with these advances came an explosion of data that has transformed a traditionally computing-bound into a data-bound problem. Here, we present HTMD, a programmable, extensible platform written in Python that aims to solve the data generation and anal. problem as well as increase reproducibility by providing a complete workspace for simulation-based discovery. So far, HTMD includes system building for CHARMM and AMBER force fields, projection methods, clustering, mol. simulation prodn., adaptive sampling, an Amazon cloud interface, Markov state models, and visualization. As a result, a single, short HTMD script can lead from a PDB structure to useful quantities such as relaxation time scales, equil. populations, metastable conformations, and kinetic rates. In this paper, we focus on the adaptive sampling and Markov state modeling features.
- 42Huang, J.; Rauscher, S.; Nawrocki, G.; Ran, T.; Feig, M.; de Groot, B. L.; Grubmüller, H.; MacKerell, A. D. CHARMM36m: An Improved Force Field for Folded and Intrinsically Disordered Proteins. Nat. Methods 2017, 14 (1), 71– 73, DOI: 10.1038/nmeth.4067Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVSiu77I&md5=0aa151fbef2ee0b5e2cfb593c54330c2CHARMM36m: an improved force field for folded and intrinsically disordered proteinsHuang, Jing; Rauscher, Sarah; Nawrocki, Grzegorz; Ran, Ting; Feig, Michael; de Groot, Bert L.; Grubmuller, Helmut; MacKerell, Alexander D. JrNature Methods (2017), 14 (1), 71-73CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The all-atom additive CHARMM36 protein force field is widely used in mol. modeling and simulations. We present its refinement, CHARMM36m (http://mackerell.umaryland.edu/charmm_ff.shtml), with improved accuracy in generating polypeptide backbone conformational ensembles for intrinsically disordered peptides and proteins.
- 43Swails, J.; ParmEd, GitHub, Inc, 2010. https://github.com/ParmEd/ParmEd. (accessed 2018–03–08).Google ScholarThere is no corresponding record for this reference.
- 44Roe, D. R.; Cheatham, T. E. PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. J. Chem. Theory Comput. 2013, 9 (7), 3084– 3095, DOI: 10.1021/ct400341pGoogle Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFehtr8%253D&md5=6f1bee934f13f180bd7e1feb6b78036dPTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory DataRoe, Daniel R.; Cheatham, Thomas E.Journal of Chemical Theory and Computation (2013), 9 (7), 3084-3095CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We describe PTRAJ and its successor CPPTRAJ, two complementary, portable, and freely available computer programs for the anal. and processing of time series of three-dimensional at. positions (i.e., coordinate trajectories) and the data therein derived. Common tools include the ability to manipulate the data to convert among trajectory formats, process groups of trajectories generated with ensemble methods (e.g., replica exchange mol. dynamics), image with periodic boundary conditions, create av. structures, strip subsets of the system, and perform calcns. such as RMS fitting, measuring distances, B-factors, radii of gyration, radial distribution functions, and time correlations, among other actions and analyses. Both the PTRAJ and CPPTRAJ programs and source code are freely available under the GNU General Public License version 3 and are currently distributed within the AmberTools 12 suite of support programs that make up part of the Amber package of computer programs (see http://ambermd.org). This overview describes the general design, features, and history of these two programs, as well as algorithmic improvements and new features available in CPPTRAJ.
- 45Aqvist, J.; Medina, C.; Samuelsson, J. E. A New Method for Predicting Binding Affinity in Computer-Aided Drug Design. Protein Eng. 1994, 7 (3), 385– 391, DOI: 10.1093/protein/7.3.385Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2c3jtlSnsw%253D%253D&md5=c45572201328309d4fc849844d1c3b71A new method for predicting binding affinity in computer-aided drug designAqvist J; Medina C; Samuelsson J EProtein engineering (1994), 7 (3), 385-91 ISSN:0269-2139.A new semi-empirical method for calculating free energies of binding from molecular dynamics (MD) simulations is presented. It is based on standard thermodynamic cycles and on a linear approximation of polar and non-polar free energy contributions from the corresponding MD averages. The method is tested on a set of endothiapepsin inhibitors and found to give accurate results both for absolute as well as relative free energies.
- 46Kabsch, W.; Sander, C. Dictionary of Protein Secondary Structure: Pattern Recognition of Hydrogen-Bonded and Geometrical Features. Biopolymers 1983, 22 (12), 2577– 2637, DOI: 10.1002/bip.360221211Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXkslegtQ%253D%253D&md5=a146e923e6c49cb542d0ad24a399d0f0Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical featuresKabsch, Wolfgang; Sander, ChristianBiopolymers (1983), 22 (12), 2577-637CODEN: BIPMAA; ISSN:0006-3525.For a successful anal. of the relation between amino acid sequence and protein structure, an unambiguous and phys. meaningful definition of secondary structure is essential. A set of simple and phys. motivated criteria for secondary structure, programmed as a pattern-recognition process of H-bonded and geometrical features extd. from x-ray coordinates were developed. Cooperative secondary structure is recognized as repeats of the elementary H-bonding patterns turn and bridge. Repeating turns are helixes, repeating bridges are ladders, connected ladders are sheets. Geometric structure is defined in terms of the concepts torsion and curvature of differential geometry. Local chain chirality is the torsional handedness of 4 consecutive Cα positions and is pos. for right-handed helixes and neg. for ideal twisted β-sheets. Curved pieces are defined as bends. Solvent exposure is given as the no. of H2O mols. in possible contact with a residue. The end result is a compilation of the primary structure, including SS bonds, secondary structure, and solvent exposure of 62 different globular proteins. The presentation is in linear form: strip graphs for an overall view and strip tables for the details of each of 10,925 residues. The dictionary is also available in computer-readable form for protein structure prediction work.
- 47Miller, B. R.; McGee, T. D.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. MMPBSA.Py: An Efficient Program for End-State Free Energy Calculations. J. Chem. Theory Comput. 2012, 8, 3314– 3321, DOI: 10.1021/ct300418hGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 48Genheden, S.; Ryde, U. The MM/PBSA and MM/GBSA Methods to Estimate Ligand-Binding Affinities. Expert Opin. Drug Discovery 2015, 10 (5), 449– 461, DOI: 10.1517/17460441.2015.1032936Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFGktr8%253D&md5=b123b88809f275564f95a2271ebd159fThe MM/PBSA and MM/GBSA methods to estimate ligand-binding affinitiesGenheden, Samuel; Ryde, UlfExpert Opinion on Drug Discovery (2015), 10 (5), 449-461CODEN: EODDBX; ISSN:1746-0441. (Informa Healthcare)Introduction: The mol. mechanics energies combined with the Poisson-Boltzmann or generalized Born and surface area continuum solvation (MM/PBSA and MM/GBSA) methods are popular approaches to est. the free energy of the binding of small ligands to biol. macromols. They are typically based on mol. dynamics simulations of the receptor-ligand complex and are therefore intermediate in both accuracy and computational effort between empirical scoring and strict alchem. perturbation methods. They have been applied to a large no. of systems with varying success. Areas covered: The authors review the use of MM/PBSA and MM/GBSA methods to calc. ligand-binding affinities, with an emphasis on calibration, testing and validation, as well as attempts to improve the methods, rather than on specific applications. Expert opinion: MM/PBSA and MM/GBSA are attractive approaches owing to their modular nature and that they do not require calcns. on a training set. They have been used successfully to reproduce and rationalize exptl. findings and to improve the results of virtual screening and docking. However, they contain several crude and questionable approxns., for example, the lack of conformational entropy and information about the no. and free energy of water mols. in the binding site. Moreover, there are many variants of the method and their performance varies strongly with the tested system. Likewise, most attempts to ameliorate the methods with more accurate approaches, for example, quantum-mech. calcns., polarizable force fields or improved solvation have deteriorated the results.
- 49Wu, H.; Noé, F. Variational Approach for Learning Markov Processes from Time Series Data. J. Nonlinear Sci. 2020, 30 (1), 23– 66, DOI: 10.1007/s00332-019-09567-yGoogle ScholarThere is no corresponding record for this reference.
- 50Mardt, A.; Pasquali, L.; Noé, F.; Wu, H. Deep Learning Markov and Koopman Models with Physical Constraints. In Proceedings of The First Mathematical and Scientific Machine Learning Conference; PMLR, 2020; pp. 451 475.Google ScholarThere is no corresponding record for this reference.
- 51Klambauer, G.; Unterthiner, T.; Mayr, A.; Hochreiter, S.; Self-Normalizing Neural Networks. In Advances in Neural Information Processing Systems, Curran Associates, Inc, 2017, Vol. 30.Google ScholarThere is no corresponding record for this reference.
- 52MacQueen, J. Some Methods for Classification and Analysis of Multivariate Observations Proceedings of the Fifth Berkeley Symposium On Mathematical Statistics And Probability, Volume 1: Statistics Le Cam, L. M.; Neyman, J. Project Euclid 1967, 5; 281 298.Google ScholarThere is no corresponding record for this reference.
- 53Bonneel, N.; van de Panne, M.; Paris, S.; Heidrich, W. Displacement Interpolation Using Lagrangian Mass Transport. ACM Trans. Graph. 2011, 30 (6), 1– 12, DOI: 10.1145/2070781.2024192Google ScholarThere is no corresponding record for this reference.
- 54Kuhn, H. W. The Hungarian Method for the Assignment Problem. Naval Res. Logistics Quarterly 1955, 2 (1–2), 83– 97, DOI: 10.1002/nav.3800020109Google ScholarThere is no corresponding record for this reference.
- 55Fong, R.; Vedaldi, A. Explanations for Attributing Deep Neural Network Predictions. In Explainable AI: Interpreting, Explaining and Visualizing Deep Learning. In Lecture Notes in Computer Science, Samek, W.; Montavon, G.; Vedaldi, A.; Hansen, L. K.; Müller, K.-R.; Springer International Publishing: Cham, 2019; pp. 149 167. DOI: DOI: 10.1007/978-3-030-28954-6_8 .Google ScholarThere is no corresponding record for this reference.
- 56Simonyan, K.; Vedaldi, A.; Zisserman, A. Deep Inside Convolutional Networks: Visualising Image Classification Models and Saliency Maps, arXiv 2014 DOI: 10.48550/arXiv.1312.6034 .Google ScholarThere is no corresponding record for this reference.
- 57Cohen, S. I. A.; Linse, S.; Luheshi, L. M.; Hellstrand, E.; White, D. A.; Rajah, L.; Otzen, D. E.; Vendruscolo, M.; Dobson, C. M.; Knowles, T. P. J. Proliferation of Amyloid-Β42 Aggregates Occurs through a Secondary Nucleation Mechanism. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (24), 9758– 9763, DOI: 10.1073/pnas.1218402110Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFOrt7fJ&md5=d9db3cfc7e3004e5cdc309a92d2c7431Proliferation of amyloid-β42 aggregates occurs through a secondary nucleation mechanismCohen, Samuel I. A.; Linse, Sara; Luheshi, Leila M.; Hellstrand, Erik; White, Duncan A.; Rajah, Luke; Otzen, Daniel E.; Vendruscolo, Michele; Dobson, Christopher M.; Knowles, Tuomas P. J.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (24), 9758-9763, S9758/1-S9758/11CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The generation of toxic oligomers during the aggregation of the amyloid-β (Aβ) peptide Aβ42 into amyloid fibrils and plaques has emerged as a central feature of the onset and progression of Alzheimer's disease, but the mol. pathways that control pathol. aggregation have proved challenging to identify. Here, the authors used a combination of kinetic studies, selective radiolabeling expts., and cell viability assays to detect directly the rates of formation of both fibrils and oligomers and the resulting cytotoxic effects. The results showed that once a small but crit. concn. of amyloid fibrils had accumulated, the toxic oligomeric species were predominantly formed from monomeric peptide mols. through a fibril-catalyzed secondary nucleation reaction, rather than through a classical mechanism of homogeneous primary nucleation. This catalytic mechanism coupled together the growth of insol. amyloid fibrils and the generation of diffusible oligomeric aggregates that are implicated as neurotoxic agents in Alzheimer's disease. These results revealed that the aggregation of Aβ42 is promoted by a pos. feedback loop that originates from the interactions between the monomeric and fibrillar forms of this peptide. These findings bring together the main mol. species implicated in the Aβ aggregation cascade and suggest that perturbation of the secondary nucleation pathway identified in this study could be an effective strategy to control the proliferation of neurotoxic Aβ42 oligomers.
- 58Arosio, P.; Knowles, T. P. J.; Linse, S. On the Lag Phase in Amyloid Fibril Formation. Phys. Chem. Chem. Phys. 2015, 17 (12), 7606– 7618, DOI: 10.1039/C4CP05563BGoogle Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCrsbo%253D&md5=552e3a6a573bf26a0c125cb4bdb2fc19On the lag phase in amyloid fibril formationArosio, Paolo; Knowles, Tuomas P. J.; Linse, SaraPhysical Chemistry Chemical Physics (2015), 17 (12), 7606-7618CODEN: PPCPFQ; ISSN:1463-9076. (Royal Society of Chemistry)A review. The formation of nanoscale amyloid fibrils from normally sol. peptides and proteins is a common form of self-assembly phenomenon that has fundamental connections with biol. functions and human diseases. The kinetics of this process has been widely studied and exhibits on a macroscopic level three characteristic stages: (1) a lag phase; (2) a growth phase; and (3) a final plateau regime. The question of which mol. events take place during each one of these phases has been a central element in the quest for a mechanism of amyloid formation. Here, the authors discuss the nature and mol. origin of the lag-phase in amyloid formation by making use of tools and concepts from phys. chem., in particular from chem. reaction kinetics. The authors discuss how, in macroscopic samples, it has become apparent that the lag-phase is not a waiting time for nuclei to form. Rather, multiple parallel processes exist and typically millions of primary nuclei form during the lag phase from monomers in soln. Thus, the lag-time represents a time that is required for the nuclei that are formed early on in the reaction to grow and proliferate in order to reach an aggregate concn. that is readily detected in bulk assays. In many cases, this proliferation takes place through secondary nucleation, where fibrils may present a catalytic surface for the formation of new aggregates. Fibrils may also break (fragmentation) and thereby provide new ends for elongation. Thus, at least 2 (primary nucleation and elongation) and in many systems at least 4 (primary nucleation, elongation, secondary nucleation, and fragmentation) microscopic processes occur during the lag phase. Moreover, these same processes occur during all 3 phases of the macroscopic aggregation process, albeit at different rates as governed by rate consts. and by the concn. of reacting species at each point in time.
- 59Tomaselli, S.; Esposito, V.; Vangone, P.; van Nuland, N. A. J.; Bonvin, A. M. J. J.; Guerrini, R.; Tancredi, T.; Temussi, P. A.; Picone, D. The α-to-β Conformational Transition of Alzheimer’s Aβ-(1–42) Peptide in Aqueous Media Is Reversible: A Step by Step Conformational Analysis Suggests the Location of β Conformation Seeding. ChemBiochem 2006, 7 (2), 257– 267, DOI: 10.1002/cbic.200500223Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhs1aktrc%253D&md5=aa0da86f9a3b30ff357490ae46dff8d8The α-to-β conformational transition of Alzheimer's Aβ-(1-42) peptide in aqueous media is reversible: a step by step conformational analysis suggests the location of β conformation seedingTomaselli, Simona; Esposito, Veronica; Vangone, Paolo; van Nuland, Nico A. J.; Bonvin, Alexandre M. J. J.; Guerrini, Remo; Tancredi, Teodorico; Temussi, Piero A.; Picone, DeliaChemBioChem (2006), 7 (2), 257-267CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Current views of the role of β-amyloid (Aβ) peptide fibrils range from regarding them as the cause of Alzheimer's pathol. to having a protective function. In the last few years, it has also been suggested that sol. oligomers might be the most important toxic species. In all cases, the study of the conformational properties of Aβ peptides in sol. form constitutes a basic approach to the design of mols. with "antiamyloid" activity. We exptl. investigated the conformational 'path' that can lead the Aβ-(1-42) peptide from the native state, which is represented by an α helix embedded in the membrane, to the final state in the amyloid fibrils, which is characterized by sheet structures. The conformational steps were monitored by using CD and NMR spectroscopy in media of varying polarities. This was achieved by changing the compn. of water and hexafluoroisopropanol (HFIP). In the presence of HFIP, β conformations can be obsd. in solns. that have very high water content (up to 99% water; vol./vol.). These can be turned back to α helixes simply by adding the appropriated amt. of HFIP. The transition of Aβ-(1-42) from α to β conformation occurs when the amt. of water is higher that 80% (vol./vol.). The NMR structure solved in HFIP/H2O with high water content showed that, on going from very apolar to polar environments, the long N-terminal helix is essentially retained, whereas the shorter C-terminal helix is lost. The complete conformational path was investigated in detail with the aid of mol. dynamics simulations in explicit solvent, which led to the localization of residues that might seed β conformations. The structures obtained might help to find regions that are more affected by environmental conditions in vivo. This could in turn aid the design of mols. able to inhibit fibril deposition or revert oligomerization processes.
- 60Shamsi, Z.; Moffett, A. S.; Shukla, D. Enhanced Unbiased Sampling of Protein Dynamics Using Evolutionary Coupling Information. Sci. Rep. 2017, 7 (1), 12700, DOI: 10.1038/s41598-017-12874-7Google ScholarThere is no corresponding record for this reference.
- 61Zimmerman, M. I.; Porter, J. R.; Sun, X.; Silva, R. R.; Bowman, G. R. Choice of Adaptive Sampling Strategy Impacts State Discovery, Transition Probabilities, and the Apparent Mechanism of Conformational Changes. J. Chem. Theory Comput. 2018, 14 (11), 5459– 5475, DOI: 10.1021/acs.jctc.8b00500Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslGqsLnI&md5=b770c1d634b1d7fc7cc8e0b9fb3b3db8Choice of Adaptive Sampling Strategy Impacts State Discovery, Transition Probabilities, and the Apparent Mechanism of Conformational ChangesZimmerman, Maxwell I.; Porter, Justin R.; Sun, Xianqiang; Silva, Roseane R.; Bowman, Gregory R.Journal of Chemical Theory and Computation (2018), 14 (11), 5459-5475CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Interest in atomically detailed simulations has grown significantly with recent advances in computational hardware and Markov state modeling (MSM) methods, yet outstanding questions remain that hinder their widespread adoption. Namely, how do alternative sampling strategies explore conformational space and how might this influence predictions generated from the data. Here, we seek to answer these questions for four commonly used sampling methods: (1) a single long simulation, (2) many short simulations run in parallel, (3) adaptive sampling, and (4) our recently developed goal-oriented sampling algorithm, FAST. We first develop a theor. framework for anal. calcg. the probability of discovering select states on simple landscapes, where we uncover the drastic effects of varying the no. and length of simulations. We then use kinetic Monte Carlo simulations on a variety of phys. inspired landscapes to characterize the probability of discovering particular states and transition pathways for each of the four methods. Consistently, we find that FAST simulations discover each target state with the highest probability, while traversing realistic pathways. Furthermore, we uncover the potential pathol. that short parallel simulations sometimes predict an incorrect transition pathway by crossing large energy barriers that long simulations would typically circumnavigate. We refer to this pathol. as "pathway tunneling". To protect against this phenomenon when using adaptive-sampling and FAST simulations, we introduce the FAST-string method. This method enhances sampling along the highest-flux transition paths to refine an MSMs transition probabilities and discriminate between competing pathways. Addnl., we compare the performance of a variety of MSM estimators in describing accurate thermodn. and kinetics. For adaptive sampling, we recommend simply normalizing the transition counts out of each state after adding small pseudocounts to avoid creating sources or sinks. Lastly, we evaluate whether our insights from simple landscapes hold for all-atom mol. dynamics simulations of the folding of the λ-repressor protein. Remarkably, we find that FAST-contacts predicts the same folding pathway as a set of long simulations but with orders of magnitude less simulation time.
- 62Betz, R. M.; Dror, R. O. How Effectively Can Adaptive Sampling Methods Capture Spontaneous Ligand Binding?. J. Chem. Theory Comput. 2019, 15 (3), 2053– 2063, DOI: 10.1021/acs.jctc.8b00913Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1agtrY%253D&md5=36f1ee6880504ea87266dce4a3402169How Effectively Can Adaptive Sampling Methods Capture Spontaneous Ligand Binding?Betz, Robin M.; Dror, Ron O.Journal of Chemical Theory and Computation (2019), 15 (3), 2053-2063CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. dynamics (MD) simulations that capture the spontaneous binding of drugs and other ligands to their target proteins can reveal a great deal of useful information, but most drug-like ligands bind on time scales longer than those accessible to individual MD simulations. Adaptive sampling methods-in which one performs multiple rounds of simulation, with the initial conditions of each round based on the results of previous rounds-offer a promising potential soln. to this problem. No comprehensive anal. of the performance gains from adaptive sampling is available for ligand binding, however, particularly for protein-ligand systems typical of those encountered in drug discovery. Moreover, most previous work presupposes knowledge of the ligand's bound pose. Here the authors outline existing methods for adaptive sampling of the ligand-binding process and introduce several improvements, with a focus on methods that do not require prior knowledge of the binding site or bound pose. The authors then evaluate these methods by comparing them to traditional, long MD simulations for realistic protein-ligand systems. The authors find that adaptive sampling simulations typically fail to reach the bound pose more efficiently than traditional MD. However, adaptive sampling identifies multiple potential binding sites more efficiently than traditional MD and also provides better characterization of binding pathways. The authors explain these results by showing that protein-ligand binding is an example of an exploration-exploitation dilemma. Existing adaptive sampling methods for ligand binding in the absence of a known bound pose vastly favor the broad exploration of protein-ligand space, sometimes failing to sufficiently exploit intermediate states as they are discovered. The authors suggest potential avenues for future research to address this shortcoming.
- 63Kleiman, D. E.; Nadeem, H.; Shukla, D. Adaptive Sampling Methods for Molecular Dynamics in the Era of Machine Learning. J. Phys. Chem. B 2023, 127 (50), 10669– 10681, DOI: 10.1021/acs.jpcb.3c04843Google ScholarThere is no corresponding record for this reference.
- 64Man, V. H.; He, X.; Derreumaux, P.; Ji, B.; Xie, X.-Q.; Nguyen, P. H.; Wang, J. Effects of All-Atom Molecular Mechanics Force Fields on Amyloid Peptide Assembly: The Case of Aβ16–22 Dimer. J. Chem. Theory Comput. 2019, 15 (2), 1440– 1452, DOI: 10.1021/acs.jctc.8b01107Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlaqsA%253D%253D&md5=b7d65b1c4eeb166b27ab435687bb2528Effects of all-atom molecular mechanics force fields on amyloid peptide assembly: The case of Aβ16-22 dimerMan, Viet Hoang; He, Xibing; Derreumaux, Philippe; Ji, Beihong; Xie, Xiang-Qun; Nguyen, Phuong H.; Wang, JunmeiJournal of Chemical Theory and Computation (2019), 15 (2), 1440-1452CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We investigated the effects of 17 widely used atomistic mol. mechanics force fields (MMFFs) on the structures and kinetics of amyloid peptide assembly. To this end, we performed large-scale all-atom mol. dynamics simulations in explicit water on the dimer of the 7-residue fragment of Alzheimer amyloid-β peptide, Aβ16-22, for a total time of 0.34 ms. We compared the effects of these MMFFs by analyzing various global reaction coordinates, secondary structure contents, the fibril population, the in-register and out-of-register architectures, and the fibril formation time at 310 K. While the AMBER94, AMBER99, and AMBER12SB force fields did not predict any β-sheets, the 7 force fields (AMBER96, GROMOS45a3, GROMOS53a5, GROMOS53a6, GROMOS43a1, GROMOS43a2, and GROMOS54a7) formed β-sheets rapidly. In contrast, the following 5 force fields (AMBER99-ILDN, AMBER14SB, CHARMM22*, CHARMM36, and CHARMM36m) were the best candidates for studying amyloid peptide assembly, as they provided good balances in terms of structures and kinetics. We also investigated the assembly mechanisms of dimeric Aβ16-22 and found that the fibril formation rate was predominantly controlled by the total β-strand content.
- 65Maier, J. A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K. E.; Simmerling, C. ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB. J. Chem. Theory Comput. 2015, 11 (8), 3696– 3713, DOI: 10.1021/acs.jctc.5b00255Google Scholar65https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFequ7rN&md5=7b803577b3b6912cc6750cfbd356596eff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SBMaier, James A.; Martinez, Carmenza; Kasavajhala, Koushik; Wickstrom, Lauren; Hauser, Kevin E.; Simmerling, CarlosJournal of Chemical Theory and Computation (2015), 11 (8), 3696-3713CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. mechanics is powerful for its speed in atomistic simulations, but an accurate force field is required. The Amber ff99SB force field improved protein secondary structure balance and dynamics from earlier force fields like ff99, but weaknesses in side chain rotamer and backbone secondary structure preferences have been identified. Here, we performed a complete refit of all amino acid side chain dihedral parameters, which had been carried over from ff94. The training set of conformations included multidimensional dihedral scans designed to improve transferability of the parameters. Improvement in all amino acids was obtained as compared to ff99SB. Parameters were also generated for alternate protonation states of ionizable side chains. Av. errors in relative energies of pairs of conformations were under 1.0 kcal/mol as compared to QM, reduced 35% from ff99SB. We also took the opportunity to make empirical adjustments to the protein backbone dihedral parameters as compared to ff99SB. Multiple small adjustments of φ and ψ parameters were tested against NMR scalar coupling data and secondary structure content for short peptides. The best results were obtained from a phys. motivated adjustment to the φ rotational profile that compensates for lack of ff99SB QM training data in the β-ppII transition region. Together, these backbone and side chain modifications (hereafter called ff14SB) not only better reproduced their benchmarks, but also improved secondary structure content in small peptides and reprodn. of NMR χ1 scalar coupling measurements for proteins in soln. We also discuss the Amber ff12SB parameter set, a preliminary version of ff14SB that includes most of its improvements.
- 66Harada, T.; Kuroda, R. CD Measurements of β-Amyloid (1–40) and (1–42) in the Condensed Phase. Biopolymers 2011, 95 (2), 127– 134, DOI: 10.1002/bip.21543Google ScholarThere is no corresponding record for this reference.
- 67Löhr, T.; Kohlhoff, K.; Heller, G. T.; Camilloni, C.; Vendruscolo, M. A Small Molecule Stabilizes the Disordered Native State of the Alzheimer’s Aβ Peptide. ACS Chem. Neurosci. 2022, 13 (12), 1738– 1745, DOI: 10.1021/acschemneuro.2c00116Google Scholar67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsVSksbnM&md5=6b68e3699c6b553694d09f306c68da96A Small Molecule Stabilizes the Disordered Native State of the Alzheimer's Aβ PeptideLohr, Thomas; Kohlhoff, Kai; Heller, Gabriella T.; Camilloni, Carlo; Vendruscolo, MicheleACS Chemical Neuroscience (2022), 13 (12), 1738-1745CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The stabilization of native states of proteins is a powerful drug discovery strategy. It is still unclear, however, whether this approach can be applied to intrinsically disordered proteins. Here, the authors report a small mol. that stabilizes the native state of the Aβ42 peptide, an intrinsically disordered protein fragment assocd. with Alzheimer's disease. This stabilization takes place by a dynamic binding mechanism, in which both the small mol. and the Aβ42 peptide remain disordered. This disordered binding mechanism involves enthalpically favorable local -stacking interactions coupled with entropically advantageous global effects. Small mols. can stabilize disordered proteins in their native states through transient non-specific interactions that provide enthalpic gain while simultaneously increasing the conformational entropy of the proteins.
- 68Reddy, G.; Straub, J. E.; Thirumalai, D. Influence of Preformed Asp23-Lys28 Salt Bridge on the Conformational Fluctuations of Monomers and Dimers of Aβ Peptides with Implications for Rates of Fibril Formation. J. Phys. Chem. B 2009, 113 (4), 1162– 1172, DOI: 10.1021/jp808914cGoogle Scholar68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXisVWqtA%253D%253D&md5=9da47509c0dc01142cad5d009c9040bfInfluence of Preformed Asp23-Lys28 Salt Bridge on the Conformational Fluctuations of Monomers and Dimers of Aβ Peptides with Implications for Rates of Fibril FormationReddy, Govardhan; Straub, John E.; Thirumalai, D.Journal of Physical Chemistry B (2009), 113 (4), 1162-1172CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Recent expts. have shown that the congener Aβ1-40[D23-K28], in which the side chains of charged residues Asp23 and Lys28 are linked by a lactam bridge, forms amyloid fibrils that are structurally similar to the wild type (WT) Aβ peptide, but at a rate that is nearly 1000 times faster. We used all atom mol. dynamics simulations in explicit water, and two force fields, of the WT dimer, a monomer with the lactam bridge (Aβ10-35-lactam[D23-K28]), and the monomer and dimers with harmonically constrained D23-K28 salt bridge (Aβ10-35[D23-K28]) to understand the origin of the enhanced fibril rate formation. The simulations show that the assembly-competent fibril-like monomer (N*) structure, which is present among the conformations sampled by the isolated monomer, with strand conformations in the residues spanning the N and C termini and a bend involving residues D23VGSNKG29, are populated to a much greater extent in Aβ10-35[D23-K28] and Aβ10-35-lactam[D23-K28] than in the WT, which has negligible probability of forming N*. The salt bridge in N* of Aβ10-35[D23-K28], whose topol. is similar to that found in the fibril, is hydrated. The redn. in the free energy barrier to fibril formation in Aβ10-35[D23-K28] and in Aβ10-35-lactam[D23-K28], compared to the WT, arises largely due to entropic restriction which enables the bend formation. A decrease in the entropy of the unfolded state and the lesser penalty for conformational rearrangement including the formation of the salt bridge in Aβ peptides with D23-K28 constraint results in a redn. in the kinetic barrier in the Aβ1-40-lactam[D23-K28] congener compared to the WT. The decrease in the barrier, which is related to the free energy cost of forming a bend, is estd. to be in the range (4-7)kBT. Although a no. of factors det. the growth of fibrils, the decrease in the free energy barrier, relative to the WT, to N* formation is a major factor in the rate enhancement in the fibril formation of Aβ1-40[D23-K28] congener. Qual. similar results were obtained using simulations of Aβ9-40 peptides and various constructs related to the Aβ10-35 systems that were probed using OPLS and CHARMM force fields. We hypothesize that mutations or other constraints that preferentially enhance the population of the N* species would speed up aggregation rates. Conversely, ligands that lock it in the fibril-like N* structure would prevent amyloid formation.
- 69Chandra, B.; Bhowmik, D.; Maity, B. K.; Mote, K. R.; Dhara, D.; Venkatramani, R.; Maiti, S.; Madhu, P. K. Major Reaction Coordinates Linking Transient Amyloid-β Oligomers to Fibrils Measured at Atomic Level. Biophys. J. 2017, 113 (4), 805– 816, DOI: 10.1016/j.bpj.2017.06.068Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Cku7vN&md5=52799f167c7d9685fa3e01234858383eMajor reaction coordinates linking transient amyloid-β oligomers to fibrils measured at atomic levelChandra, Bappaditya; Bhowmik, Debanjan; Maity, Barun Kumar; Mote, Kaustubh R.; Dhara, Debabrata; Venkatramani, Ravindra; Maiti, Sudipta; Madhu, Perunthiruthy K.Biophysical Journal (2017), 113 (4), 805-816CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The structural underpinnings for the higher toxicity of the oligomeric intermediates of amyloidogenic peptides, compared to the mature fibrils, remain unknown at present. The transient nature and heterogeneity of the oligomers make it difficult to follow their structure. Here, using vibrational and solid-state NMR spectroscopy, and mol. dynamics simulations, we show that freely aggregating Aβ40 oligomers in physiol. solns. have an intramol. antiparallel configuration that is distinct from the intermol. parallel β-sheet structure obsd. in mature fibrils. The intramol. hydrogen-bonding network flips nearly 90°, and the two β-strands of each monomeric unit move apart, to give rise to the well-known intermol. in-register parallel β-sheet structure in the mature fibrils. Solid-state NMR distance measurements capture the interstrand sepn. within monomer units during the transition from the oligomer to the fibril form. We further find that the D23-K28 salt-bridge, a major feature of the Aβ40 fibrils and a focal point of mutations linked to early onset Alzheimer's disease, is not detectable in the small oligomers. Mol. dynamics simulations capture the correlation between changes in the D23-K28 distance and the flipping of the monomer secondary structure between antiparallel and parallel β-sheet architectures. Overall, we propose interstrand sepn. and salt-bridge formation as key reaction coordinates describing the structural transition of the small Aβ40 oligomers to fibrils.
- 70Nemergut, M.; Marques, S. M.; Uhrik, L.; Vanova, T.; Nezvedova, M.; Gadara, D. C.; Jha, D.; Tulis, J.; Novakova, V.; Planas-Iglesias, J. Domino-like Effect of C112R Mutation on ApoE4 Aggregation and Its Reduction by Alzheimer’s Disease Drug Candidate. Mol. Neurodegener. 2023, 18 (1), 38, DOI: 10.1186/s13024-023-00620-9Google Scholar70https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXht1SitbjL&md5=acdfc26c7af26791accfaec4f3a3c83eDomino-like effect of C112R mutation on ApoE4 aggregation and its reduction by Alzheimer's Disease drug candidateNemergut, Michal; Marques, Sergio M.; Uhrik, Lukas; Vanova, Tereza; Nezvedova, Marketa; Gadara, Darshak Chandulal; Jha, Durga; Tulis, Jan; Novakova, Veronika; Planas-Iglesias, Joan; Kunka, Antonin; Legrand, Anthony; Hribkova, Hana; Pospisilova, Veronika; Sedmik, Jiri; Raska, Jan; Prokop, Zbynek; Damborsky, Jiri; Bohaciakova, Dasa; Spacil, Zdenek; Hernychova, Lenka; Bednar, David; Marek, MartinMolecular Neurodegeneration (2023), 18 (1), 38CODEN: MNOEAZ; ISSN:1750-1326. (BioMed Central Ltd.)Apolipoprotein E (ApoE) ε4 genotype is the most prevalent risk factor for late-onset Alzheimers Disease (AD). Although ApoE4 differs from its non-pathol. ApoE3 isoform only by the C112R mutation, the mol. mechanism of its proteinopathy is unknown. Here, we reveal the mol. mechanism of ApoE4 aggregation using a combination of exptl. and computational techniques, including X-ray crystallog., site-directed mutagenesis, hydrogen-deuterium mass spectrometry (HDX-MS), static light scattering and mol. dynamics simulations. Treatment of ApoE ε3/ε3 and ε4/ε4 cerebral organoids with tramiprosate was used to compare the effect of tramiprosate on ApoE4 aggregation at the cellular level. We found that C112R substitution in ApoE4 induces long-distance (> 15 A) conformational changes leading to the formation of a V-shaped dimeric unit that is geometrically different and more aggregation-prone than the ApoE3 structure. AD drug candidate tramiprosate and its metabolite 3-sulfopropanoic acid induce ApoE3-like conformational behavior in ApoE4 and reduce its aggregation propensity. Anal. of ApoE ε4/ε4 cerebral organoids treated with tramiprosate revealed its effect on cholesteryl esters, the storage products of excess cholesterol. Our results connect the ApoE4 structure with its aggregation propensity, providing a new druggable target for neurodegeneration and ageing.
- 71Walsh, D. M.; Thulin, E.; Minogue, A. M.; Gustavsson, N.; Pang, E.; Teplow, D. B.; Linse, S. A Facile Method for Expression and Purification of the Alzheimer’s Disease-Associated Amyloid Beta-Peptide. FEBS J. 2009, 276 (5), 1266– 1281, DOI: 10.1111/j.1742-4658.2008.06862.xGoogle Scholar71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXivFensbo%253D&md5=f644980c816e4b65d8920ed8aee05d0aA facile method for expression and purification of the Alzheimer's disease-associated amyloid β-peptideWalsh, Dominic M.; Thulin, Eva; Minogue, Aedin M.; Gustavsson, Niklas; Pang, Eric; Teplow, David B.; Linse, SaraFEBS Journal (2009), 276 (5), 1266-1281CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)The authors report the development of a high-level bacterial expression system for the Alzheimer's disease-assocd. amyloid β-peptide (Aβ), together with a scalable and inexpensive purifn. procedure. Aβ(1-40) and Aβ(1-42) coding sequences together with added ATG codons were cloned directly into a Pet vector to facilitate prodn. of Met-Aβ(1-40) and Met-Aβ(1-42), referred to as Aβ(M1-40) and Aβ(M1-42), resp. The expression sequences were designed using codons preferred by Escherichia coli, and the two peptides were expressed in this host in inclusion bodies. Peptides were purified from inclusion bodies using a combination of anion-exchange chromatog. and centrifugal filtration. The method described requires little specialized equipment and provides a facile and inexpensive procedure for prodn. of large amts. of very pure Aβ peptides. Recombinant peptides generated using this protocol produced amyloid fibrils that were indistinguishable from those formed by chem. synthesized Aβ1-40 and Aβ1-42. Formation of fibrils by all peptides was concn.-dependent, and exhibited kinetics typical of a nucleation-dependent polymn. reaction. Recombinant and synthetic peptides exhibited a similar toxic effect on hippocampal neurons, with acute treatment causing inhibition of MTT redn., and chronic treatment resulting in neuritic degeneration and cell loss.
- 72Thacker, D.; Sanagavarapu, K.; Frohm, B.; Meisl, G.; Knowles, T. P. J.; Linse, S. The Role of Fibril Structure and Surface Hydrophobicity in Secondary Nucleation of Amyloid Fibrils. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (41), 25272– 25283, DOI: 10.1073/pnas.2002956117Google Scholar72https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVKkt7jO&md5=58081f0fe2ca0ad466ac4491c9b8f627The role of fibril structure and surface hydrophobicity in secondary nucleation of amyloid fibrilsThacker, Dev; Sanagavarapu, Kalyani; Frohm, Birgitta; Meisl, Georg; Knowles, Tuomas P. J.; Linse, SaraProceedings of the National Academy of Sciences of the United States of America (2020), 117 (41), 25272-25283CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Crystals, nanoparticles, and fibrils catalyze the generation of new aggregates on their surface from the same type of monomeric building blocks as the parent assemblies. This secondary nucleation process can be many orders of magnitude faster than primary nucleation. In the case of amyloid fibrils assocd. with Alzheimer's disease, this process leads to the multiplication and propagation of aggregates, whereby short-lived oligomeric intermediates cause neurotoxicity. Understanding the catalytic activity is a fundamental goal in elucidating the mol. mechanisms of Alzheimer's and assocd. diseases. Here the authors explore the role of fibril structure and hydrophobicity by asking whether the V18, A21, V40, and A42 side chains which are exposed on the Aβ42 fibril surface as continuous hydrophobic patches play a role in secondary nucleation. Single, double, and quadruple serine substitutions were made. Kinetic analyses of aggregation data at multiple monomer concns. reveal that all seven mutants retain the dominance of secondary nucleation as the main mechanism of fibril proliferation. This finding highlights the generality of secondary nucleation and its independence of the detailed mol. structure. Cryo-electron micrographs reveal that the V18S substitution causes fibrils to adopt a distinct morphol. with longer twist distance than variants lacking this substitution. Self- and cross-seeding data show that surface catalysis is only efficient between peptides of identical morphol., indicating a templating role of secondary nucleation with structural conversion at the fibril surface. The authors' findings thus provide clear evidence that the propagation of amyloid fibril strains is possible even in systems dominated by secondary nucleation rather than fragmentation.
- 73Yang, H.; Yang, S.; Kong, J.; Dong, A.; Yu, S. Obtaining Information about Protein Secondary Structures in Aqueous Solution Using Fourier Transform IR Spectroscopy. Nat. Protoc. 2015, 10 (3), 382– 396, DOI: 10.1038/nprot.2015.024Google Scholar73https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXit1artbw%253D&md5=690ba9ad6159923b076c27f09822cf4cObtaining information about protein secondary structures in aqueous solution using Fourier transform IR spectroscopyYang, Huayan; Yang, Shouning; Kong, Jilie; Dong, Aichun; Yu, ShaoningNature Protocols (2015), 10 (3), 382-396CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)Fourier transform IR (FTIR) spectroscopy is a nondestructive technique for structural characterization of proteins and polypeptides. The IR spectral data of polymers are usually interpreted in terms of the vibrations of a structural repeat. The repeat units in proteins give rise to nine characteristic IR absorption bands (amides A, B and I-VII). Amide I bands (1,700-1,600 cm-1) are the most prominent and sensitive vibrational bands of the protein backbone, and they relate to protein secondary structural components. In this protocol, we have detailed the principles that underlie the detn. of protein secondary structure by FTIR spectroscopy, as well as the basic steps involved in protein sample prepn., instrument operation, FTIR spectra collection and spectra anal. in order to est. protein secondary-structural components in aq. (both H2O and deuterium oxide (D2O)) soln. using algorithms, such as second-deriv., deconvolution and curve fitting. Small amts. of high-purity (>95%) proteins at high concns. (>3 mg ml-1) are needed in this protocol; typically, the procedure can be completed in 1-2 d.
- 74Hafsa, N. E.; Arndt, D.; Wishart, D. S. CSI 3.0: A Web Server for Identifying Secondary and Super-Secondary Structure in Proteins Using NMR Chemical Shifts. Nucleic Acids Res. 2015, 43 (W1), W370– W377, DOI: 10.1093/nar/gkv494Google ScholarThere is no corresponding record for this reference.
- 75Borcherds, W. M.; Daughdrill, G. W. Using NMR Chemical Shifts to Determine Residue-Specific Secondary Structure Populations for Intrinsically Disordered Proteins. Methods Enzymol. 2018, 611, 101– 136, DOI: 10.1016/bs.mie.2018.09.011Google ScholarThere is no corresponding record for this reference.
- 76Schumann, F. H.; Riepl, H.; Maurer, T.; Gronwald, W.; Neidig, K.-P.; Kalbitzer, H. R. Combined Chemical Shift Changes and Amino Acid Specific Chemical Shift Mapping of Protein–Protein Interactions. J. Biomol. NMR 2007, 39 (4), 275– 289, DOI: 10.1007/s10858-007-9197-zGoogle ScholarThere is no corresponding record for this reference.
- 77Heller, G. T.; Aprile, F. A.; Michaels, T. C. T.; Limbocker, R.; Perni, M.; Ruggeri, F. S.; Mannini, B.; Löhr, T.; Bonomi, M.; Camilloni, C. Small-Molecule Sequestration of Amyloid-β as a Drug Discovery Strategy for Alzheimer’s Disease. Sci. Adv. 2020, 6 (45), eabb5924 DOI: 10.1126/sciadv.abb5924Google ScholarThere is no corresponding record for this reference.
- 78Habchi, J.; Arosio, P.; Perni, M.; Costa, A. R.; Yagi-Utsumi, M.; Joshi, P.; Chia, S.; Cohen, S. I. A.; Müller, M. B. D.; Linse, S. An Anticancer Drug Suppresses the Primary Nucleation Reaction That Initiates the Production of the Toxic Aβ42 Aggregates Linked with Alzheimer’s Disease. Sci. Adv. 2016, 2 (2), e1501244 DOI: 10.1126/sciadv.1501244Google Scholar78https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvVWku7g%253D&md5=5cab04625df0052f5d64f57be85007bdAn anticancer drug suppresses the primary nucleation reaction that initiates the production of the toxic Ab42 aggregates linked with Alzheimer's diseaseHabchi, Johnny; Arosio, Paolo; Perni, Michele; Costa, Ana Rita; Yagi-Utsumi, Maho; Joshi, Priyanka; Chia, Sean; Cohen, Samuel I. A.; Muller, Martin B. D.; Linse, Sara; Nollen, Ellen A. A.; Dobson, Christopher M.; Knowles, Tuomas P. J.; Vendruscolo, MicheleScience Advances (2016), 2 (2), e1501244/1-e1501244/14CODEN: SACDAF; ISSN:2375-2548. (American Association for the Advancement of Science)The conversion of the β-amyloid (Aβ) peptide into pathogenic aggregates is linked to the onset and progression of Alzheimer's disease. Although this observation has prompted an extensive search for therapeutic agents to modulate the concn. of Aβ or inhibit its aggregation, all clin. trials with these objectives have so far failed, at least in part because of a lack of understanding of the mol. mechanisms underlying the process of aggregation and its inhibition. To address this problem, we describe a chem. kinetics approach for rational drug discovery, in which the effects of small mols. on the rates of specific microscopic steps in the self-assembly of Aβ42, the most aggregation-prone variant of Aβ, are analyzed quant. By applying this approach, we report that bexarotene, an anticancer drug approved by the U.S. Food and Drug Administration, selectively targets the primary nucleation step in Aβ42 aggregation, delays the formation of toxic species in neuroblastoma cells, and completely suppresses Aβ42 deposition and its consequences in a Caenorhabditis elegans model of Aβ42-mediated toxicity. These results suggest that the prevention of the primary nucleation of Aβ42 by compds. such as bexarotene could potentially reduce the risk of onset of Alzheimer's disease and, more generally, that our strategy provides a general framework for the rational identification of a range of candidate drugs directed against neurodegenerative disorders.
- 79Granata, D.; Baftizadeh, F.; Habchi, J.; Galvagnion, C.; De Simone, A.; Camilloni, C.; Laio, A.; Vendruscolo, M. The Inverted Free Energy Landscape of an Intrinsically Disordered Peptide by Simulations and Experiments. Sci. Rep. 2015, 5 (1), 15449, DOI: 10.1038/srep15449Google ScholarThere is no corresponding record for this reference.
- 80Chong, S.-H.; Ham, S. Folding Free Energy Landscape of Ordered and Intrinsically Disordered Proteins. Sci. Rep. 2019, 9 (1), 14927, DOI: 10.1038/s41598-019-50825-6Google Scholar80https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnptVersw%253D%253D&md5=a20e190c3fa60943db197bf1a798b90aFolding Free Energy Landscape of Ordered and Intrinsically Disordered ProteinsChong Song-Ho; Ham SihyunScientific reports (2019), 9 (1), 14927 ISSN:.Folding funnel is the essential concept of the free energy landscape for ordered proteins. How does this concept apply to intrinsically disordered proteins (IDPs)? Here, we address this fundamental question through the explicit characterization of the free energy landscapes of the representative α-helical (HP-35) and β-sheet (WW domain) proteins and of an IDP (pKID) that folds upon binding to its partner (KIX). We demonstrate that HP-35 and WW domain indeed exhibit the steep folding funnel: the landscape slope for these proteins is ca. -50 kcal/mol, meaning that the free energy decreases by ~5 kcal/mol upon the formation of 10% native contacts. On the other hand, the landscape of pKID is funneled but considerably shallower (slope of -24 kcal/mol), which explains why pKID is disordered in free environments. Upon binding to KIX, the landscape of pKID now becomes significantly steep (slope of -54 kcal/mol), which enables otherwise disordered pKID to fold. We also show that it is the pKID-KIX intermolecular interactions originating from hydrophobic residues that mainly confer the steep folding funnel. The present work not only provides the quantitative characterization of the protein folding free energy landscape, but also establishes the usefulness of the folding funnel concept to IDPs.
- 81Saravanan, K. M.; Zhang, H.; Zhang, H.; Xi, W.; Wei, Y. On the Conformational Dynamics of β-Amyloid Forming Peptides: A Computational Perspective. Front. Bioeng. Biotechnol. 2020, 8, 532, DOI: 10.3389/fbioe.2020.00532Google Scholar81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jhs1Sntg%253D%253D&md5=356542cee75cb7444d118574fcacd7ffOn the Conformational Dynamics of β-Amyloid Forming Peptides: A Computational PerspectiveSaravanan Konda Mani; Zhang Haiping; Zhang Huiling; Xi Wenhui; Wei YanjieFrontiers in bioengineering and biotechnology (2020), 8 (), 532 ISSN:2296-4185.Understanding the conformational dynamics of proteins and peptides involved in important functions is still a difficult task in computational structural biology. Because such conformational transitions in β-amyloid (Aβ) forming peptides play a crucial role in many neurological disorders, researchers from different scientific fields have been trying to address issues related to the folding of Aβ forming peptides together. Many theoretical models have been proposed in the recent years for studying Aβ peptides using mathematical, physicochemical, and molecular dynamics simulation, and machine learning approaches. In this article, we have comprehensively reviewed the developmental advances in the theoretical models for Aβ peptide folding and interactions, particularly in the context of neurological disorders. Furthermore, we have extensively reviewed the advances in molecular dynamics simulation as a tool used for studying the conversions between polymorphic amyloid forms and applications of using machine learning approaches in predicting Aβ peptides and aggregation-prone regions in proteins. We have also provided details on the theoretical advances in the study of Aβ peptides, which would enhance our understanding of these peptides at the molecular level and eventually lead to the development of targeted therapies for certain acute neurological disorders such as Alzheimer's disease in the future.
- 82Grasso, G.; Danani, A. Molecular Simulations of Amyloid Beta Assemblies. Adv. Phys.: x 2020, 5 (1), 1770627, DOI: 10.1080/23746149.2020.1770627Google Scholar82https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFanu77F&md5=3f1db67e2d05a8204ce25dd35f6c66ceMolecular simulations of amyloid beta assembliesGrasso, Gianvito; Danani, AndreaAdvances in Physics: X (2020), 5 (1), 1770627CODEN: APXDAR; ISSN:2374-6149. (Taylor & Francis Ltd.)Several neurodegenerative disorders arise from the abnormal protein aggregation in the nervous tissue lead- ing tointracellular inclusions or extracellular aggregates in specific brain areas. In case of Alzheimer Disease, the accumulation of the Amyloid Beta peptide in the brain is proposed to be an early important event in the pathogenesis. Despite significant research efforts in this field, the mol. mechanisms of protein misfolding and aggregation remain somewhat unrevealed.Within this framework, computer simulations represent a power- ful tool able to connect macroscopic exptl. fiend- ings to nanoscale mol. events.However, from the computational point of View, insufficient sampling often limits the ability of computer simulations to fully address this point. One of the main challenges of MD simulations is the ability to sample exptl. relevant millise- cond to second timescales.The present review describes the applications of mol. dynamics techniques to elucidate the conformational states and the aggregation pathway of the Amyloid Beta peptide responsible for AD. Moreover, the computational studies focused on the impact of Amyloid Beta assemblies on cell membranes will be also described. Finally, the interaction mechanisms between promising small mols. and Amyloid Beta assemblies will be discussed within the field of designing new efficient drugs against neurodegenerative disorders.
- 83Haass, C.; Kaether, C.; Thinakaran, G.; Sisodia, S. Trafficking and Proteolytic Processing of APP. Cold Spring Harbor Perspect. Med. 2012, 2 (5), a006270, DOI: 10.1101/cshperspect.a006270Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntlenur0%253D&md5=b050f8e78d9221e56ef34a63e8fda596Trafficking and proteolytic processing of APPHaass, Christian; Kaether, Christoph; Thinakaran, Copal; Sisodia, SangramCold Spring Harbor Perspectives in Medicine (2012), 2 (5), a006270/1-a006270/25CODEN: CSHPFV; ISSN:2157-1422. (Cold Spring Harbor Laboratory Press)A review. Accumulations of insol. deposits of amyloid β-peptide are major pathol. hallmarks of Alzheimer disease. Amyloid β-peptide is derived by sequential proteolytic processing from a large type 1 trans-membrane protein, the β-amyloid precursor protein. The proteolytic enzymes involved in its processing are named secretases. β- And γ-secretase liberate by sequential cleavage the neurotoxic amyloid β-peptide, whereas α-secretase prevents its generation by cleaving within the middle of the amyloid domain. In this chapter we describe the cell biol. and biochem. characteristics of the three secretase activities involved in the proteolytic processing of the precursor protein. In addn. we outline how the precursor protein maturates and traffics through the secretory pathway to reach the subcellular locations where the individual secretases are preferentially active. Furthermore, we illuminate how neuronal activity and mutations which cause familial Alzheimer disease affect amyloid β-peptide generation and therefore disease onset and progression.
- 84Zhou, M.; Wen, H.; Lei, H.; Zhang, T. Molecular Dynamics Study of Conformation Transition from Helix to Sheet of Aβ42 Peptide. J. Mol. Graphics Modell. 2021, 109, 108027, DOI: 10.1016/j.jmgm.2021.108027Google Scholar84https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVamsr%252FJ&md5=2f9eda592333471c3d2f135c4f193c59Molecular dynamics study of conformation transition from helix to sheet of Aβ42 peptideZhou, Min; Wen, Huilin; Lei, Huimin; Zhang, TaoJournal of Molecular Graphics & Modelling (2021), 109 (), 108027CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Ltd.)Aβ42 peptides can form helix and sheet structure under different conditions. The conformational conversion is closely assocd. with Aβ peptides aggregation and their neurotoxicity. But the transition from helix to sheet is not be clearly understood. In this study we performed microsecond timescale MD simulations of Aβ42 peptide to investigate the conformation transition from α-helix to β-sheet. Markov state model (MSM) was built to facilitate identification of crucial intermediate states and possible transition pathway. Based on the anal., we found that the region Y10-A21 in the middle of Aβ42 peptide plays an initial role in this transition. MSM model revealed that the collapse of helical structure in this region might trigger the formation of sheet structure. Moreover, we further simulated the aggregation of Aβ42 peptides with different conformations. We found that the Aβ42 peptides forming sheet structure have higher aggregation potential compared with peptides with helix structure. These results demonstrate that we can prevent the aggregation of Aβ42 peptides by stabilizing the helix structure in the region of Y10-A21. In addn., this study provides new insight into better understanding the conformational transition and aggregation of Aβ42 peptides.
- 85Shuaib, S.; Goyal, B. Scrutiny of the Mechanism of Small Molecule Inhibitor Preventing Conformational Transition of Amyloid-Β42 Monomer: Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2018, 36 (3), 663– 678, DOI: 10.1080/07391102.2017.1291363Google ScholarThere is no corresponding record for this reference.
- 86Liu, F.; Ma, Z.; Sang, J.; Lu, F. Edaravone Inhibits the Conformational Transition of Amyloid-Β42: Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2020, 38 (8), 2377– 2388, DOI: 10.1080/07391102.2019.1632225Google ScholarThere is no corresponding record for this reference.
- 87Narang, S. S.; Goyal, D.; Goyal, B. Inhibition of Alzheimer’s Amyloid-Β42 Peptide Aggregation by a Bi-Functional Bis-Tryptoline Triazole: Key Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2020, 38 (6), 1598– 1611, DOI: 10.1080/07391102.2019.1614093Google ScholarThere is no corresponding record for this reference.
- 88Cao, Y.; Jiang, X.; Han, W. Self-Assembly Pathways of β-Sheet-Rich Amyloid-β(1–40) Dimers: Markov State Model Analysis on Millisecond Hybrid-Resolution Simulations. J. Chem. Theory Comput. 2017, 13 (11), 5731– 5744, DOI: 10.1021/acs.jctc.7b00803Google Scholar88https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1CisLzE&md5=761fcde5aab69118ef6332fda366f37bSelf-Assembly Pathways of β-Sheet-Rich Amyloid-β(1-40) Dimers: Markov State Model Analysis on Millisecond Hybrid-Resolution SimulationsCao, Yang; Jiang, Xuehan; Han, WeiJournal of Chemical Theory and Computation (2017), 13 (11), 5731-5744CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Early oligomerization during amyloid-β (Aβ) aggregation is essential for Aβ neurotoxicity. Understanding how unstructured Aβs assemble into oligomers, esp. those rich in β-sheets, is essential but remains challenging as the assembly process is too transient for exptl. characterization and too slow for mol. dynamics simulations. So far, at. simulations are limited only to studies of either oligomer structures or assembly pathways for short Aβ segments. Here, to overcome the computational challenge, we combined in this study a hybrid-resoln. model and adaptive sampling techniques to perform over 2.7 ms of simulations of formation of full-length Aβ40 dimers that are the earliest toxic oligomeric species. The Markov state model was further employed to characterize the transition pathways and assocd. kinetics. The results showed that for 2 major forms of β-sheet-rich structures reported exptl., the corresponding assembly mechanisms were markedly different. Hairpin-contg. structures were formed by direct binding of sol. Aβ in β-hairpin-like conformations. The formation of parallel, in-register structures resembling fibrils occurred ∼100-fold more slowly and involved a rapid encounter of Aβ in arbitrary conformations followed by a slow structural conversion. The structural conversion proceeded via diverse pathways but always required transient unfolding of encounter complexes. We found that the transition kinetics could be affected differently by intramol./intermol. interactions involving individual residues in a conformation-dependent manner. In particular, the interactions involving Aβ's N-terminal part promoted the assembly into hairpin-contg. structures but delayed the formation of fibril-like structures, thus explaining puzzling observations reported previously regarding the roles of this region in the early assembly process.
- 89Rojas, A. V.; Liwo, A.; Scheraga, H. A. A Study of the α-Helical Intermediate Preceding the Aggregation of the Amino-Terminal Fragment of the β Amyloid Peptide (Aβ1–28). J. Phys. Chem. B 2011, 115 (44), 12978– 12983, DOI: 10.1021/jp2050993Google Scholar89https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlaqtrrI&md5=bb4a962dd9c9ef4527c600252e2f3b71A Study of the α-Helical Intermediate Preceding the Aggregation of the Amino-Terminal Fragment of the β Amyloid Peptide (Aβ1-28)Rojas, Ana V.; Liwo, Adam; Scheraga, Harold A.Journal of Physical Chemistry B (2011), 115 (44), 12978-12983CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)The β amyloid (Aβ) peptide aggregates to form β-rich structures that are known to trigger Alzheimer's disease. Expts. suggest that an α-helical intermediate precedes the formation of these aggregates. However, a description at the mol. level of the α-to-β transition has not been obtained. Because it has been proposed that the transition might be initiated in the amino-terminal region of Aβ, we studied the aggregation of the 28-residue amino-terminal fragment of Aβ (Aβ1-28) using mol. dynamics (MD) and a coarse-grained force field. Simulations starting from extended and helical conformations showed that oligomerization is initiated by the formation of intermol. β-sheets between the residues in the N-terminal regions. In simulations starting from the α-helical conformation, forcing residues 17-21 to remain in the initial (helical) conformation prevents aggregation but allows for the formation of dimers, indicating that oligomerization, initiated along the nonhelical N-terminal regions, cannot progress without the α-to-β transition propagating along the chains.
- 90Tarasoff-Conway, J. M.; Carare, R. O.; Osorio, R. S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B. V. Clearance Systems in the Brain-Implications for Alzheimer Disease. Nat. Rev. Neurol. 2015, 11 (8), 457– 470, DOI: 10.1038/nrneurol.2015.119Google Scholar90https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1Wqt73N&md5=52b11eec4c752ab3fdaa14b721478061Clearance systems in the brain-implications for Alzheimer diseaseTarasoff-Conway, Jenna M.; Carare, Roxana O.; Osorio, Ricardo S.; Glodzik, Lidia; Butler, Tracy; Fieremans, Els; Axel, Leon; Rusinek, Henry; Nicholson, Charles; Zlokovic, Berislav V.; Frangione, Blas; Blennow, Kaj; Menard, Joel; Zetterberg, Henrik; Wisniewski, Thomas; de Leon, Mony J.Nature Reviews Neurology (2015), 11 (8), 457-470CODEN: NRNACP; ISSN:1759-4758. (Nature Publishing Group)Accumulation of toxic protein aggregates-amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles is the pathol. hallmark of Alzheimer disease (AD). Aβ accumulation has been hypothesized to result from an imbalance between Aβ prodn. and clearance; indeed, Aβ clearance seems to be impaired in both early and late forms of AD. To develop efficient strategies to slow down or halt AD, it is crit. to understand how Aβ is cleared from the brain. Extracellular Aβ deposits can be removed from the brain by various clearance systems, most importantly, transport across the blood-brain barrier. Findings from the past few years suggest that astroglial-mediated interstitial fluid (ISF) bulk flow, known as the glymphatic system, might contribute to a larger portion of extracellular Aβ (eAβ) clearance than previously thought. The meningeal lymphatic vessels, discovered in 2015, might provide another clearance route. Because these clearance systems act together to drive eAβ from the brain, any alteration to their function could contribute to AD. An understanding of Aβ clearance might provide strategies to reduce excess Aβ deposits and delay, or even prevent, disease onset. In this Review, we describe the clearance systems of the brain as they relate to proteins implicated in AD pathol., with the main focus on Aβ.
- 91Patterson, B. W.; Elbert, D. L.; Mawuenyega, K. G.; Kasten, T.; Ovod, V.; Ma, S.; Xiong, C.; Chott, R.; Yarasheski, K.; Sigurdson, W. Age and Amyloid Effects on Human Central Nervous System Amyloid-Beta Kinetics. Ann. Neurol. 2015, 78 (3), 439– 453, DOI: 10.1002/ana.24454Google ScholarThere is no corresponding record for this reference.
- 92Yamazaki, Y.; Zhao, N.; Caulfield, T. R.; Liu, C.-C.; Bu, G. Apolipoprotein E and Alzheimer Disease: Pathobiology and Targeting Strategies. Nat. Rev. Neurol. 2019, 15 (9), 501– 518, DOI: 10.1038/s41582-019-0228-7Google ScholarThere is no corresponding record for this reference.
- 93Bye, J. W.; Falconer, R. J. Thermal Stability of Lysozyme as a Function of Ion Concentration: A Reappraisal of the Relationship between the Hofmeister Series and Protein Stability. Protein Sci. 2013, 22 (11), 1563– 1570, DOI: 10.1002/pro.2355Google Scholar93https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1eksrfP&md5=bde6c804bbabbe81d8e60cdeaba2e3d4Thermal stability of lysozyme as a function of ion concentration: A reappraisal of the relationship between the Hofmeister series and protein stabilityBye, Jordan W.; Falconer, Robert J.Protein Science (2013), 22 (11), 1563-1570CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Anion and cation effects on the structural stability of lysozyme were investigated using differential scanning calorimetry. At low concns. (<5 mM) anions and cations alter the stability of lysozyme but they do not follow the Hofmeister (or inverse Hofmeister) series. At higher concns. protein stabilization follows the well-established Hofmeister series. Our hypothesis is that there are three mechanisms at work. At low concns. the anions interact with charged side chains where the presence of the ion can alter the structural stability of the protein. At higher concns. the low charge d. anions perchlorate and iodide interact weakly with the protein. Their presence however reduces the Gibbs free energy required to hydrate the core of the protein that is exposed during unfolding therefore destabilizing the structure. At higher concns. the high charge d. anions phosphate and sulfate compete for water with the protein as it unfolds increasing the Gibbs free energy required to hydrate the newly exposed core of the protein therefore stabilizing the structure.
- 94Martens, Y. A.; Zhao, N.; Liu, C.-C.; Kanekiyo, T.; Yang, A. J.; Goate, A. M.; Holtzman, D. M.; Bu, G. ApoE Cascade Hypothesis in the Pathogenesis of Alzheimer’s Disease and Related Dementias. Neuron 2022, 110 (8), 1304– 1317, DOI: 10.1016/j.neuron.2022.03.004Google ScholarThere is no corresponding record for this reference.
- 95Chai, A. B.; Lam, H. H. J.; Kockx, M.; Gelissen, I. C. Apolipoprotein E Isoform-Dependent Effects on the Processing of Alzheimer’s Amyloid-β. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2021, 1866 (9), 158980, DOI: 10.1016/j.bbalip.2021.158980Google ScholarThere is no corresponding record for this reference.
- 96Tijms, B. M.; Vromen, E. M.; Mjaavatten, O.; Holstege, H.; Reus, L. M.; van der Lee, S.; Wesenhagen, K. E. J.; Lorenzini, L.; Vermunt, L.; Venkatraghavan, V. Cerebrospinal Fluid Proteomics in Patients with Alzheimer’s Disease Reveals Five Molecular Subtypes with Distinct Genetic Risk Profiles. Nat. Aging 2024, 4 (1), 33– 47, DOI: 10.1038/s43587-023-00550-7Google ScholarThere is no corresponding record for this reference.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 96 other publications.
- 1Gustavsson, A.; Norton, N.; Fast, T.; Frölich, L.; Georges, J.; Holzapfel, D.; Kirabali, T.; Krolak-Salmon, P.; Rossini, P. M.; Ferretti, M. T. Global Estimates on the Number of Persons across the Alzheimer’s Disease Continuum. Alzheimer’s Dementia 2022, 19, 658– 670, DOI: 10.1002/alz.12694There is no corresponding record for this reference.
- 2Benilova, I.; Karran, E.; De Strooper, B. The Toxic Aβ Oligomer and Alzheimer’s Disease: An Emperor in Need of Clothes. Nat. Neurosci. 2012, 15 (3), 349– 357, DOI: 10.1038/nn.30282https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtlOltrc%253D&md5=071f3c9b1e0fe9b2e66363c44d87e381The toxic Aβ oligomer and Alzheimer's disease: an emperor in need of clothesBenilova, Iryna; Karran, Eric; De Strooper, BartNature Neuroscience (2012), 15 (3), 349-357CODEN: NANEFN; ISSN:1097-6256. (Nature Publishing Group)A review. The 'toxic Aβ oligomer' hypothesis has attracted considerable attention among Alzheimer's disease researchers as a way of resolving the lack of correlation between deposited amyloid-β (Aβ) in amyloid plaques-in terms of both amt. and location-and cognitive impairment or neurodegeneration. However, the lack of a common, agreed-upon exptl. description of the toxic Aβ oligomer makes interpretation and direct comparison of data between different research groups impossible. Here we critically review the evidence supporting toxic Aβ oligomers as drivers of neurodegeneration and make some suggestions that might facilitate progress in this complex field.
- 3Karran, E.; De Strooper, B. The Amyloid Hypothesis in Alzheimer Disease: New Insights from New Therapeutics. Nat. Rev. Drug Discov. 2022, 21 (4), 306– 318, DOI: 10.1038/s41573-022-00391-w3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xkt1aru7s%253D&md5=44187b2d3c8e4aae3a27a9c18c80c6c3The amyloid hypothesis in Alzheimer disease: new insights from new therapeuticsKarran, Eric; De Strooper, BartNature Reviews Drug Discovery (2022), 21 (4), 306-318CODEN: NRDDAG; ISSN:1474-1776. (Nature Portfolio)Many drugs that target amyloid-beta (Abeta) in Alzheimer disease (AD) have failed to demonstrate clin. efficacy. However, four anti-Abeta antibodies have been shown to mediate the removal of amyloid plaque from brains of patients with AD, and the FDA has recently granted accelerated approval to one of these, aducanumab, using redn. of amyloid plaque as a surrogate end point. The rationale for approval and the extent of the clin. benefit from these antibodies are under intense debate. With the aim of informing this debate, we review clin. trial data for drugs that target Abeta from the perspective of the temporal interplay between the two pathognomonic protein aggregates in AD - Abeta plaques and tau neurofibrillary tangles - and their relationship to cognitive impairment, highlighting differences in drug properties that could affect their clin. performance. On this basis, we propose that Abeta pathol. drives tau pathol., that amyloid plaque would need to be reduced to a low level (®20 centiloids) to reveal significant clin. benefit and that there will be a lag between the removal of amyloid and the potential to observe a clin. benefit. We conclude that the speed of amyloid removal from the brain by a potential therapy will be important in demonstrating clin. benefit in the context of a clin. trial.
- 4Castellani, R. J.; Plascencia-Villa, G.; Perry, G. The Amyloid Cascade and Alzheimer’s Disease Therapeutics: Theory versus Observation. Lab. Invest. 2019, 99 (7), 958– 970, DOI: 10.1038/s41374-019-0231-zThere is no corresponding record for this reference.
- 5Matiiv, A. B.; Trubitsina, N. P.; Matveenko, A. G.; Barbitoff, Y. A.; Zhouravleva, G. A.; Bondarev, S. A. Amyloid and Amyloid-Like Aggregates: Diversity and the Term Crisis. Biochemistry (Moscow) 2020, 85 (9), 1011– 1034, DOI: 10.1134/S0006297920090035There is no corresponding record for this reference.
- 6Bhattacharya, S.; Lin, X. Recent Advances in Computational Protocols Addressing Intrinsically Disordered Proteins. Biomolecules 2019, 9 (4), 146, DOI: 10.3390/biom90401466https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXovFCgsb4%253D&md5=42bb3479001b582aeaa37b9e25c8db57Recent advances in computational protocols addressing intrinsically disordered proteinsBhattacharya, Supriyo; Lin, XingchengBiomolecules (2019), 9 (4), 146CODEN: BIOMHC; ISSN:2218-273X. (MDPI AG)A review. Intrinsically disordered proteins (IDP) are abundant in the human genome and have recently emerged as major therapeutic targets for various diseases. Unlike traditional proteins that adopt a definitive structure, IDPs in free soln. are disordered and exist as an ensemble of conformations. This enables the IDPs to signal through multiple signaling pathways and serve as scaffolds for multi-protein complexes. The challenge in studying IDPs exptl. stems from their disordered nature. NMR (NMR), CD, small angle X-ray scattering, and single mol. Forster resonance energy transfer (FRET) can give the local structural information and overall dimension of IDPs, but seldom provide a unified picture of the whole protein. To understand the conformational dynamics of IDPs and how their structural ensembles recognize multiple binding partners and small mol. inhibitors, knowledge-based and physics-based sampling techniques are utilized in-silico, guided by exptl. structural data. However, efficient sampling of the IDP conformational ensemble requires traversing the numerous degrees of freedom in the IDP energy landscape, as well as force-fields that accurately model the protein and solvent interactions. In this review, we have provided an overview of the current state of computational methods for studying IDP structure and dynamics and discussed the major challenges faced in this field.
- 7Saurabh, S.; Nadendla, K.; Purohit, S. S.; Sivakumar, P. M.; Cetinel, S. Fuzzy Drug Targets: Disordered Proteins in the Drug-Discovery Realm. ACS Omega 2023, 8 (11), 9729– 9747, DOI: 10.1021/acsomega.2c07708There is no corresponding record for this reference.
- 8Paul, A.; Samantray, S.; Anteghini, M.; Khaled, M.; Strodel, B. Thermodynamics and Kinetics of the Amyloid-β Peptide Revealed by Markov State Models Based on MD Data in Agreement with Experiment. Chem. Sci. 2021, 12 (19), 6652– 6669, DOI: 10.1039/D0SC04657D8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXovVaqtrk%253D&md5=53401b7f733acd3039b84361fe413cbcThermodynamics and kinetics of the amyloid-β peptide revealed by Markov state models based on MD data in agreement with experimentPaul, Arghadwip; Samantray, Suman; Anteghini, Marco; Khaled, Mohammed; Strodel, BirgitChemical Science (2021), 12 (19), 6652-6669CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)The amlyoid-β peptide (Aβ) is closely linked to the development of Alzheimer's disease. Mol. dynamics (MD) simulations have become an indispensable tool for studying the behavior of this peptide at the atomistic level. General key aspects of MD simulations are the force field used for modeling the peptide and its environment, which is important for accurate modeling of the system of interest, and the length of the simulations, which dets. whether or not equil. is reached. In this study we address these points by analyzing 30-μs MD simulations acquired for Aβ40 using seven different force fields. We assess the convergence of these simulations based on the convergence of various structural properties and of NMR and fluorescence spectroscopic observables. Moreover, we calc Markov state models for the different MD simulations, which provide an unprecedented view of the thermodn. and kinetics of the amyloid-β peptide. This further allows us to provide answers for pertinent questions, like: which force fields are suitable for modeling Aβ (a99SB-UCB and a99SB-ILDN/TIP4P-D); what does Aβ peptide really look like (mostly extended and disordered) and; how long does it take MD simulations of Aβ to attain equil. (at least 20-30μs). We believe the analyses presented in this study will provide a useful ref. guide for important questions relating to the structure and dynamics of Aβ in particular, and by extension other similar disordered proteins.
- 9McGibbon, R. T.; Pande, V. S. Variational Cross-Validation of Slow Dynamical Modes in Molecular Kinetics. J. Chem. Phys. 2015, 142 (12), 124105, DOI: 10.1063/1.49162929https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXlsVWnur0%253D&md5=dc34e6f64a11721767439e91e68e778aVariational cross-validation of slow dynamical modes in molecular kineticsMcGibbon, Robert T.; Pande, Vijay S.Journal of Chemical Physics (2015), 142 (12), 124105/1-124105/12CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Markov state models are a widely used method for approximating the eigenspectrum of the mol. dynamics propagator, yielding insight into the long-timescale statistical kinetics and slow dynamical modes of biomol. systems. However, the lack of a unified theor. framework for choosing between alternative models has hampered progress, esp. for non-experts applying these methods to novel biol. systems. Here, we consider cross-validation with a new objective function for estimators of these slow dynamical modes, a generalized matrix Rayleigh quotient (GMRQ), which measures the ability of a rank-m projection operator to capture the slow subspace of the system. It is shown that a variational theorem bounds the GMRQ from above by the sum of the first m eigenvalues of the system's propagator, but that this bound can be violated when the requisite matrix elements are estd. subject to statistical uncertainty. This overfitting can be detected and avoided through cross-validation. These result make it possible to construct Markov state models for protein dynamics in a way that appropriately captures the tradeoff between systematic and statistical errors. (c) 2015 American Institute of Physics.
- 10Spiriti, J.; Noé, F.; Wong, C. F. Simulation of Ligand Dissociation Kinetics from the Protein Kinase PYK2. J. Comput. Chem. 2022, 43 (28), 1911– 1922, DOI: 10.1002/jcc.26991There is no corresponding record for this reference.
- 11Dominic, A. J. I.; Cao, S.; Montoya-Castillo, A.; Huang, X. Memory Unlocks the Future of Biomolecular Dynamics: Transformative Tools to Uncover Physical Insights Accurately and Efficiently. J. Am. Chem. Soc. 2023, 145 (18), 9916– 9927, DOI: 10.1021/jacs.3c0109511https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXosFyksr8%253D&md5=be056dee3d0e6c4987050f956bb70a2eMemory Unlocks the Future of Biomolecular Dynamics: Transformative Tools to Uncover Physical Insights Accurately and EfficientlyDominic, Anthony J.; Cao, Siqin; Montoya-Castillo, Andres; Huang, XuhuiJournal of the American Chemical Society (2023), 145 (18), 9916-9927CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)A review. Conformational changes underpin function and encode complex biomol. mechanisms. Gaining at.-level detail of how such changes occur has the potential to reveal these mechanisms and is of crit. importance in identifying drug targets, facilitating rational drug design, and enabling bioengineering applications. While the past two decades have brought Markov state model techniques to the point where practitioners can regularly use them to glimpse the long-time dynamics of slow conformations in complex systems, many systems are still beyond their reach. In this Perspective, we discuss how including memory (i.e., non-Markovian effects) can reduce the computational cost to predict the long-time dynamics in these complex systems by orders of magnitude and with greater accuracy and resoln. than state-of-the-art Markov state models. We illustrate how memory lies at the heart of successful and promising techniques, ranging from the Fokker-Planck and generalized Langevin equations to deep-learning recurrent neural networks and generalized master equations. We delineate how these techniques work, identify insights that they can offer in biomol. systems, and discuss their advantages and disadvantages in practical settings. We show how generalized master equations can enable the investigation of, for example, the gate-opening process in RNA polymerase II and demonstrate how our recent advances tame the deleterious influence of statistical underconvergence of the mol. dynamics simulations used to parameterize these techniques. This represents a significant leap forward that will enable our memory-based techniques to interrogate systems that are currently beyond the reach of even the best Markov state models. We conclude by discussing some current challenges and future prospects for how exploiting memory will open the door to many exciting opportunities.
- 12Noé, F.; Wu, H.; Prinz, J.-H.; Plattner, N. Projected and Hidden Markov Models for Calculating Kinetics and Metastable States of Complex Molecules. J. Chem. Phys. 2013, 139 (18), 184114, DOI: 10.1063/1.482881612https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslKqt77P&md5=27bea55f9a13aae7f3231bb36ad2c1ceProjected and hidden Markov models for calculating kinetics and metastable states of complex moleculesNoe, Frank; Wu, Hao; Prinz, Jan-Hendrik; Plattner, NuriaJournal of Chemical Physics (2013), 139 (18), 184114/1-184114/17CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Markov state models (MSMs) have been successful in computing metastable states, slow relaxation timescales and assocd. structural changes, and stationary or kinetic exptl. observables of complex mols. from large amts. of mol. dynamics simulation data. However, MSMs approx. the true dynamics by assuming a Markov chain on a clusters discretization of the state space. This approxn. is difficult to make for high-dimensional biomol. systems, and the quality and reproducibility of MSMs has, therefore, been limited. Here, we discard the assumption that dynamics are Markovian on the discrete clusters. Instead, we only assume that the full phase-space mol. dynamics is Markovian, and a projection of this full dynamics is obsd. on the discrete states, leading to the concept of Projected Markov Models (PMMs). Robust estn. methods for PMMs are not yet available, but we derive a practically feasible approxn. via Hidden Markov Models (HMMs). It is shown how various mol. observables of interest that are often computed from MSMs can be computed from HMMs/PMMs. The new framework is applicable to both, simulation and single-mol. exptl. data. We demonstrate its versatility by applications to educative model systems, a 1 ms Anton MD simulation of the bovine pancreatic trypsin inhibitor protein, and an optical tweezer force probe trajectory of an RNA hairpin. (c) 2013 American Institute of Physics.
- 13Suárez, E.; Wiewiora, R. P.; Wehmeyer, C.; Noé, F.; Chodera, J. D.; Zuckerman, D. M. What Markov State Models Can and Cannot Do: Correlation versus Path-Based Observables in Protein-Folding Models. J. Chem. Theory Comput. 2021, 17 (5), 3119– 3133, DOI: 10.1021/acs.jctc.0c01154There is no corresponding record for this reference.
- 14Dominic, A. J.; Sayer, T.; Cao, S.; Markland, T. E.; Huang, X.; Montoya-Castillo, A. Building Insightful, Memory-Enriched Models to Capture Long-Time Biochemical Processes from Short-Time Simulations. Proc. Natl. Acad. Sci. U. S. A. 2023, 120 (12), e2221048120 DOI: 10.1073/pnas.2221048120There is no corresponding record for this reference.
- 15Wehmeyer, C.; Noé, F. Time-Lagged Autoencoders: Deep Learning of Slow Collective Variables for Molecular Kinetics. J. Chem. Phys. 2018, 148 (24), 241703, DOI: 10.1063/1.501139915https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXkslahsro%253D&md5=9e455103c2f05e0a6c0b16f93a9cf074Time-lagged autoencoders: Deep learning of slow collective variables for molecular kineticsWehmeyer, Christoph; Noe, FrankJournal of Chemical Physics (2018), 148 (24), 241703/1-241703/9CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Inspired by the success of deep learning techniques in the phys. and chem. sciences, we apply a modification of an autoencoder type deep neural network to the task of dimension redn. of mol. dynamics data. We can show that our time-lagged autoencoder reliably finds low-dimensional embeddings for high-dimensional feature spaces which capture the slow dynamics of the underlying stochastic processes - beyond the capabilities of linear dimension redn. techniques. (c) 2018 American Institute of Physics.
- 16Mardt, A.; Pasquali, L.; Wu, H.; Noé, F. VAMPnets for Deep Learning of Molecular Kinetics. Nat. Commun. 2018, 9 (1), 5, DOI: 10.1038/s41467-017-02388-116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MzmvFGqtA%253D%253D&md5=a5871b9d4dc574f431de970e765e4262VAMPnets for deep learning of molecular kineticsMardt Andreas; Pasquali Luca; Wu Hao; Noe FrankNature communications (2018), 9 (1), 5 ISSN:.There is an increasing demand for computing the relevant structures, equilibria, and long-timescale kinetics of biomolecular processes, such as protein-drug binding, from high-throughput molecular dynamics simulations. Current methods employ transformation of simulated coordinates into structural features, dimension reduction, clustering the dimension-reduced data, and estimation of a Markov state model or related model of the interconversion rates between molecular structures. This handcrafted approach demands a substantial amount of modeling expertise, as poor decisions at any step will lead to large modeling errors. Here we employ the variational approach for Markov processes (VAMP) to develop a deep learning framework for molecular kinetics using neural networks, dubbed VAMPnets. A VAMPnet encodes the entire mapping from molecular coordinates to Markov states, thus combining the whole data processing pipeline in a single end-to-end framework. Our method performs equally or better than state-of-the-art Markov modeling methods and provides easily interpretable few-state kinetic models.
- 17Löhr, T.; Kohlhoff, K.; Heller, G. T.; Camilloni, C.; Vendruscolo, M. A Kinetic Ensemble of the Alzheimer’s Aβ Peptide. Nat. Comput. Sci. 2021, 1 (1), 71– 78, DOI: 10.1038/s43588-020-00003-wThere is no corresponding record for this reference.
- 18Ghorbani, M.; Prasad, S.; Klauda, J. B.; Brooks, B. R. GraphVAMPNet, using graph neural networks and variational approach to Markov processes for dynamical modeling of biomolecules. J. Chem. Phys. 2022, 156 (18), 184103, DOI: 10.1063/5.008560718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38Xht1Oitr%252FL&md5=fd3c7fd1ee3f3af64afa1f53c257eff9GraphVAMPNet, using graph neural networks and variational approach to Markov processes for dynamical modeling of biomoleculesGhorbani, Mahdi; Prasad, Samarjeet; Klauda, Jeffery B.; Brooks, Bernard R.Journal of Chemical Physics (2022), 156 (18), 184103CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)Finding a low dimensional representation of data from long-timescale trajectories of biomol. processes, such as protein folding or ligand-receptor binding, is of fundamental importance, and kinetic models, such as Markov modeling, have proven useful in describing the kinetics of these systems. Recently, an unsupervised machine learning technique called VAMPNet was introduced to learn the low dimensional representation and the linear dynamical model in an end-to-end manner. VAMPNet is based on the variational approach for Markov processes and relies on neural networks to learn the coarse-grained dynamics. In this paper, we combine VAMPNet and graph neural networks to generate an end-to-end framework to efficiently learn high-level dynamics and metastable states from the long-timescale mol. dynamics trajectories. This method bears the advantages of graph representation learning and uses graph message passing operations to generate an embedding for each datapoint, which is used in the VAMPNet to generate a coarse-grained dynamical model. This type of mol. representation results in a higher resoln. and a more interpretable Markov model than the std. VAMPNet, enabling a more detailed kinetic study of the biomol. processes. Our GraphVAMPNet approach is also enhanced with an attention mechanism to find the important residues for classification into different metastable states. (c) 2022 American Institute of Physics.
- 19Liu, B.; Xue, M.; Qiu, Y.; Konovalov, K. A.; O’Connor, M. S.; Huang, X. GraphVAMPnets for Uncovering Slow Collective Variables of Self-Assembly Dynamics. J. Chem. Phys. 2023, 159 (9), 094901, DOI: 10.1063/5.0158903There is no corresponding record for this reference.
- 20Mardt, A.; Hempel, T.; Clementi, C.; Noé, F. Deep Learning to Decompose Macromolecules into Independent Markovian Domains. Nat. Commun. 2022, 13 (1), 7101, DOI: 10.1038/s41467-022-34603-z20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XivFOmsL7N&md5=eee8e2dd74b7a123c9f547e4a4b53eefDeep learning to decompose macromolecules into independent Markovian domainsMardt, Andreas; Hempel, Tim; Clementi, Cecilia; Noe, FrankNature Communications (2022), 13 (1), 7101CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)The increasing interest in modeling the dynamics of ever larger proteins has revealed a fundamental problem with models that describe the mol. system as being in a global configuration state. This notion limits our ability to gather sufficient statistics of state probabilities or state-to-state transitions because for large mol. systems the no. of metastable states grows exponentially with size. In this manuscript, we approach this challenge by introducing a method that combines our recent progress on independent Markov decompn. (IMD) with VAMPnets, a deep learning approach to Markov modeling. We establish a training objective that quantifies how well a given decompn. of the mol. system into independent subdomains with Markovian dynamics approximates the overall dynamics. By constructing an end-to-end learning framework, the decompn. into such subdomains and their individual Markov state models are simultaneously learned, providing a data-efficient and easily interpretable summary of the complex system dynamics. While learning the dynamical coupling between Markovian subdomains is still an open issue, the present results are a significant step towards learning Ising models of large mol. complexes from simulation data.
- 21Chen, W.; Sidky, H.; Ferguson, A. L. Nonlinear Discovery of Slow Molecular Modes Using State-Free Reversible VAMPnets. J. Chem. Phys. 2019, 150 (21), 214114, DOI: 10.1063/1.509252121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFalurrM&md5=4b423a81430965b8d714eac2daba848bNonlinear discovery of slow molecular modes using state-free reversible VAMPnetsChen, Wei; Sidky, Hythem; Ferguson, Andrew L.Journal of Chemical Physics (2019), 150 (21), 214114/1-214114/16CODEN: JCPSA6; ISSN:0021-9606. (American Institute of Physics)The success of enhanced sampling mol. simulations that accelerate along collective variables (CVs) is predicated on the availability of variables coincident with the slow collective motions governing the long-time conformational dynamics of a system. It is challenging to intuit these slow CVs for all but the simplest mol. systems, and their data-driven discovery directly from mol. simulation trajectories has been a central focus of the mol. simulation community to both unveil the important phys. mechanisms and drive enhanced sampling. In this work, we introduce state-free reversible VAMPnets (SRV) as a deep learning architecture that learns nonlinear CV approximants to the leading slow eigenfunctions of the spectral decompn. of the transfer operator that evolves equil.-scaled probability distributions through time. Orthogonality of the learned CVs is naturally imposed within network training without added regularization. The CVs are inherently explicit and differentiable functions of the input coordinates making them well-suited to use in enhanced sampling calcns. We demonstrate the utility of SRVs in capturing parsimonious nonlinear representations of complex system dynamics in applications to 1D and 2D toy systems where the true eigenfunctions are exactly calculable and to mol. dynamics simulations of alanine dipeptide and the WW domain protein. (c) 2019 American Institute of Physics.
- 22Kleiman, D. E.; Shukla, D. Active Learning of the Conformational Ensemble of Proteins Using Maximum Entropy VAMPNets. J. Chem. Theory Comput. 2023, 19 (14), 4377– 4388, DOI: 10.1021/acs.jctc.3c00040There is no corresponding record for this reference.
- 23Mardt, A.; Noé, F. Progress in Deep Markov State Modeling: Coarse Graining and Experimental Data Restraints. J. Chem. Phys. 2021, 155 (21), 214106, DOI: 10.1063/5.0064668There is no corresponding record for this reference.
- 24Tolar, M.; Abushakra, S.; Hey, J. A.; Porsteinsson, A.; Sabbagh, M. Aducanumab, Gantenerumab, BAN2401, and ALZ-801─the First Wave of Amyloid-Targeting Drugs for Alzheimer’s Disease with Potential for near Term Approval. Alzheimer’s Res. Ther. 2020, 12 (1), 95, DOI: 10.1186/s13195-020-00663-wThere is no corresponding record for this reference.
- 25Gervais, F.; Paquette, J.; Morissette, C.; Krzywkowski, P.; Yu, M.; Azzi, M.; Lacombe, D.; Kong, X.; Aman, A.; Laurin, J. Targeting Soluble Aβ Peptide with Tramiprosate for the Treatment of Brain Amyloidosis. Neurobiol. Aging 2007, 28 (4), 537– 547, DOI: 10.1016/j.neurobiolaging.2006.02.01525https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXitFyks74%253D&md5=f871b841546682e832b666dc875a68efTargeting soluble Aβ peptide with Tramiprosate for the treatment of brain amyloidosisGervais, Francine; Paquette, Julie; Morissette, Celine; Krzywkowski, Pascale; Yu, Mathilde; Azzi, Mounia; Lacombe, Diane; Kong, Xianqi; Aman, Ahmed; Laurin, Julie; Szarek, Walter A.; Tremblay, PatrickNeurobiology of Aging (2007), 28 (4), 537-547CODEN: NEAGDO; ISSN:0197-4580. (Elsevier)Amyloid β-peptide (Aβ) is a major constituent of senile plaques in Alzheimer's disease (AD). Neurotoxicity results from the conformational transition of Aβ from random-coil to β-sheet and its oligomerization. Among a series of ionic compds. able to interact with sol. Aβ, Tramiprosate (3-amino-1-propanesulfonic acid; 3APS; Alzhemed) was found to maintain Aβ in a non-fibrillar form, to decrease Aβ42-induced cell death in neuronal cell cultures, and to inhibit amyloid deposition. Tramiprosate crosses the murine blood-brain barrier (BBB) to exert its activity. Treatment of TgCRND8 mice with Tramiprosate resulted in significant redn. (∼30%) in the brain amyloid plaque load and a significant decrease in the cerebral levels of sol. and insol. Aβ40 and Aβ42 (∼20-30%). A dose-dependent redn. (up to 60%) of plasma Aβ levels was also obsd., suggesting that Tramiprosate influences the central pool of Aβ, changing either its efflux or its metab. in the brain. We propose that Tramiprosate, which targets sol. Aβ, represents a new and promising therapeutic class of drugs for the treatment of AD.
- 26Caltagirone, C.; Ferrannini, L.; Marchionni, N.; Nappi, G.; Scapagnini, G.; Trabucchi, M. The Potential Protective Effect of Tramiprosate (Homotaurine) against Alzheimer’s Disease: A Review. Aging: Clin. Exp. Res. 2012, 24 (6), 580– 587, DOI: 10.1007/BF03654836There is no corresponding record for this reference.
- 27Zou, X.; Himbert, S.; Dujardin, A.; Juhasz, J.; Ros, S.; Stöver, H. D. H.; Rheinstädter, M. C. Curcumin and Homotaurine Suppress Amyloid-Β25–35 Aggregation in Synthetic Brain Membranes. ACS Chem. Neurosci. 2021, 12 (8), 1395– 1405, DOI: 10.1021/acschemneuro.1c00057There is no corresponding record for this reference.
- 28Abushakra, S.; Porsteinsson, A.; Vellas, B.; Cummings, J.; Gauthier, S.; Hey, J. A.; Power, A.; Hendrix, S.; Wang, P.; Shen, L.; Sampalis, J.; Tolar, M. Clinical Benefits of Tramiprosate in Alzheimer’s Disease Are Associated with Higher Number of APOE4 Alleles: The “APOE4 Gene-Dose Effect. J. Prev. Alzheimers Dis. 2016, 3 (4), 219– 228, DOI: 10.14283/jpad.2016.115There is no corresponding record for this reference.
- 29Tian, J.; Dang, H.; Wallner, M.; Olsen, R.; Kaufman, D. L. Homotaurine, a Safe Blood-Brain Barrier Permeable GABAA-R-Specific Agonist, Ameliorates Disease in Mouse Models of Multiple Sclerosis. Sci. Rep. 2018, 8 (1), 16555, DOI: 10.1038/s41598-018-32733-329https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvotF2gtw%253D%253D&md5=08d91caa6e786493d66bb024fdd3c482Homotaurine, a safe blood-brain barrier permeable GABAA-R-specific agonist, ameliorates disease in mouse models of multiple sclerosisTian Jide; Dang Hoa; Wallner Martin; Olsen Richard; Kaufman Daniel LScientific reports (2018), 8 (1), 16555 ISSN:.There is a need for treatments that can safely promote regulatory lymphocyte responses. T cells express GABA receptors (GABAA-Rs) and GABA administration can inhibit Th1-mediated processes such as type 1 diabetes and rheumatoid arthritis in mouse models. Whether GABAA-R agonists can also inhibit Th17-driven processes such as experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS), is an open question. GABA does not pass through the blood-brain barrier (BBB) making it ill-suited to inhibit the spreading of autoreactivity within the CNS. Homotaurine is a BBB-permeable amino acid that antagonizes amyloid fibril formation and was found to be safe but ineffective in long-term Alzheimer's disease clinical trials. Homotaurine also acts as GABAA-R agonist with better pharmacokinetics than that of GABA. Working with both monophasic and relapsing-remitting mouse models of EAE, we show that oral administration of homotaurine can (1) enhance CD8(+)CD122(+)PD-1(+) and CD4(+)Foxp3(+) Treg, but not Breg, responses, (2) inhibit autoreactive Th17 and Th1 responses, and (3) effectively ameliorate ongoing disease. These observations demonstrate the potential of BBB-permeable GABAA-R agonists as a new class of treatment to enhance CD8(+) and CD4(+) Treg responses and limit Th17 and Th1-medaited inflammation in the CNS.
- 30Manzano, S.; Agüera, L.; Aguilar, M.; Olazarán, J. A Review on Tramiprosate (Homotaurine) in Alzheimer’s Disease and Other Neurocognitive Disorders. Front. Neurol. 2020, 11, 614, DOI: 10.3389/fneur.2020.0061430https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jptlSjtA%253D%253D&md5=9b5ddd9c0751224da9c5ace8c3892ed1A Review on Tramiprosate (Homotaurine) in Alzheimer's Disease and Other Neurocognitive DisordersManzano Sagrario; Aguera Luis; Aguilar Miquel; Olazaran JavierFrontiers in neurology (2020), 11 (), 614 ISSN:1664-2295.Alzheimer's disease (AD) is the most prevalent neurodegenerative condition, especially among elderly people. The presence of cortical β-amyloid deposition, together with tau phosphorylation and intracellular accumulation of neurofibrillary tangles (NFT) is the main neuropathologic criteria for AD diagnosis. Additionally, a role of inflammatory, mitochondrial, and metabolic factors has been suggested. Tramiprosate binds to soluble amyloid, thus inhibiting its aggregation in the brain. It reduced oligomeric and fibrillar (plaque) amyloid, diminished hippocampal atrophy, improved cholinergic transmission, and stabilized cognition in preclinical and clinical studies. In this narrative review, current information on the efficacy and safety of tramiprosate, both in AD and in other neurocognitive disorders, is presented. Possible directions for future studies with tramiprosate are also discussed.
- 31Hey, J. A.; Yu, J. Y.; Versavel, M.; Abushakra, S.; Kocis, P.; Power, A.; Kaplan, P. L.; Amedio, J.; Tolar, M. Clinical Pharmacokinetics and Safety of ALZ-801, a Novel Prodrug of Tramiprosate in Development for the Treatment of Alzheimer’s Disease. Clin. Pharmacokinet. 2018, 57 (3), 315– 333, DOI: 10.1007/s40262-017-0608-331https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslWjurbP&md5=7fa4fd8ac6aa9ec857efbdd52aea7d59Clinical Pharmacokinetics and Safety of ALZ-801, a Novel Prodrug of Tramiprosate in Development for the Treatment of Alzheimer's DiseaseHey, John A.; Yu, Jeremy Y.; Versavel, Mark; Abushakra, Susan; Kocis, Petr; Power, Aidan; Kaplan, Paul L.; Amedio, John; Tolar, MartinClinical Pharmacokinetics (2018), 57 (3), 315-333CODEN: CPKNDH; ISSN:0312-5963. (Springer International Publishing AG)ALZ-801 is an orally available, valine-conjugated prodrug of tramiprosate. Tramiprosate, the active agent, is a small-mol. β-amyloid (Aβ) anti-oligomer and aggregation inhibitor that was evaluated extensively in preclin. and clin. investigations for the treatment of Alzheimer's disease (AD). Tramiprosate has been found to inhibit β-amyloid oligomer formation by a multi-ligand enveloping mechanism of action that stabilizes Aβ42 monomers, resulting in the inhibition of formation of oligomers and subsequent aggregation. Although promising as an AD treatment, tramiprosate exhibited two limiting deficiencies: high intersubject pharmacokinetic (PK) variability likely due to extensive gastrointestinal metab., and mild-to-moderate incidence of nausea and vomiting. To address these, we developed an optimized prodrug, ALZ-801, which retains the favorable efficacy attributes of tramiprosate while improving oral PK variability and gastrointestinal tolerability. In this study, we summarize the phase I bridging program to evaluate the safety, tolerability and PK for ALZ-801 after single and multiple rising dose administration in healthy volunteers. Randomized, placebo-controlled, phase I studies in 127 healthy male and female adult and elderly volunteers included [1] a single ascending dose (SAD) study; [2] a 14-day multiple ascending dose (MAD) study; and [3] a single-dose tablet food-effect study. This program was conducted with both a loose-filled capsule and an immediate-release tablet formulation, under both fasted and fed conditions. Safety and tolerability were assessed, and plasma and urine were collected for liq. chromatog.-mass spectrometry (LC-MS) detn. and non-compartmental PK anal. In addn., we defined the target dose of ALZ-801 that delivers a steady-state plasma area under the curve (AUC) exposure of tramiprosate equiv. to that studied in the tramiprosate phase III study. ALZ-801 was well tolerated and there were no severe or serious adverse events (AEs) or lab. findings. The most common AEs were transient mild nausea and some instances of vomiting, which were not dose-related and showed development of tolerance after continued use. ALZ-801 produced dose-dependent max. plasma concn. (Cmax) and AUC exposures of tramiprosate, which were equiv. to that after oral tramiprosate, but with a substantially reduced intersubject variability and a longer elimination half-life. Administration of ALZ-801 with food markedly reduced the incidence of gastrointestinal symptoms compared with the fasted state, without affecting plasma tramiprosate exposure. An immediate-release tablet formulation of ALZ-801 displayed plasma exposure and low variability similar to the loose-filled capsule. ALZ-801 also showed excellent dose-proportionality without accumulation or decrease in plasma exposure of tramiprosate over 14 days. Based on these data, 265 mg of ALZ-801 twice daily was found to achieve a steady-state AUC exposure of tramiprosate equiv. to 150 mg twice daily of oral tramiprosate in the previous phase III trials. ALZ-801, when administered in capsule and tablet forms, showed excellent oral safety and tolerability in healthy adults and elderly volunteers, with significantly improved PK characteristics over oral tramiprosate. A clin. dose of ALZ-801 (265 mg twice daily) was established that achieves the AUC exposure of 150 mg of tramiprosate twice daily, which showed pos. cognitive and functional improvements in apolipoprotein E4/4 homozygous AD patients. These bridging data support the phase III development of ALZ-801 in patients with AD.
- 32A Phase 3, Multicenter, Randomized, Double-Blind, Placebo-Controlled Study of the Efficacy, Safety and Biomarker Effects of ALZ-801 in Subjects With Early Alzheimer’s Disease and APOE4/4 Genotype, ClinicalTrials.gov ID; Clinical trial registration NCT04770220; https://clinicaltrials.gov/ct2/show/NCT04770220 (accessed 2022–07–21).There is no corresponding record for this reference.
- 33Kocis, P.; Tolar, M.; Yu, J.; Sinko, W.; Ray, S.; Blennow, K.; Fillit, H.; Hey, J. A. Elucidating the Aβ42 Anti-Aggregation Mechanism of Action of Tramiprosate in Alzheimer’s Disease: Integrating Molecular Analytical Methods, Pharmacokinetic and Clinical Data. CNS Drugs 2017, 31 (6), 495– 509, DOI: 10.1007/s40263-017-0434-zThere is no corresponding record for this reference.
- 34Hey, J. A.; Kocis, P.; Hort, J.; Abushakra, S.; Power, A.; Vyhnálek, M.; Yu, J. Y.; Tolar, M. Discovery and Identification of an Endogenous Metabolite of Tramiprosate and Its Prodrug ALZ-801 That Inhibits Beta Amyloid Oligomer Formation in the Human Brain. CNS Drugs 2018, 32 (9), 849– 861, DOI: 10.1007/s40263-018-0554-0There is no corresponding record for this reference.
- 35Liang, C.; Savinov, S. N.; Fejzo, J.; Eyles, S. J.; Chen, J. Modulation of Amyloid-Β42 Conformation by Small Molecules Through Nonspecific Binding. J. Chem. Theory Comput. 2019, 15 (10), 5169– 5174, DOI: 10.1021/acs.jctc.9b0059935https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12jt77L&md5=30ffad7b37865a67ad8ef17affc0df02Modulation of Amyloid-β42 Conformation by Small Molecules Through Nonspecific BindingLiang, Chungwen; Savinov, Sergey N.; Fejzo, Jasna; Eyles, Stephen J.; Chen, JianhanJournal of Chemical Theory and Computation (2019), 15 (10), 5169-5174CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Aggregation of amyloid-β (Aβ) peptides is a crucial step in the progression of Alzheimer's disease (AD). Identifying aggregation inhibitors against AD has been a great challenge. We report an atomistic simulation study of the inhibition mechanism of two small mols., homotaurine and scyllo-inositol, which are AD drug candidates currently under investigation. We show that both small mols. promote a conformational change of the Aβ42 monomer toward a more collapsed phase through a nonspecific binding mechanism. This finding provides atomistic-level insights into designing potential drug candidates for future AD treatments.
- 36Hanwell, M. D.; Curtis, D. E.; Lonie, D. C.; Vandermeersch, T.; Zurek, E.; Hutchison, G. R. Avogadro: An Advanced Semantic Chemical Editor, Visualization, and Analysis Platform. J. Cheminf. 2012, 4 (1), 17, DOI: 10.1186/1758-2946-4-1736https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVGksLg%253D&md5=f10400f51db314afa780e99403ca748aAvogadro: an advanced semantic chemical editor, visualization, and analysis platformHanwell, Marcus D.; Curtis, Donald E.; Lonie, David C.; Vandermeersch, Tim; Zurek, Eva; Hutchison, Geoffrey R.Journal of Cheminformatics (2012), 4 (), 17CODEN: JCOHB3; ISSN:1758-2946. (Chemistry Central Ltd.)Background: The Avogadro project has developed an advanced mol. editor and visualizer designed for cross-platform use in computational chem., mol. modeling, bioinformatics, materials science, and related areas. It offers flexible, high quality rendering, and a powerful plugin architecture. Typical uses include building mol. structures, formatting input files, and analyzing output of a wide variety of computational chem. packages. By using the CML file format as its native document type, Avogadro seeks to enhance the semantic accessibility of chem. data types. Results: The work presented here details the Avogadro library, which is a framework providing a code library and application programming interface (API) with three-dimensional visualization capabilities; and has direct applications to research and education in the fields of chem., physics, materials science, and biol. The Avogadro application provides a rich graphical interface using dynamically loaded plugins through the library itself. The application and library can each be extended by implementing a plugin module in C++ or Python to explore different visualization techniques, build/manipulate mol. structures, and interact with other programs. We describe some example extensions, one which uses a genetic algorithm to find stable crystal structures, and one which interfaces with the PackMol program to create packed, solvated structures for mol. dynamics simulations. The 1.0 release series of Avogadro is the main focus of the results discussed here. Conclusions: Avogadro offers a semantic chem. builder and platform for visualization and anal. For users, it offers an easy-to-use builder, integrated support for downloading from common databases such as PubChem and the Protein Data Bank, extg. chem. data from a wide variety of formats, including computational chem. output, and native, semantic support for the CML file format. For developers, it can be easily extended via a powerful plugin mechanism to support new features in org. chem., inorg. complexes, drug design, materials, biomols., and simulations.
- 37Frisch, M. J.; Trucks, G. W.; Schlegel, H. B.; Scuseria, G. E.; Robb, M. A.; Cheeseman, J. R.; Scalmani, G.; Barone, V.; Mennucci, B.; Petersson, G. A., Gaussian 09. In Revision E.01, Gaussian, Inc., 2009.There is no corresponding record for this reference.
- 38Case, D. A.; Betz, R. M.; Cerutti, D. S.; Cheatham, III, T. E.; Darden, T. A.; Duke, R. E.; Giese, T. J.; Gohlke, H.; Goetz, A. W.; Homeyer, N., ; AMBER 16, University of California, San Francisco, 2016.There is no corresponding record for this reference.
- 39Rose, P. W.; Bi, C.; Bluhm, W. F.; Christie, C. H.; Dimitropoulos, D.; Dutta, S.; Green, R. K.; Goodsell, D. S.; Prlić, A.; Quesada, M. The RCSB Protein Data Bank: New Resources for Research and Education. Nucleic Acids Res. 2012, 41 (D1), D475– D482, DOI: 10.1093/nar/gks1200There is no corresponding record for this reference.
- 40Bas, D. C.; Rogers, D. M.; Jensen, J. H. Very fast prediction and rationalization of pKa values for protein–ligand complexes. Proteins: Struct., Funct., Bioinf. 2008, 73 (3), 765– 783, DOI: 10.1002/prot.2210240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtlCgsbjO&md5=34f63cf947000be5482b745675ff8a8aVery fast prediction and rationalization of pKa values for protein-ligand complexesBas, Delphine C.; Rogers, David M.; Jensen, Jan H.Proteins: Structure, Function, and Bioinformatics (2008), 73 (3), 765-783CODEN: PSFBAF ISSN:. (Wiley-Liss, Inc.)The PROPKA method for the prediction of the pKa values of ionizable residues in proteins is extended to include the effect of non-proteinaceous ligands on protein pKa values as well as predict the change in pKa values of ionizable groups on the ligand itself. This new version of PROPKA (PROPKA 2.0) is, as much as possible, developed by adapting the empirical rules underlying PROPKA 1.0 to ligand functional groups. Thus, the speed of PROPKA is retained, so that the pKa values of all ionizable groups are computed in a matter of seconds for most proteins. This adaptation is validated by comparing PROPKA 2.0 predictions to exptl. data for 26 protein-ligand complexes including trypsin, thrombin, three pepsins, HIV-1 protease, chymotrypsin, xylanase, hydroxynitrile lyase, and dihydrofolate reductase. For trypsin and thrombin, large protonation state changes (|n| > 0.5) have been obsd. exptl. for 4 out of 14 ligand complexes. PROPKA 2.0 and Klebe's PEOE approach both identify three of the four large protonation state changes. The protonation state changes due to plasmepsin II, cathepsin D and endothiapepsin binding to pepstatin are predicted to within 0.4 proton units at pH 6.5 and 7.0, resp. The PROPKA 2.0 results indicate that structural changes due to ligand binding contribute significantly to the proton uptake/release, as do residues far away from the binding site, primarily due to the change in the local environment of a particular residue and hence the change in the local hydrogen bonding network. Overall the results suggest that PROPKA 2.0 provides a good description of the protein-ligand interactions that have an important effect on the pKa values of titratable groups, thereby permitting fast and accurate detn. of the protonation states of key residues and ligand functional groups within the binding or active site of a protein.
- 41Doerr, S.; Harvey, M. J.; Noé, F.; De Fabritiis, G. HTMD: High-Throughput Molecular Dynamics for Molecular Discovery. J. Chem. Theory Comput. 2016, 12 (4), 1845– 1852, DOI: 10.1021/acs.jctc.6b0004941https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xjs12hu7o%253D&md5=c7ce0d9a709642ad5e28e202655a9a9dHTMD: High-Throughput Molecular Dynamics for Molecular DiscoveryDoerr, S.; Harvey, M. J.; Noe, Frank; De Fabritiis, G.Journal of Chemical Theory and Computation (2016), 12 (4), 1845-1852CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Recent advances in mol. simulations have allowed scientists to investigate slower biol. processes than ever before. Together with these advances came an explosion of data that has transformed a traditionally computing-bound into a data-bound problem. Here, we present HTMD, a programmable, extensible platform written in Python that aims to solve the data generation and anal. problem as well as increase reproducibility by providing a complete workspace for simulation-based discovery. So far, HTMD includes system building for CHARMM and AMBER force fields, projection methods, clustering, mol. simulation prodn., adaptive sampling, an Amazon cloud interface, Markov state models, and visualization. As a result, a single, short HTMD script can lead from a PDB structure to useful quantities such as relaxation time scales, equil. populations, metastable conformations, and kinetic rates. In this paper, we focus on the adaptive sampling and Markov state modeling features.
- 42Huang, J.; Rauscher, S.; Nawrocki, G.; Ran, T.; Feig, M.; de Groot, B. L.; Grubmüller, H.; MacKerell, A. D. CHARMM36m: An Improved Force Field for Folded and Intrinsically Disordered Proteins. Nat. Methods 2017, 14 (1), 71– 73, DOI: 10.1038/nmeth.406742https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVSiu77I&md5=0aa151fbef2ee0b5e2cfb593c54330c2CHARMM36m: an improved force field for folded and intrinsically disordered proteinsHuang, Jing; Rauscher, Sarah; Nawrocki, Grzegorz; Ran, Ting; Feig, Michael; de Groot, Bert L.; Grubmuller, Helmut; MacKerell, Alexander D. JrNature Methods (2017), 14 (1), 71-73CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The all-atom additive CHARMM36 protein force field is widely used in mol. modeling and simulations. We present its refinement, CHARMM36m (http://mackerell.umaryland.edu/charmm_ff.shtml), with improved accuracy in generating polypeptide backbone conformational ensembles for intrinsically disordered peptides and proteins.
- 43Swails, J.; ParmEd, GitHub, Inc, 2010. https://github.com/ParmEd/ParmEd. (accessed 2018–03–08).There is no corresponding record for this reference.
- 44Roe, D. R.; Cheatham, T. E. PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. J. Chem. Theory Comput. 2013, 9 (7), 3084– 3095, DOI: 10.1021/ct400341p44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptFehtr8%253D&md5=6f1bee934f13f180bd7e1feb6b78036dPTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory DataRoe, Daniel R.; Cheatham, Thomas E.Journal of Chemical Theory and Computation (2013), 9 (7), 3084-3095CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We describe PTRAJ and its successor CPPTRAJ, two complementary, portable, and freely available computer programs for the anal. and processing of time series of three-dimensional at. positions (i.e., coordinate trajectories) and the data therein derived. Common tools include the ability to manipulate the data to convert among trajectory formats, process groups of trajectories generated with ensemble methods (e.g., replica exchange mol. dynamics), image with periodic boundary conditions, create av. structures, strip subsets of the system, and perform calcns. such as RMS fitting, measuring distances, B-factors, radii of gyration, radial distribution functions, and time correlations, among other actions and analyses. Both the PTRAJ and CPPTRAJ programs and source code are freely available under the GNU General Public License version 3 and are currently distributed within the AmberTools 12 suite of support programs that make up part of the Amber package of computer programs (see http://ambermd.org). This overview describes the general design, features, and history of these two programs, as well as algorithmic improvements and new features available in CPPTRAJ.
- 45Aqvist, J.; Medina, C.; Samuelsson, J. E. A New Method for Predicting Binding Affinity in Computer-Aided Drug Design. Protein Eng. 1994, 7 (3), 385– 391, DOI: 10.1093/protein/7.3.38545https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK2c3jtlSnsw%253D%253D&md5=c45572201328309d4fc849844d1c3b71A new method for predicting binding affinity in computer-aided drug designAqvist J; Medina C; Samuelsson J EProtein engineering (1994), 7 (3), 385-91 ISSN:0269-2139.A new semi-empirical method for calculating free energies of binding from molecular dynamics (MD) simulations is presented. It is based on standard thermodynamic cycles and on a linear approximation of polar and non-polar free energy contributions from the corresponding MD averages. The method is tested on a set of endothiapepsin inhibitors and found to give accurate results both for absolute as well as relative free energies.
- 46Kabsch, W.; Sander, C. Dictionary of Protein Secondary Structure: Pattern Recognition of Hydrogen-Bonded and Geometrical Features. Biopolymers 1983, 22 (12), 2577– 2637, DOI: 10.1002/bip.36022121146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2cXkslegtQ%253D%253D&md5=a146e923e6c49cb542d0ad24a399d0f0Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical featuresKabsch, Wolfgang; Sander, ChristianBiopolymers (1983), 22 (12), 2577-637CODEN: BIPMAA; ISSN:0006-3525.For a successful anal. of the relation between amino acid sequence and protein structure, an unambiguous and phys. meaningful definition of secondary structure is essential. A set of simple and phys. motivated criteria for secondary structure, programmed as a pattern-recognition process of H-bonded and geometrical features extd. from x-ray coordinates were developed. Cooperative secondary structure is recognized as repeats of the elementary H-bonding patterns turn and bridge. Repeating turns are helixes, repeating bridges are ladders, connected ladders are sheets. Geometric structure is defined in terms of the concepts torsion and curvature of differential geometry. Local chain chirality is the torsional handedness of 4 consecutive Cα positions and is pos. for right-handed helixes and neg. for ideal twisted β-sheets. Curved pieces are defined as bends. Solvent exposure is given as the no. of H2O mols. in possible contact with a residue. The end result is a compilation of the primary structure, including SS bonds, secondary structure, and solvent exposure of 62 different globular proteins. The presentation is in linear form: strip graphs for an overall view and strip tables for the details of each of 10,925 residues. The dictionary is also available in computer-readable form for protein structure prediction work.
- 47Miller, B. R.; McGee, T. D.; Swails, J. M.; Homeyer, N.; Gohlke, H.; Roitberg, A. E. MMPBSA.Py: An Efficient Program for End-State Free Energy Calculations. J. Chem. Theory Comput. 2012, 8, 3314– 3321, DOI: 10.1021/ct300418h47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtV2gtrzP&md5=cc4148bd8f70c7cad94fd3ec6f580e52MMPBSA.py: An Efficient Program for End-State Free Energy CalculationsMiller, Bill R., III; McGee, T. Dwight, Jr.; Swails, Jason M.; Homeyer, Nadine; Gohlke, Holger; Roitberg, Adrian E.Journal of Chemical Theory and Computation (2012), 8 (9), 3314-3321CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)MM-PBSA is a post-processing end-state method to calc. free energies of mols. in soln. MMPBSA.py is a program written in Python for streamlining end-state free energy calcns. using ensembles derived from mol. dynamics (MD) or Monte Carlo (MC) simulations. Several implicit solvation models are available with MMPBSA.py, including the Poisson-Boltzmann Model, the Generalized Born Model, and the Ref. Interaction Site Model. Vibrational frequencies may be calcd. using normal mode or quasi-harmonic anal. to approx. the solute entropy. Specific interactions can also be dissected using free energy decompn. or alanine scanning. A parallel implementation significantly speeds up the calcn. by dividing frames evenly across available processors. MMPBSA.py is an efficient, user-friendly program with the flexibility to accommodate the needs of users performing end-state free energy calcns. The source code can be downloaded at http://ambermd.org/ with AmberTools, released under the GNU General Public License.
- 48Genheden, S.; Ryde, U. The MM/PBSA and MM/GBSA Methods to Estimate Ligand-Binding Affinities. Expert Opin. Drug Discovery 2015, 10 (5), 449– 461, DOI: 10.1517/17460441.2015.103293648https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntFGktr8%253D&md5=b123b88809f275564f95a2271ebd159fThe MM/PBSA and MM/GBSA methods to estimate ligand-binding affinitiesGenheden, Samuel; Ryde, UlfExpert Opinion on Drug Discovery (2015), 10 (5), 449-461CODEN: EODDBX; ISSN:1746-0441. (Informa Healthcare)Introduction: The mol. mechanics energies combined with the Poisson-Boltzmann or generalized Born and surface area continuum solvation (MM/PBSA and MM/GBSA) methods are popular approaches to est. the free energy of the binding of small ligands to biol. macromols. They are typically based on mol. dynamics simulations of the receptor-ligand complex and are therefore intermediate in both accuracy and computational effort between empirical scoring and strict alchem. perturbation methods. They have been applied to a large no. of systems with varying success. Areas covered: The authors review the use of MM/PBSA and MM/GBSA methods to calc. ligand-binding affinities, with an emphasis on calibration, testing and validation, as well as attempts to improve the methods, rather than on specific applications. Expert opinion: MM/PBSA and MM/GBSA are attractive approaches owing to their modular nature and that they do not require calcns. on a training set. They have been used successfully to reproduce and rationalize exptl. findings and to improve the results of virtual screening and docking. However, they contain several crude and questionable approxns., for example, the lack of conformational entropy and information about the no. and free energy of water mols. in the binding site. Moreover, there are many variants of the method and their performance varies strongly with the tested system. Likewise, most attempts to ameliorate the methods with more accurate approaches, for example, quantum-mech. calcns., polarizable force fields or improved solvation have deteriorated the results.
- 49Wu, H.; Noé, F. Variational Approach for Learning Markov Processes from Time Series Data. J. Nonlinear Sci. 2020, 30 (1), 23– 66, DOI: 10.1007/s00332-019-09567-yThere is no corresponding record for this reference.
- 50Mardt, A.; Pasquali, L.; Noé, F.; Wu, H. Deep Learning Markov and Koopman Models with Physical Constraints. In Proceedings of The First Mathematical and Scientific Machine Learning Conference; PMLR, 2020; pp. 451 475.There is no corresponding record for this reference.
- 51Klambauer, G.; Unterthiner, T.; Mayr, A.; Hochreiter, S.; Self-Normalizing Neural Networks. In Advances in Neural Information Processing Systems, Curran Associates, Inc, 2017, Vol. 30.There is no corresponding record for this reference.
- 52MacQueen, J. Some Methods for Classification and Analysis of Multivariate Observations Proceedings of the Fifth Berkeley Symposium On Mathematical Statistics And Probability, Volume 1: Statistics Le Cam, L. M.; Neyman, J. Project Euclid 1967, 5; 281 298.There is no corresponding record for this reference.
- 53Bonneel, N.; van de Panne, M.; Paris, S.; Heidrich, W. Displacement Interpolation Using Lagrangian Mass Transport. ACM Trans. Graph. 2011, 30 (6), 1– 12, DOI: 10.1145/2070781.2024192There is no corresponding record for this reference.
- 54Kuhn, H. W. The Hungarian Method for the Assignment Problem. Naval Res. Logistics Quarterly 1955, 2 (1–2), 83– 97, DOI: 10.1002/nav.3800020109There is no corresponding record for this reference.
- 55Fong, R.; Vedaldi, A. Explanations for Attributing Deep Neural Network Predictions. In Explainable AI: Interpreting, Explaining and Visualizing Deep Learning. In Lecture Notes in Computer Science, Samek, W.; Montavon, G.; Vedaldi, A.; Hansen, L. K.; Müller, K.-R.; Springer International Publishing: Cham, 2019; pp. 149 167. DOI: DOI: 10.1007/978-3-030-28954-6_8 .There is no corresponding record for this reference.
- 56Simonyan, K.; Vedaldi, A.; Zisserman, A. Deep Inside Convolutional Networks: Visualising Image Classification Models and Saliency Maps, arXiv 2014 DOI: 10.48550/arXiv.1312.6034 .There is no corresponding record for this reference.
- 57Cohen, S. I. A.; Linse, S.; Luheshi, L. M.; Hellstrand, E.; White, D. A.; Rajah, L.; Otzen, D. E.; Vendruscolo, M.; Dobson, C. M.; Knowles, T. P. J. Proliferation of Amyloid-Β42 Aggregates Occurs through a Secondary Nucleation Mechanism. Proc. Natl. Acad. Sci. U. S. A. 2013, 110 (24), 9758– 9763, DOI: 10.1073/pnas.121840211057https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFOrt7fJ&md5=d9db3cfc7e3004e5cdc309a92d2c7431Proliferation of amyloid-β42 aggregates occurs through a secondary nucleation mechanismCohen, Samuel I. A.; Linse, Sara; Luheshi, Leila M.; Hellstrand, Erik; White, Duncan A.; Rajah, Luke; Otzen, Daniel E.; Vendruscolo, Michele; Dobson, Christopher M.; Knowles, Tuomas P. J.Proceedings of the National Academy of Sciences of the United States of America (2013), 110 (24), 9758-9763, S9758/1-S9758/11CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The generation of toxic oligomers during the aggregation of the amyloid-β (Aβ) peptide Aβ42 into amyloid fibrils and plaques has emerged as a central feature of the onset and progression of Alzheimer's disease, but the mol. pathways that control pathol. aggregation have proved challenging to identify. Here, the authors used a combination of kinetic studies, selective radiolabeling expts., and cell viability assays to detect directly the rates of formation of both fibrils and oligomers and the resulting cytotoxic effects. The results showed that once a small but crit. concn. of amyloid fibrils had accumulated, the toxic oligomeric species were predominantly formed from monomeric peptide mols. through a fibril-catalyzed secondary nucleation reaction, rather than through a classical mechanism of homogeneous primary nucleation. This catalytic mechanism coupled together the growth of insol. amyloid fibrils and the generation of diffusible oligomeric aggregates that are implicated as neurotoxic agents in Alzheimer's disease. These results revealed that the aggregation of Aβ42 is promoted by a pos. feedback loop that originates from the interactions between the monomeric and fibrillar forms of this peptide. These findings bring together the main mol. species implicated in the Aβ aggregation cascade and suggest that perturbation of the secondary nucleation pathway identified in this study could be an effective strategy to control the proliferation of neurotoxic Aβ42 oligomers.
- 58Arosio, P.; Knowles, T. P. J.; Linse, S. On the Lag Phase in Amyloid Fibril Formation. Phys. Chem. Chem. Phys. 2015, 17 (12), 7606– 7618, DOI: 10.1039/C4CP05563B58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitVCrsbo%253D&md5=552e3a6a573bf26a0c125cb4bdb2fc19On the lag phase in amyloid fibril formationArosio, Paolo; Knowles, Tuomas P. J.; Linse, SaraPhysical Chemistry Chemical Physics (2015), 17 (12), 7606-7618CODEN: PPCPFQ; ISSN:1463-9076. (Royal Society of Chemistry)A review. The formation of nanoscale amyloid fibrils from normally sol. peptides and proteins is a common form of self-assembly phenomenon that has fundamental connections with biol. functions and human diseases. The kinetics of this process has been widely studied and exhibits on a macroscopic level three characteristic stages: (1) a lag phase; (2) a growth phase; and (3) a final plateau regime. The question of which mol. events take place during each one of these phases has been a central element in the quest for a mechanism of amyloid formation. Here, the authors discuss the nature and mol. origin of the lag-phase in amyloid formation by making use of tools and concepts from phys. chem., in particular from chem. reaction kinetics. The authors discuss how, in macroscopic samples, it has become apparent that the lag-phase is not a waiting time for nuclei to form. Rather, multiple parallel processes exist and typically millions of primary nuclei form during the lag phase from monomers in soln. Thus, the lag-time represents a time that is required for the nuclei that are formed early on in the reaction to grow and proliferate in order to reach an aggregate concn. that is readily detected in bulk assays. In many cases, this proliferation takes place through secondary nucleation, where fibrils may present a catalytic surface for the formation of new aggregates. Fibrils may also break (fragmentation) and thereby provide new ends for elongation. Thus, at least 2 (primary nucleation and elongation) and in many systems at least 4 (primary nucleation, elongation, secondary nucleation, and fragmentation) microscopic processes occur during the lag phase. Moreover, these same processes occur during all 3 phases of the macroscopic aggregation process, albeit at different rates as governed by rate consts. and by the concn. of reacting species at each point in time.
- 59Tomaselli, S.; Esposito, V.; Vangone, P.; van Nuland, N. A. J.; Bonvin, A. M. J. J.; Guerrini, R.; Tancredi, T.; Temussi, P. A.; Picone, D. The α-to-β Conformational Transition of Alzheimer’s Aβ-(1–42) Peptide in Aqueous Media Is Reversible: A Step by Step Conformational Analysis Suggests the Location of β Conformation Seeding. ChemBiochem 2006, 7 (2), 257– 267, DOI: 10.1002/cbic.20050022359https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhs1aktrc%253D&md5=aa0da86f9a3b30ff357490ae46dff8d8The α-to-β conformational transition of Alzheimer's Aβ-(1-42) peptide in aqueous media is reversible: a step by step conformational analysis suggests the location of β conformation seedingTomaselli, Simona; Esposito, Veronica; Vangone, Paolo; van Nuland, Nico A. J.; Bonvin, Alexandre M. J. J.; Guerrini, Remo; Tancredi, Teodorico; Temussi, Piero A.; Picone, DeliaChemBioChem (2006), 7 (2), 257-267CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)Current views of the role of β-amyloid (Aβ) peptide fibrils range from regarding them as the cause of Alzheimer's pathol. to having a protective function. In the last few years, it has also been suggested that sol. oligomers might be the most important toxic species. In all cases, the study of the conformational properties of Aβ peptides in sol. form constitutes a basic approach to the design of mols. with "antiamyloid" activity. We exptl. investigated the conformational 'path' that can lead the Aβ-(1-42) peptide from the native state, which is represented by an α helix embedded in the membrane, to the final state in the amyloid fibrils, which is characterized by sheet structures. The conformational steps were monitored by using CD and NMR spectroscopy in media of varying polarities. This was achieved by changing the compn. of water and hexafluoroisopropanol (HFIP). In the presence of HFIP, β conformations can be obsd. in solns. that have very high water content (up to 99% water; vol./vol.). These can be turned back to α helixes simply by adding the appropriated amt. of HFIP. The transition of Aβ-(1-42) from α to β conformation occurs when the amt. of water is higher that 80% (vol./vol.). The NMR structure solved in HFIP/H2O with high water content showed that, on going from very apolar to polar environments, the long N-terminal helix is essentially retained, whereas the shorter C-terminal helix is lost. The complete conformational path was investigated in detail with the aid of mol. dynamics simulations in explicit solvent, which led to the localization of residues that might seed β conformations. The structures obtained might help to find regions that are more affected by environmental conditions in vivo. This could in turn aid the design of mols. able to inhibit fibril deposition or revert oligomerization processes.
- 60Shamsi, Z.; Moffett, A. S.; Shukla, D. Enhanced Unbiased Sampling of Protein Dynamics Using Evolutionary Coupling Information. Sci. Rep. 2017, 7 (1), 12700, DOI: 10.1038/s41598-017-12874-7There is no corresponding record for this reference.
- 61Zimmerman, M. I.; Porter, J. R.; Sun, X.; Silva, R. R.; Bowman, G. R. Choice of Adaptive Sampling Strategy Impacts State Discovery, Transition Probabilities, and the Apparent Mechanism of Conformational Changes. J. Chem. Theory Comput. 2018, 14 (11), 5459– 5475, DOI: 10.1021/acs.jctc.8b0050061https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslGqsLnI&md5=b770c1d634b1d7fc7cc8e0b9fb3b3db8Choice of Adaptive Sampling Strategy Impacts State Discovery, Transition Probabilities, and the Apparent Mechanism of Conformational ChangesZimmerman, Maxwell I.; Porter, Justin R.; Sun, Xianqiang; Silva, Roseane R.; Bowman, Gregory R.Journal of Chemical Theory and Computation (2018), 14 (11), 5459-5475CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Interest in atomically detailed simulations has grown significantly with recent advances in computational hardware and Markov state modeling (MSM) methods, yet outstanding questions remain that hinder their widespread adoption. Namely, how do alternative sampling strategies explore conformational space and how might this influence predictions generated from the data. Here, we seek to answer these questions for four commonly used sampling methods: (1) a single long simulation, (2) many short simulations run in parallel, (3) adaptive sampling, and (4) our recently developed goal-oriented sampling algorithm, FAST. We first develop a theor. framework for anal. calcg. the probability of discovering select states on simple landscapes, where we uncover the drastic effects of varying the no. and length of simulations. We then use kinetic Monte Carlo simulations on a variety of phys. inspired landscapes to characterize the probability of discovering particular states and transition pathways for each of the four methods. Consistently, we find that FAST simulations discover each target state with the highest probability, while traversing realistic pathways. Furthermore, we uncover the potential pathol. that short parallel simulations sometimes predict an incorrect transition pathway by crossing large energy barriers that long simulations would typically circumnavigate. We refer to this pathol. as "pathway tunneling". To protect against this phenomenon when using adaptive-sampling and FAST simulations, we introduce the FAST-string method. This method enhances sampling along the highest-flux transition paths to refine an MSMs transition probabilities and discriminate between competing pathways. Addnl., we compare the performance of a variety of MSM estimators in describing accurate thermodn. and kinetics. For adaptive sampling, we recommend simply normalizing the transition counts out of each state after adding small pseudocounts to avoid creating sources or sinks. Lastly, we evaluate whether our insights from simple landscapes hold for all-atom mol. dynamics simulations of the folding of the λ-repressor protein. Remarkably, we find that FAST-contacts predicts the same folding pathway as a set of long simulations but with orders of magnitude less simulation time.
- 62Betz, R. M.; Dror, R. O. How Effectively Can Adaptive Sampling Methods Capture Spontaneous Ligand Binding?. J. Chem. Theory Comput. 2019, 15 (3), 2053– 2063, DOI: 10.1021/acs.jctc.8b0091362https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1agtrY%253D&md5=36f1ee6880504ea87266dce4a3402169How Effectively Can Adaptive Sampling Methods Capture Spontaneous Ligand Binding?Betz, Robin M.; Dror, Ron O.Journal of Chemical Theory and Computation (2019), 15 (3), 2053-2063CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. dynamics (MD) simulations that capture the spontaneous binding of drugs and other ligands to their target proteins can reveal a great deal of useful information, but most drug-like ligands bind on time scales longer than those accessible to individual MD simulations. Adaptive sampling methods-in which one performs multiple rounds of simulation, with the initial conditions of each round based on the results of previous rounds-offer a promising potential soln. to this problem. No comprehensive anal. of the performance gains from adaptive sampling is available for ligand binding, however, particularly for protein-ligand systems typical of those encountered in drug discovery. Moreover, most previous work presupposes knowledge of the ligand's bound pose. Here the authors outline existing methods for adaptive sampling of the ligand-binding process and introduce several improvements, with a focus on methods that do not require prior knowledge of the binding site or bound pose. The authors then evaluate these methods by comparing them to traditional, long MD simulations for realistic protein-ligand systems. The authors find that adaptive sampling simulations typically fail to reach the bound pose more efficiently than traditional MD. However, adaptive sampling identifies multiple potential binding sites more efficiently than traditional MD and also provides better characterization of binding pathways. The authors explain these results by showing that protein-ligand binding is an example of an exploration-exploitation dilemma. Existing adaptive sampling methods for ligand binding in the absence of a known bound pose vastly favor the broad exploration of protein-ligand space, sometimes failing to sufficiently exploit intermediate states as they are discovered. The authors suggest potential avenues for future research to address this shortcoming.
- 63Kleiman, D. E.; Nadeem, H.; Shukla, D. Adaptive Sampling Methods for Molecular Dynamics in the Era of Machine Learning. J. Phys. Chem. B 2023, 127 (50), 10669– 10681, DOI: 10.1021/acs.jpcb.3c04843There is no corresponding record for this reference.
- 64Man, V. H.; He, X.; Derreumaux, P.; Ji, B.; Xie, X.-Q.; Nguyen, P. H.; Wang, J. Effects of All-Atom Molecular Mechanics Force Fields on Amyloid Peptide Assembly: The Case of Aβ16–22 Dimer. J. Chem. Theory Comput. 2019, 15 (2), 1440– 1452, DOI: 10.1021/acs.jctc.8b0110764https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlaqsA%253D%253D&md5=b7d65b1c4eeb166b27ab435687bb2528Effects of all-atom molecular mechanics force fields on amyloid peptide assembly: The case of Aβ16-22 dimerMan, Viet Hoang; He, Xibing; Derreumaux, Philippe; Ji, Beihong; Xie, Xiang-Qun; Nguyen, Phuong H.; Wang, JunmeiJournal of Chemical Theory and Computation (2019), 15 (2), 1440-1452CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)We investigated the effects of 17 widely used atomistic mol. mechanics force fields (MMFFs) on the structures and kinetics of amyloid peptide assembly. To this end, we performed large-scale all-atom mol. dynamics simulations in explicit water on the dimer of the 7-residue fragment of Alzheimer amyloid-β peptide, Aβ16-22, for a total time of 0.34 ms. We compared the effects of these MMFFs by analyzing various global reaction coordinates, secondary structure contents, the fibril population, the in-register and out-of-register architectures, and the fibril formation time at 310 K. While the AMBER94, AMBER99, and AMBER12SB force fields did not predict any β-sheets, the 7 force fields (AMBER96, GROMOS45a3, GROMOS53a5, GROMOS53a6, GROMOS43a1, GROMOS43a2, and GROMOS54a7) formed β-sheets rapidly. In contrast, the following 5 force fields (AMBER99-ILDN, AMBER14SB, CHARMM22*, CHARMM36, and CHARMM36m) were the best candidates for studying amyloid peptide assembly, as they provided good balances in terms of structures and kinetics. We also investigated the assembly mechanisms of dimeric Aβ16-22 and found that the fibril formation rate was predominantly controlled by the total β-strand content.
- 65Maier, J. A.; Martinez, C.; Kasavajhala, K.; Wickstrom, L.; Hauser, K. E.; Simmerling, C. ff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SB. J. Chem. Theory Comput. 2015, 11 (8), 3696– 3713, DOI: 10.1021/acs.jctc.5b0025565https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFequ7rN&md5=7b803577b3b6912cc6750cfbd356596eff14SB: Improving the Accuracy of Protein Side Chain and Backbone Parameters from ff99SBMaier, James A.; Martinez, Carmenza; Kasavajhala, Koushik; Wickstrom, Lauren; Hauser, Kevin E.; Simmerling, CarlosJournal of Chemical Theory and Computation (2015), 11 (8), 3696-3713CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Mol. mechanics is powerful for its speed in atomistic simulations, but an accurate force field is required. The Amber ff99SB force field improved protein secondary structure balance and dynamics from earlier force fields like ff99, but weaknesses in side chain rotamer and backbone secondary structure preferences have been identified. Here, we performed a complete refit of all amino acid side chain dihedral parameters, which had been carried over from ff94. The training set of conformations included multidimensional dihedral scans designed to improve transferability of the parameters. Improvement in all amino acids was obtained as compared to ff99SB. Parameters were also generated for alternate protonation states of ionizable side chains. Av. errors in relative energies of pairs of conformations were under 1.0 kcal/mol as compared to QM, reduced 35% from ff99SB. We also took the opportunity to make empirical adjustments to the protein backbone dihedral parameters as compared to ff99SB. Multiple small adjustments of φ and ψ parameters were tested against NMR scalar coupling data and secondary structure content for short peptides. The best results were obtained from a phys. motivated adjustment to the φ rotational profile that compensates for lack of ff99SB QM training data in the β-ppII transition region. Together, these backbone and side chain modifications (hereafter called ff14SB) not only better reproduced their benchmarks, but also improved secondary structure content in small peptides and reprodn. of NMR χ1 scalar coupling measurements for proteins in soln. We also discuss the Amber ff12SB parameter set, a preliminary version of ff14SB that includes most of its improvements.
- 66Harada, T.; Kuroda, R. CD Measurements of β-Amyloid (1–40) and (1–42) in the Condensed Phase. Biopolymers 2011, 95 (2), 127– 134, DOI: 10.1002/bip.21543There is no corresponding record for this reference.
- 67Löhr, T.; Kohlhoff, K.; Heller, G. T.; Camilloni, C.; Vendruscolo, M. A Small Molecule Stabilizes the Disordered Native State of the Alzheimer’s Aβ Peptide. ACS Chem. Neurosci. 2022, 13 (12), 1738– 1745, DOI: 10.1021/acschemneuro.2c0011667https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsVSksbnM&md5=6b68e3699c6b553694d09f306c68da96A Small Molecule Stabilizes the Disordered Native State of the Alzheimer's Aβ PeptideLohr, Thomas; Kohlhoff, Kai; Heller, Gabriella T.; Camilloni, Carlo; Vendruscolo, MicheleACS Chemical Neuroscience (2022), 13 (12), 1738-1745CODEN: ACNCDM; ISSN:1948-7193. (American Chemical Society)The stabilization of native states of proteins is a powerful drug discovery strategy. It is still unclear, however, whether this approach can be applied to intrinsically disordered proteins. Here, the authors report a small mol. that stabilizes the native state of the Aβ42 peptide, an intrinsically disordered protein fragment assocd. with Alzheimer's disease. This stabilization takes place by a dynamic binding mechanism, in which both the small mol. and the Aβ42 peptide remain disordered. This disordered binding mechanism involves enthalpically favorable local -stacking interactions coupled with entropically advantageous global effects. Small mols. can stabilize disordered proteins in their native states through transient non-specific interactions that provide enthalpic gain while simultaneously increasing the conformational entropy of the proteins.
- 68Reddy, G.; Straub, J. E.; Thirumalai, D. Influence of Preformed Asp23-Lys28 Salt Bridge on the Conformational Fluctuations of Monomers and Dimers of Aβ Peptides with Implications for Rates of Fibril Formation. J. Phys. Chem. B 2009, 113 (4), 1162– 1172, DOI: 10.1021/jp808914c68https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXisVWqtA%253D%253D&md5=9da47509c0dc01142cad5d009c9040bfInfluence of Preformed Asp23-Lys28 Salt Bridge on the Conformational Fluctuations of Monomers and Dimers of Aβ Peptides with Implications for Rates of Fibril FormationReddy, Govardhan; Straub, John E.; Thirumalai, D.Journal of Physical Chemistry B (2009), 113 (4), 1162-1172CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)Recent expts. have shown that the congener Aβ1-40[D23-K28], in which the side chains of charged residues Asp23 and Lys28 are linked by a lactam bridge, forms amyloid fibrils that are structurally similar to the wild type (WT) Aβ peptide, but at a rate that is nearly 1000 times faster. We used all atom mol. dynamics simulations in explicit water, and two force fields, of the WT dimer, a monomer with the lactam bridge (Aβ10-35-lactam[D23-K28]), and the monomer and dimers with harmonically constrained D23-K28 salt bridge (Aβ10-35[D23-K28]) to understand the origin of the enhanced fibril rate formation. The simulations show that the assembly-competent fibril-like monomer (N*) structure, which is present among the conformations sampled by the isolated monomer, with strand conformations in the residues spanning the N and C termini and a bend involving residues D23VGSNKG29, are populated to a much greater extent in Aβ10-35[D23-K28] and Aβ10-35-lactam[D23-K28] than in the WT, which has negligible probability of forming N*. The salt bridge in N* of Aβ10-35[D23-K28], whose topol. is similar to that found in the fibril, is hydrated. The redn. in the free energy barrier to fibril formation in Aβ10-35[D23-K28] and in Aβ10-35-lactam[D23-K28], compared to the WT, arises largely due to entropic restriction which enables the bend formation. A decrease in the entropy of the unfolded state and the lesser penalty for conformational rearrangement including the formation of the salt bridge in Aβ peptides with D23-K28 constraint results in a redn. in the kinetic barrier in the Aβ1-40-lactam[D23-K28] congener compared to the WT. The decrease in the barrier, which is related to the free energy cost of forming a bend, is estd. to be in the range (4-7)kBT. Although a no. of factors det. the growth of fibrils, the decrease in the free energy barrier, relative to the WT, to N* formation is a major factor in the rate enhancement in the fibril formation of Aβ1-40[D23-K28] congener. Qual. similar results were obtained using simulations of Aβ9-40 peptides and various constructs related to the Aβ10-35 systems that were probed using OPLS and CHARMM force fields. We hypothesize that mutations or other constraints that preferentially enhance the population of the N* species would speed up aggregation rates. Conversely, ligands that lock it in the fibril-like N* structure would prevent amyloid formation.
- 69Chandra, B.; Bhowmik, D.; Maity, B. K.; Mote, K. R.; Dhara, D.; Venkatramani, R.; Maiti, S.; Madhu, P. K. Major Reaction Coordinates Linking Transient Amyloid-β Oligomers to Fibrils Measured at Atomic Level. Biophys. J. 2017, 113 (4), 805– 816, DOI: 10.1016/j.bpj.2017.06.06869https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXht1Cku7vN&md5=52799f167c7d9685fa3e01234858383eMajor reaction coordinates linking transient amyloid-β oligomers to fibrils measured at atomic levelChandra, Bappaditya; Bhowmik, Debanjan; Maity, Barun Kumar; Mote, Kaustubh R.; Dhara, Debabrata; Venkatramani, Ravindra; Maiti, Sudipta; Madhu, Perunthiruthy K.Biophysical Journal (2017), 113 (4), 805-816CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)The structural underpinnings for the higher toxicity of the oligomeric intermediates of amyloidogenic peptides, compared to the mature fibrils, remain unknown at present. The transient nature and heterogeneity of the oligomers make it difficult to follow their structure. Here, using vibrational and solid-state NMR spectroscopy, and mol. dynamics simulations, we show that freely aggregating Aβ40 oligomers in physiol. solns. have an intramol. antiparallel configuration that is distinct from the intermol. parallel β-sheet structure obsd. in mature fibrils. The intramol. hydrogen-bonding network flips nearly 90°, and the two β-strands of each monomeric unit move apart, to give rise to the well-known intermol. in-register parallel β-sheet structure in the mature fibrils. Solid-state NMR distance measurements capture the interstrand sepn. within monomer units during the transition from the oligomer to the fibril form. We further find that the D23-K28 salt-bridge, a major feature of the Aβ40 fibrils and a focal point of mutations linked to early onset Alzheimer's disease, is not detectable in the small oligomers. Mol. dynamics simulations capture the correlation between changes in the D23-K28 distance and the flipping of the monomer secondary structure between antiparallel and parallel β-sheet architectures. Overall, we propose interstrand sepn. and salt-bridge formation as key reaction coordinates describing the structural transition of the small Aβ40 oligomers to fibrils.
- 70Nemergut, M.; Marques, S. M.; Uhrik, L.; Vanova, T.; Nezvedova, M.; Gadara, D. C.; Jha, D.; Tulis, J.; Novakova, V.; Planas-Iglesias, J. Domino-like Effect of C112R Mutation on ApoE4 Aggregation and Its Reduction by Alzheimer’s Disease Drug Candidate. Mol. Neurodegener. 2023, 18 (1), 38, DOI: 10.1186/s13024-023-00620-970https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXht1SitbjL&md5=acdfc26c7af26791accfaec4f3a3c83eDomino-like effect of C112R mutation on ApoE4 aggregation and its reduction by Alzheimer's Disease drug candidateNemergut, Michal; Marques, Sergio M.; Uhrik, Lukas; Vanova, Tereza; Nezvedova, Marketa; Gadara, Darshak Chandulal; Jha, Durga; Tulis, Jan; Novakova, Veronika; Planas-Iglesias, Joan; Kunka, Antonin; Legrand, Anthony; Hribkova, Hana; Pospisilova, Veronika; Sedmik, Jiri; Raska, Jan; Prokop, Zbynek; Damborsky, Jiri; Bohaciakova, Dasa; Spacil, Zdenek; Hernychova, Lenka; Bednar, David; Marek, MartinMolecular Neurodegeneration (2023), 18 (1), 38CODEN: MNOEAZ; ISSN:1750-1326. (BioMed Central Ltd.)Apolipoprotein E (ApoE) ε4 genotype is the most prevalent risk factor for late-onset Alzheimers Disease (AD). Although ApoE4 differs from its non-pathol. ApoE3 isoform only by the C112R mutation, the mol. mechanism of its proteinopathy is unknown. Here, we reveal the mol. mechanism of ApoE4 aggregation using a combination of exptl. and computational techniques, including X-ray crystallog., site-directed mutagenesis, hydrogen-deuterium mass spectrometry (HDX-MS), static light scattering and mol. dynamics simulations. Treatment of ApoE ε3/ε3 and ε4/ε4 cerebral organoids with tramiprosate was used to compare the effect of tramiprosate on ApoE4 aggregation at the cellular level. We found that C112R substitution in ApoE4 induces long-distance (> 15 A) conformational changes leading to the formation of a V-shaped dimeric unit that is geometrically different and more aggregation-prone than the ApoE3 structure. AD drug candidate tramiprosate and its metabolite 3-sulfopropanoic acid induce ApoE3-like conformational behavior in ApoE4 and reduce its aggregation propensity. Anal. of ApoE ε4/ε4 cerebral organoids treated with tramiprosate revealed its effect on cholesteryl esters, the storage products of excess cholesterol. Our results connect the ApoE4 structure with its aggregation propensity, providing a new druggable target for neurodegeneration and ageing.
- 71Walsh, D. M.; Thulin, E.; Minogue, A. M.; Gustavsson, N.; Pang, E.; Teplow, D. B.; Linse, S. A Facile Method for Expression and Purification of the Alzheimer’s Disease-Associated Amyloid Beta-Peptide. FEBS J. 2009, 276 (5), 1266– 1281, DOI: 10.1111/j.1742-4658.2008.06862.x71https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXivFensbo%253D&md5=f644980c816e4b65d8920ed8aee05d0aA facile method for expression and purification of the Alzheimer's disease-associated amyloid β-peptideWalsh, Dominic M.; Thulin, Eva; Minogue, Aedin M.; Gustavsson, Niklas; Pang, Eric; Teplow, David B.; Linse, SaraFEBS Journal (2009), 276 (5), 1266-1281CODEN: FJEOAC; ISSN:1742-464X. (Wiley-Blackwell)The authors report the development of a high-level bacterial expression system for the Alzheimer's disease-assocd. amyloid β-peptide (Aβ), together with a scalable and inexpensive purifn. procedure. Aβ(1-40) and Aβ(1-42) coding sequences together with added ATG codons were cloned directly into a Pet vector to facilitate prodn. of Met-Aβ(1-40) and Met-Aβ(1-42), referred to as Aβ(M1-40) and Aβ(M1-42), resp. The expression sequences were designed using codons preferred by Escherichia coli, and the two peptides were expressed in this host in inclusion bodies. Peptides were purified from inclusion bodies using a combination of anion-exchange chromatog. and centrifugal filtration. The method described requires little specialized equipment and provides a facile and inexpensive procedure for prodn. of large amts. of very pure Aβ peptides. Recombinant peptides generated using this protocol produced amyloid fibrils that were indistinguishable from those formed by chem. synthesized Aβ1-40 and Aβ1-42. Formation of fibrils by all peptides was concn.-dependent, and exhibited kinetics typical of a nucleation-dependent polymn. reaction. Recombinant and synthetic peptides exhibited a similar toxic effect on hippocampal neurons, with acute treatment causing inhibition of MTT redn., and chronic treatment resulting in neuritic degeneration and cell loss.
- 72Thacker, D.; Sanagavarapu, K.; Frohm, B.; Meisl, G.; Knowles, T. P. J.; Linse, S. The Role of Fibril Structure and Surface Hydrophobicity in Secondary Nucleation of Amyloid Fibrils. Proc. Natl. Acad. Sci. U. S. A. 2020, 117 (41), 25272– 25283, DOI: 10.1073/pnas.200295611772https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitVKkt7jO&md5=58081f0fe2ca0ad466ac4491c9b8f627The role of fibril structure and surface hydrophobicity in secondary nucleation of amyloid fibrilsThacker, Dev; Sanagavarapu, Kalyani; Frohm, Birgitta; Meisl, Georg; Knowles, Tuomas P. J.; Linse, SaraProceedings of the National Academy of Sciences of the United States of America (2020), 117 (41), 25272-25283CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Crystals, nanoparticles, and fibrils catalyze the generation of new aggregates on their surface from the same type of monomeric building blocks as the parent assemblies. This secondary nucleation process can be many orders of magnitude faster than primary nucleation. In the case of amyloid fibrils assocd. with Alzheimer's disease, this process leads to the multiplication and propagation of aggregates, whereby short-lived oligomeric intermediates cause neurotoxicity. Understanding the catalytic activity is a fundamental goal in elucidating the mol. mechanisms of Alzheimer's and assocd. diseases. Here the authors explore the role of fibril structure and hydrophobicity by asking whether the V18, A21, V40, and A42 side chains which are exposed on the Aβ42 fibril surface as continuous hydrophobic patches play a role in secondary nucleation. Single, double, and quadruple serine substitutions were made. Kinetic analyses of aggregation data at multiple monomer concns. reveal that all seven mutants retain the dominance of secondary nucleation as the main mechanism of fibril proliferation. This finding highlights the generality of secondary nucleation and its independence of the detailed mol. structure. Cryo-electron micrographs reveal that the V18S substitution causes fibrils to adopt a distinct morphol. with longer twist distance than variants lacking this substitution. Self- and cross-seeding data show that surface catalysis is only efficient between peptides of identical morphol., indicating a templating role of secondary nucleation with structural conversion at the fibril surface. The authors' findings thus provide clear evidence that the propagation of amyloid fibril strains is possible even in systems dominated by secondary nucleation rather than fragmentation.
- 73Yang, H.; Yang, S.; Kong, J.; Dong, A.; Yu, S. Obtaining Information about Protein Secondary Structures in Aqueous Solution Using Fourier Transform IR Spectroscopy. Nat. Protoc. 2015, 10 (3), 382– 396, DOI: 10.1038/nprot.2015.02473https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXit1artbw%253D&md5=690ba9ad6159923b076c27f09822cf4cObtaining information about protein secondary structures in aqueous solution using Fourier transform IR spectroscopyYang, Huayan; Yang, Shouning; Kong, Jilie; Dong, Aichun; Yu, ShaoningNature Protocols (2015), 10 (3), 382-396CODEN: NPARDW; ISSN:1750-2799. (Nature Publishing Group)Fourier transform IR (FTIR) spectroscopy is a nondestructive technique for structural characterization of proteins and polypeptides. The IR spectral data of polymers are usually interpreted in terms of the vibrations of a structural repeat. The repeat units in proteins give rise to nine characteristic IR absorption bands (amides A, B and I-VII). Amide I bands (1,700-1,600 cm-1) are the most prominent and sensitive vibrational bands of the protein backbone, and they relate to protein secondary structural components. In this protocol, we have detailed the principles that underlie the detn. of protein secondary structure by FTIR spectroscopy, as well as the basic steps involved in protein sample prepn., instrument operation, FTIR spectra collection and spectra anal. in order to est. protein secondary-structural components in aq. (both H2O and deuterium oxide (D2O)) soln. using algorithms, such as second-deriv., deconvolution and curve fitting. Small amts. of high-purity (>95%) proteins at high concns. (>3 mg ml-1) are needed in this protocol; typically, the procedure can be completed in 1-2 d.
- 74Hafsa, N. E.; Arndt, D.; Wishart, D. S. CSI 3.0: A Web Server for Identifying Secondary and Super-Secondary Structure in Proteins Using NMR Chemical Shifts. Nucleic Acids Res. 2015, 43 (W1), W370– W377, DOI: 10.1093/nar/gkv494There is no corresponding record for this reference.
- 75Borcherds, W. M.; Daughdrill, G. W. Using NMR Chemical Shifts to Determine Residue-Specific Secondary Structure Populations for Intrinsically Disordered Proteins. Methods Enzymol. 2018, 611, 101– 136, DOI: 10.1016/bs.mie.2018.09.011There is no corresponding record for this reference.
- 76Schumann, F. H.; Riepl, H.; Maurer, T.; Gronwald, W.; Neidig, K.-P.; Kalbitzer, H. R. Combined Chemical Shift Changes and Amino Acid Specific Chemical Shift Mapping of Protein–Protein Interactions. J. Biomol. NMR 2007, 39 (4), 275– 289, DOI: 10.1007/s10858-007-9197-zThere is no corresponding record for this reference.
- 77Heller, G. T.; Aprile, F. A.; Michaels, T. C. T.; Limbocker, R.; Perni, M.; Ruggeri, F. S.; Mannini, B.; Löhr, T.; Bonomi, M.; Camilloni, C. Small-Molecule Sequestration of Amyloid-β as a Drug Discovery Strategy for Alzheimer’s Disease. Sci. Adv. 2020, 6 (45), eabb5924 DOI: 10.1126/sciadv.abb5924There is no corresponding record for this reference.
- 78Habchi, J.; Arosio, P.; Perni, M.; Costa, A. R.; Yagi-Utsumi, M.; Joshi, P.; Chia, S.; Cohen, S. I. A.; Müller, M. B. D.; Linse, S. An Anticancer Drug Suppresses the Primary Nucleation Reaction That Initiates the Production of the Toxic Aβ42 Aggregates Linked with Alzheimer’s Disease. Sci. Adv. 2016, 2 (2), e1501244 DOI: 10.1126/sciadv.150124478https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXlvVWku7g%253D&md5=5cab04625df0052f5d64f57be85007bdAn anticancer drug suppresses the primary nucleation reaction that initiates the production of the toxic Ab42 aggregates linked with Alzheimer's diseaseHabchi, Johnny; Arosio, Paolo; Perni, Michele; Costa, Ana Rita; Yagi-Utsumi, Maho; Joshi, Priyanka; Chia, Sean; Cohen, Samuel I. A.; Muller, Martin B. D.; Linse, Sara; Nollen, Ellen A. A.; Dobson, Christopher M.; Knowles, Tuomas P. J.; Vendruscolo, MicheleScience Advances (2016), 2 (2), e1501244/1-e1501244/14CODEN: SACDAF; ISSN:2375-2548. (American Association for the Advancement of Science)The conversion of the β-amyloid (Aβ) peptide into pathogenic aggregates is linked to the onset and progression of Alzheimer's disease. Although this observation has prompted an extensive search for therapeutic agents to modulate the concn. of Aβ or inhibit its aggregation, all clin. trials with these objectives have so far failed, at least in part because of a lack of understanding of the mol. mechanisms underlying the process of aggregation and its inhibition. To address this problem, we describe a chem. kinetics approach for rational drug discovery, in which the effects of small mols. on the rates of specific microscopic steps in the self-assembly of Aβ42, the most aggregation-prone variant of Aβ, are analyzed quant. By applying this approach, we report that bexarotene, an anticancer drug approved by the U.S. Food and Drug Administration, selectively targets the primary nucleation step in Aβ42 aggregation, delays the formation of toxic species in neuroblastoma cells, and completely suppresses Aβ42 deposition and its consequences in a Caenorhabditis elegans model of Aβ42-mediated toxicity. These results suggest that the prevention of the primary nucleation of Aβ42 by compds. such as bexarotene could potentially reduce the risk of onset of Alzheimer's disease and, more generally, that our strategy provides a general framework for the rational identification of a range of candidate drugs directed against neurodegenerative disorders.
- 79Granata, D.; Baftizadeh, F.; Habchi, J.; Galvagnion, C.; De Simone, A.; Camilloni, C.; Laio, A.; Vendruscolo, M. The Inverted Free Energy Landscape of an Intrinsically Disordered Peptide by Simulations and Experiments. Sci. Rep. 2015, 5 (1), 15449, DOI: 10.1038/srep15449There is no corresponding record for this reference.
- 80Chong, S.-H.; Ham, S. Folding Free Energy Landscape of Ordered and Intrinsically Disordered Proteins. Sci. Rep. 2019, 9 (1), 14927, DOI: 10.1038/s41598-019-50825-680https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3MnptVersw%253D%253D&md5=a20e190c3fa60943db197bf1a798b90aFolding Free Energy Landscape of Ordered and Intrinsically Disordered ProteinsChong Song-Ho; Ham SihyunScientific reports (2019), 9 (1), 14927 ISSN:.Folding funnel is the essential concept of the free energy landscape for ordered proteins. How does this concept apply to intrinsically disordered proteins (IDPs)? Here, we address this fundamental question through the explicit characterization of the free energy landscapes of the representative α-helical (HP-35) and β-sheet (WW domain) proteins and of an IDP (pKID) that folds upon binding to its partner (KIX). We demonstrate that HP-35 and WW domain indeed exhibit the steep folding funnel: the landscape slope for these proteins is ca. -50 kcal/mol, meaning that the free energy decreases by ~5 kcal/mol upon the formation of 10% native contacts. On the other hand, the landscape of pKID is funneled but considerably shallower (slope of -24 kcal/mol), which explains why pKID is disordered in free environments. Upon binding to KIX, the landscape of pKID now becomes significantly steep (slope of -54 kcal/mol), which enables otherwise disordered pKID to fold. We also show that it is the pKID-KIX intermolecular interactions originating from hydrophobic residues that mainly confer the steep folding funnel. The present work not only provides the quantitative characterization of the protein folding free energy landscape, but also establishes the usefulness of the folding funnel concept to IDPs.
- 81Saravanan, K. M.; Zhang, H.; Zhang, H.; Xi, W.; Wei, Y. On the Conformational Dynamics of β-Amyloid Forming Peptides: A Computational Perspective. Front. Bioeng. Biotechnol. 2020, 8, 532, DOI: 10.3389/fbioe.2020.0053281https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jhs1Sntg%253D%253D&md5=356542cee75cb7444d118574fcacd7ffOn the Conformational Dynamics of β-Amyloid Forming Peptides: A Computational PerspectiveSaravanan Konda Mani; Zhang Haiping; Zhang Huiling; Xi Wenhui; Wei YanjieFrontiers in bioengineering and biotechnology (2020), 8 (), 532 ISSN:2296-4185.Understanding the conformational dynamics of proteins and peptides involved in important functions is still a difficult task in computational structural biology. Because such conformational transitions in β-amyloid (Aβ) forming peptides play a crucial role in many neurological disorders, researchers from different scientific fields have been trying to address issues related to the folding of Aβ forming peptides together. Many theoretical models have been proposed in the recent years for studying Aβ peptides using mathematical, physicochemical, and molecular dynamics simulation, and machine learning approaches. In this article, we have comprehensively reviewed the developmental advances in the theoretical models for Aβ peptide folding and interactions, particularly in the context of neurological disorders. Furthermore, we have extensively reviewed the advances in molecular dynamics simulation as a tool used for studying the conversions between polymorphic amyloid forms and applications of using machine learning approaches in predicting Aβ peptides and aggregation-prone regions in proteins. We have also provided details on the theoretical advances in the study of Aβ peptides, which would enhance our understanding of these peptides at the molecular level and eventually lead to the development of targeted therapies for certain acute neurological disorders such as Alzheimer's disease in the future.
- 82Grasso, G.; Danani, A. Molecular Simulations of Amyloid Beta Assemblies. Adv. Phys.: x 2020, 5 (1), 1770627, DOI: 10.1080/23746149.2020.177062782https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXisFanu77F&md5=3f1db67e2d05a8204ce25dd35f6c66ceMolecular simulations of amyloid beta assembliesGrasso, Gianvito; Danani, AndreaAdvances in Physics: X (2020), 5 (1), 1770627CODEN: APXDAR; ISSN:2374-6149. (Taylor & Francis Ltd.)Several neurodegenerative disorders arise from the abnormal protein aggregation in the nervous tissue lead- ing tointracellular inclusions or extracellular aggregates in specific brain areas. In case of Alzheimer Disease, the accumulation of the Amyloid Beta peptide in the brain is proposed to be an early important event in the pathogenesis. Despite significant research efforts in this field, the mol. mechanisms of protein misfolding and aggregation remain somewhat unrevealed.Within this framework, computer simulations represent a power- ful tool able to connect macroscopic exptl. fiend- ings to nanoscale mol. events.However, from the computational point of View, insufficient sampling often limits the ability of computer simulations to fully address this point. One of the main challenges of MD simulations is the ability to sample exptl. relevant millise- cond to second timescales.The present review describes the applications of mol. dynamics techniques to elucidate the conformational states and the aggregation pathway of the Amyloid Beta peptide responsible for AD. Moreover, the computational studies focused on the impact of Amyloid Beta assemblies on cell membranes will be also described. Finally, the interaction mechanisms between promising small mols. and Amyloid Beta assemblies will be discussed within the field of designing new efficient drugs against neurodegenerative disorders.
- 83Haass, C.; Kaether, C.; Thinakaran, G.; Sisodia, S. Trafficking and Proteolytic Processing of APP. Cold Spring Harbor Perspect. Med. 2012, 2 (5), a006270, DOI: 10.1101/cshperspect.a00627083https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXntlenur0%253D&md5=b050f8e78d9221e56ef34a63e8fda596Trafficking and proteolytic processing of APPHaass, Christian; Kaether, Christoph; Thinakaran, Copal; Sisodia, SangramCold Spring Harbor Perspectives in Medicine (2012), 2 (5), a006270/1-a006270/25CODEN: CSHPFV; ISSN:2157-1422. (Cold Spring Harbor Laboratory Press)A review. Accumulations of insol. deposits of amyloid β-peptide are major pathol. hallmarks of Alzheimer disease. Amyloid β-peptide is derived by sequential proteolytic processing from a large type 1 trans-membrane protein, the β-amyloid precursor protein. The proteolytic enzymes involved in its processing are named secretases. β- And γ-secretase liberate by sequential cleavage the neurotoxic amyloid β-peptide, whereas α-secretase prevents its generation by cleaving within the middle of the amyloid domain. In this chapter we describe the cell biol. and biochem. characteristics of the three secretase activities involved in the proteolytic processing of the precursor protein. In addn. we outline how the precursor protein maturates and traffics through the secretory pathway to reach the subcellular locations where the individual secretases are preferentially active. Furthermore, we illuminate how neuronal activity and mutations which cause familial Alzheimer disease affect amyloid β-peptide generation and therefore disease onset and progression.
- 84Zhou, M.; Wen, H.; Lei, H.; Zhang, T. Molecular Dynamics Study of Conformation Transition from Helix to Sheet of Aβ42 Peptide. J. Mol. Graphics Modell. 2021, 109, 108027, DOI: 10.1016/j.jmgm.2021.10802784https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitVamsr%252FJ&md5=2f9eda592333471c3d2f135c4f193c59Molecular dynamics study of conformation transition from helix to sheet of Aβ42 peptideZhou, Min; Wen, Huilin; Lei, Huimin; Zhang, TaoJournal of Molecular Graphics & Modelling (2021), 109 (), 108027CODEN: JMGMFI; ISSN:1093-3263. (Elsevier Ltd.)Aβ42 peptides can form helix and sheet structure under different conditions. The conformational conversion is closely assocd. with Aβ peptides aggregation and their neurotoxicity. But the transition from helix to sheet is not be clearly understood. In this study we performed microsecond timescale MD simulations of Aβ42 peptide to investigate the conformation transition from α-helix to β-sheet. Markov state model (MSM) was built to facilitate identification of crucial intermediate states and possible transition pathway. Based on the anal., we found that the region Y10-A21 in the middle of Aβ42 peptide plays an initial role in this transition. MSM model revealed that the collapse of helical structure in this region might trigger the formation of sheet structure. Moreover, we further simulated the aggregation of Aβ42 peptides with different conformations. We found that the Aβ42 peptides forming sheet structure have higher aggregation potential compared with peptides with helix structure. These results demonstrate that we can prevent the aggregation of Aβ42 peptides by stabilizing the helix structure in the region of Y10-A21. In addn., this study provides new insight into better understanding the conformational transition and aggregation of Aβ42 peptides.
- 85Shuaib, S.; Goyal, B. Scrutiny of the Mechanism of Small Molecule Inhibitor Preventing Conformational Transition of Amyloid-Β42 Monomer: Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2018, 36 (3), 663– 678, DOI: 10.1080/07391102.2017.1291363There is no corresponding record for this reference.
- 86Liu, F.; Ma, Z.; Sang, J.; Lu, F. Edaravone Inhibits the Conformational Transition of Amyloid-Β42: Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2020, 38 (8), 2377– 2388, DOI: 10.1080/07391102.2019.1632225There is no corresponding record for this reference.
- 87Narang, S. S.; Goyal, D.; Goyal, B. Inhibition of Alzheimer’s Amyloid-Β42 Peptide Aggregation by a Bi-Functional Bis-Tryptoline Triazole: Key Insights from Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2020, 38 (6), 1598– 1611, DOI: 10.1080/07391102.2019.1614093There is no corresponding record for this reference.
- 88Cao, Y.; Jiang, X.; Han, W. Self-Assembly Pathways of β-Sheet-Rich Amyloid-β(1–40) Dimers: Markov State Model Analysis on Millisecond Hybrid-Resolution Simulations. J. Chem. Theory Comput. 2017, 13 (11), 5731– 5744, DOI: 10.1021/acs.jctc.7b0080388https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhs1CisLzE&md5=761fcde5aab69118ef6332fda366f37bSelf-Assembly Pathways of β-Sheet-Rich Amyloid-β(1-40) Dimers: Markov State Model Analysis on Millisecond Hybrid-Resolution SimulationsCao, Yang; Jiang, Xuehan; Han, WeiJournal of Chemical Theory and Computation (2017), 13 (11), 5731-5744CODEN: JCTCCE; ISSN:1549-9618. (American Chemical Society)Early oligomerization during amyloid-β (Aβ) aggregation is essential for Aβ neurotoxicity. Understanding how unstructured Aβs assemble into oligomers, esp. those rich in β-sheets, is essential but remains challenging as the assembly process is too transient for exptl. characterization and too slow for mol. dynamics simulations. So far, at. simulations are limited only to studies of either oligomer structures or assembly pathways for short Aβ segments. Here, to overcome the computational challenge, we combined in this study a hybrid-resoln. model and adaptive sampling techniques to perform over 2.7 ms of simulations of formation of full-length Aβ40 dimers that are the earliest toxic oligomeric species. The Markov state model was further employed to characterize the transition pathways and assocd. kinetics. The results showed that for 2 major forms of β-sheet-rich structures reported exptl., the corresponding assembly mechanisms were markedly different. Hairpin-contg. structures were formed by direct binding of sol. Aβ in β-hairpin-like conformations. The formation of parallel, in-register structures resembling fibrils occurred ∼100-fold more slowly and involved a rapid encounter of Aβ in arbitrary conformations followed by a slow structural conversion. The structural conversion proceeded via diverse pathways but always required transient unfolding of encounter complexes. We found that the transition kinetics could be affected differently by intramol./intermol. interactions involving individual residues in a conformation-dependent manner. In particular, the interactions involving Aβ's N-terminal part promoted the assembly into hairpin-contg. structures but delayed the formation of fibril-like structures, thus explaining puzzling observations reported previously regarding the roles of this region in the early assembly process.
- 89Rojas, A. V.; Liwo, A.; Scheraga, H. A. A Study of the α-Helical Intermediate Preceding the Aggregation of the Amino-Terminal Fragment of the β Amyloid Peptide (Aβ1–28). J. Phys. Chem. B 2011, 115 (44), 12978– 12983, DOI: 10.1021/jp205099389https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlaqtrrI&md5=bb4a962dd9c9ef4527c600252e2f3b71A Study of the α-Helical Intermediate Preceding the Aggregation of the Amino-Terminal Fragment of the β Amyloid Peptide (Aβ1-28)Rojas, Ana V.; Liwo, Adam; Scheraga, Harold A.Journal of Physical Chemistry B (2011), 115 (44), 12978-12983CODEN: JPCBFK; ISSN:1520-5207. (American Chemical Society)The β amyloid (Aβ) peptide aggregates to form β-rich structures that are known to trigger Alzheimer's disease. Expts. suggest that an α-helical intermediate precedes the formation of these aggregates. However, a description at the mol. level of the α-to-β transition has not been obtained. Because it has been proposed that the transition might be initiated in the amino-terminal region of Aβ, we studied the aggregation of the 28-residue amino-terminal fragment of Aβ (Aβ1-28) using mol. dynamics (MD) and a coarse-grained force field. Simulations starting from extended and helical conformations showed that oligomerization is initiated by the formation of intermol. β-sheets between the residues in the N-terminal regions. In simulations starting from the α-helical conformation, forcing residues 17-21 to remain in the initial (helical) conformation prevents aggregation but allows for the formation of dimers, indicating that oligomerization, initiated along the nonhelical N-terminal regions, cannot progress without the α-to-β transition propagating along the chains.
- 90Tarasoff-Conway, J. M.; Carare, R. O.; Osorio, R. S.; Glodzik, L.; Butler, T.; Fieremans, E.; Axel, L.; Rusinek, H.; Nicholson, C.; Zlokovic, B. V. Clearance Systems in the Brain-Implications for Alzheimer Disease. Nat. Rev. Neurol. 2015, 11 (8), 457– 470, DOI: 10.1038/nrneurol.2015.11990https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1Wqt73N&md5=52b11eec4c752ab3fdaa14b721478061Clearance systems in the brain-implications for Alzheimer diseaseTarasoff-Conway, Jenna M.; Carare, Roxana O.; Osorio, Ricardo S.; Glodzik, Lidia; Butler, Tracy; Fieremans, Els; Axel, Leon; Rusinek, Henry; Nicholson, Charles; Zlokovic, Berislav V.; Frangione, Blas; Blennow, Kaj; Menard, Joel; Zetterberg, Henrik; Wisniewski, Thomas; de Leon, Mony J.Nature Reviews Neurology (2015), 11 (8), 457-470CODEN: NRNACP; ISSN:1759-4758. (Nature Publishing Group)Accumulation of toxic protein aggregates-amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles is the pathol. hallmark of Alzheimer disease (AD). Aβ accumulation has been hypothesized to result from an imbalance between Aβ prodn. and clearance; indeed, Aβ clearance seems to be impaired in both early and late forms of AD. To develop efficient strategies to slow down or halt AD, it is crit. to understand how Aβ is cleared from the brain. Extracellular Aβ deposits can be removed from the brain by various clearance systems, most importantly, transport across the blood-brain barrier. Findings from the past few years suggest that astroglial-mediated interstitial fluid (ISF) bulk flow, known as the glymphatic system, might contribute to a larger portion of extracellular Aβ (eAβ) clearance than previously thought. The meningeal lymphatic vessels, discovered in 2015, might provide another clearance route. Because these clearance systems act together to drive eAβ from the brain, any alteration to their function could contribute to AD. An understanding of Aβ clearance might provide strategies to reduce excess Aβ deposits and delay, or even prevent, disease onset. In this Review, we describe the clearance systems of the brain as they relate to proteins implicated in AD pathol., with the main focus on Aβ.
- 91Patterson, B. W.; Elbert, D. L.; Mawuenyega, K. G.; Kasten, T.; Ovod, V.; Ma, S.; Xiong, C.; Chott, R.; Yarasheski, K.; Sigurdson, W. Age and Amyloid Effects on Human Central Nervous System Amyloid-Beta Kinetics. Ann. Neurol. 2015, 78 (3), 439– 453, DOI: 10.1002/ana.24454There is no corresponding record for this reference.
- 92Yamazaki, Y.; Zhao, N.; Caulfield, T. R.; Liu, C.-C.; Bu, G. Apolipoprotein E and Alzheimer Disease: Pathobiology and Targeting Strategies. Nat. Rev. Neurol. 2019, 15 (9), 501– 518, DOI: 10.1038/s41582-019-0228-7There is no corresponding record for this reference.
- 93Bye, J. W.; Falconer, R. J. Thermal Stability of Lysozyme as a Function of Ion Concentration: A Reappraisal of the Relationship between the Hofmeister Series and Protein Stability. Protein Sci. 2013, 22 (11), 1563– 1570, DOI: 10.1002/pro.235593https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1eksrfP&md5=bde6c804bbabbe81d8e60cdeaba2e3d4Thermal stability of lysozyme as a function of ion concentration: A reappraisal of the relationship between the Hofmeister series and protein stabilityBye, Jordan W.; Falconer, Robert J.Protein Science (2013), 22 (11), 1563-1570CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Anion and cation effects on the structural stability of lysozyme were investigated using differential scanning calorimetry. At low concns. (<5 mM) anions and cations alter the stability of lysozyme but they do not follow the Hofmeister (or inverse Hofmeister) series. At higher concns. protein stabilization follows the well-established Hofmeister series. Our hypothesis is that there are three mechanisms at work. At low concns. the anions interact with charged side chains where the presence of the ion can alter the structural stability of the protein. At higher concns. the low charge d. anions perchlorate and iodide interact weakly with the protein. Their presence however reduces the Gibbs free energy required to hydrate the core of the protein that is exposed during unfolding therefore destabilizing the structure. At higher concns. the high charge d. anions phosphate and sulfate compete for water with the protein as it unfolds increasing the Gibbs free energy required to hydrate the newly exposed core of the protein therefore stabilizing the structure.
- 94Martens, Y. A.; Zhao, N.; Liu, C.-C.; Kanekiyo, T.; Yang, A. J.; Goate, A. M.; Holtzman, D. M.; Bu, G. ApoE Cascade Hypothesis in the Pathogenesis of Alzheimer’s Disease and Related Dementias. Neuron 2022, 110 (8), 1304– 1317, DOI: 10.1016/j.neuron.2022.03.004There is no corresponding record for this reference.
- 95Chai, A. B.; Lam, H. H. J.; Kockx, M.; Gelissen, I. C. Apolipoprotein E Isoform-Dependent Effects on the Processing of Alzheimer’s Amyloid-β. Biochim. Biophys. Acta, Mol. Cell Biol. Lipids 2021, 1866 (9), 158980, DOI: 10.1016/j.bbalip.2021.158980There is no corresponding record for this reference.
- 96Tijms, B. M.; Vromen, E. M.; Mjaavatten, O.; Holstege, H.; Reus, L. M.; van der Lee, S.; Wesenhagen, K. E. J.; Lorenzini, L.; Vermunt, L.; Venkatraghavan, V. Cerebrospinal Fluid Proteomics in Patients with Alzheimer’s Disease Reveals Five Molecular Subtypes with Distinct Genetic Risk Profiles. Nat. Aging 2024, 4 (1), 33– 47, DOI: 10.1038/s43587-023-00550-7There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/jacsau.4c00182.
Detailed materials and methods, complementary to the concise descriptions in this main text, Supplementary discussions (Notes 1–10), Supplementary figures: structure of Aβ42 (Figure S1), comparison of the Amber ff14SB and CHARMM36m force fields (Figures S2 and S3), comparison of different adaptive sampling protocols (Figures S4 and S5), temporal alignment and concatenation of the adaptive sampling and classical MDs (Figures S6–S9), conventional Markov state model analysis (Figures S10–S12), variational Markov state analysis using VAMPnets (Figures S13–S18), comparative Markov state model analysis (Figure S19), time-based evolution of the states (Figure S20), radius of gyration by state (Figure S21), characterization of learned conformational states via network gradients (Figures S22 and S23), interactions of Aβ42 with the small molecules (Figures S24–S26), experimental validation (Figures S27–S31), Supplementary tables: comparison of computational protocols for the simulation of Aβ42 (Table S1), analysis of classical MDs (Table S2), summary of the effects of small molecules (Table S3), and intramolecular interactions of Aβ42 (Table S4) (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.