1
|
Cao S, Nüske F, Liu B, Soley MB, Huang X. AMUSET-TICA: A Tensor-Based Approach for Identifying Slow Collective Variables in Biomolecular Dynamics. J Chem Theory Comput 2025; 21:4855-4866. [PMID: 40254940 DOI: 10.1021/acs.jctc.5c00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Elucidating collective variables (CVs) for biomolecular dynamics is crucial for understanding numerous biological processes. By leveraging the tensor-train data structure, a multilinear version of the AMUSE (Algorithm for Multiple Unknown Signals) algorithm for Koopman approximation (AMUSEt) was recently developed to identify CVs for biomolecular dynamics. To find slow CVs, AMUSEt transforms input features (e.g., pairwise atomic distances) into nonlinear basis functions (e.g., Gaussian functions) and encodes these nonlinear basis functions within a tensor-train structure via time-lagged correlation functions. Due to the need to fit these tensor-train data structures into computer memory, AMUSEt can handle only a limited number of input features. Consequently, AMUSEt relies on manually selecting and ranking features based on physical intuition to fully capture the slow dynamics. However, when applied to complex biological systems with numerous features, this selection and ranking process becomes increasingly challenging. To address this challenge, here we present AMUSET-TICA (AMUSEt-based Time-lagged Independent Component Analysis), a CV-identification method using time-structure-independent components (tICs) as the input features for AMUSEt. The key insight of AMUSET-TICA lies in its highly effective embedding of high-dimensional atomistic protein conformations, achieved by expanding orthogonal tICs into overlapping Gaussian basis functions through a tensor-product data structure. This eliminates the need for manually selecting and ranking input features for a wide range of biomolecular systems. We demonstrate that AMUSET-TICA consistently and significantly outperforms AMUSEt and tICA in identifying slow CVs for three different biomolecular systems: alanine dipeptide, the N-terminal domain of L9 (NTL9), and the FIP35 WW domain. For all these systems, the CVs generated by AMUSET-TICA accurately describe the slowest dynamical modes underlying these biological conformational changes. Furthermore, we show that AMUSET-TICA achieves performance comparable to deep-learning approaches like VAMPnets in identifying the slowest dynamical modes, while being significantly more computationally efficient in terms of CPU time. In addition, the CVs yielded by AMUSET-TICA provide insights into the folding mechanisms of NTL9 and the FIP35 WW domain, including CV3 and CV4 of the WW domain, which capture its two parallel folding pathways. We expect AMUSET-TICA can be widely applied to facilitate the investigation of biomolecular dynamics.
Collapse
Affiliation(s)
- Siqin Cao
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Feliks Nüske
- Max-Planck-Institute for Dynamics of Complex Technical Systems, Magdeburg 39106, Germany
| | - Bojun Liu
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Micheline B Soley
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Xuhui Huang
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
2
|
Orr AA, Uwakweh AO, Li X, Karanji AK, Hoag SW, Deredge DJ, MacKerell AD. Mapping the distribution and affinities of ligand interaction sites on human serum albumin. Biophys J 2025:S0006-3495(25)00170-5. [PMID: 40134214 DOI: 10.1016/j.bpj.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
Ligands in many instances interact with a protein at multiple sites with a range of affinities. In this study, ligand-protein interaction sites on human serum albumin (HSA) are mapped using the site-identification by ligand competitive saturation (SILCS)-Biologics approach in conjunction with hydrogen-deuterium exchange (HDX)-mass spectrometry (MS) experiments. Ligands studied include known HSA binders, ibuprofen and ketoprofen, and compounds arginine, alanine, sucrose, and trehalose, excipients used in therapeutic formulations of protein-based drugs. In addition, the impact of excipient binding to HSA on its stability is investigated through temperature-ramp stability studies monitoring solution viscosity. For the studied ligands, interactions that correspond to known drug-binding sites (DSs) are identified. These include previously identified ibuprofen and ketoprofen interaction sites as well as additional sites and, in the case of the excipients, the ligands are shown to also bind at previously unidentified interaction sites, termed excipient sites (ESs) with 20 or more sites identified for the studied compounds. HDX-MS titrations were used to determine dissociation constants for a subset of the interaction sites for ibuprofen, ketoprofen, arginine, and sucrose, which exhibited Kd values in the low micromolar to millimolar range in satisfactory agreement with SILCS-Biologics predicted affinities, validating the computational approach to identify both high- and low-affinity interaction sites. The stability studies indicate the excipients offer protection at low excipient/protein ratios up to 66 with destabilization occurring at ratios above 132 with the exception of sucrose at the t0 time point, indicating that the more favorable affinities of sucrose seen in the SILCS-Biologics and HDX-MS analyses contribute to protein stabilization. These results indicate that ligands can bind to large numbers of interaction sites on proteins, with those interactions having implications for the development of formulations for therapeutic proteins.
Collapse
Affiliation(s)
- Asuka A Orr
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland; SilcsBio LLC, Baltimore, Maryland
| | - Agbo-Oma Uwakweh
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Xun Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Ahmad Kiani Karanji
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Stephen W Hoag
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland
| | - Daniel J Deredge
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland.
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, Maryland.
| |
Collapse
|
3
|
Nussinov R, Yavuz BR, Jang H. Allostery in Disease: Anticancer Drugs, Pockets, and the Tumor Heterogeneity Challenge. J Mol Biol 2025:169050. [PMID: 40021049 DOI: 10.1016/j.jmb.2025.169050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Charting future innovations is challenging. Yet, allosteric and orthosteric anticancer drugs are undergoing a revolution and taxing unresolved dilemmas await. Among the imaginative innovations, here we discuss cereblon and thalidomide derivatives as a means of recruiting neosubstrates and their degradation, allosteric heterogeneous bifunctional drugs like PROTACs, drugging phosphatases, inducers of targeted posttranslational protein modifications, antibody-drug conjugates, exploiting membrane interactions to increase local concentration, stabilizing the folded state, and more. These couple with harnessing allosteric cryptic pockets whose discovery offers more options to modulate the affinity of orthosteric, active site inhibitors. Added to these are strategies to counter drug resistance through drug combinations co-targeting pathways to bypass signaling blockades. Here, we discuss on the molecular and cellular levels, such inspiring advances, provide examples of their applications, their mechanisms and rational. We start with an overview on difficult to target proteins and their properties-rarely, if ever-conceptualized before, discuss emerging innovative drugs, and proceed to the increasingly popular allosteric cryptic pockets-their advantages-and critically, issues to be aware of. We follow with drug resistance and in-depth discussion of tumor heterogeneity. Heterogeneity is a hallmark of highly aggressive cancers, the core of drug resistance unresolved challenge. We discuss potential ways to target heterogeneity by predicting it. The increase in experimental and clinical data, computed (cell-type specific) interactomes, capturing transient cryptic pockets, learned drug resistance, workings of regulatory mechanisms, heterogeneity, and resistance-based cell signaling drug combinations, assisted by AI-driven reasoning and recognition, couple with creative allosteric drug discovery, charting future innovations.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, the United States of America; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, the United States of America; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America
| |
Collapse
|
4
|
Qiao X, Li X, Zhang M, Liu N, Wu Y, Lu S, Chen T. Targeting cryptic allosteric sites of G protein-coupled receptors as a novel strategy for biased drug discovery. Pharmacol Res 2025; 212:107574. [PMID: 39755133 DOI: 10.1016/j.phrs.2024.107574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/06/2025]
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of membrane receptors and are highly effective targets for therapeutic drugs. GPCRs couple different downstream effectors, including G proteins (such as Gi/o, Gs, G12, and Gq) and β-arrestins (such as β-arrestin 1 and β-arrestin 2) to mediate diverse cellular and physiological responses. Biased signaling allows for the specific activation of certain pathways from the full range of receptors' signaling capabilities. Targeting more variable allosteric sites, which are spatially different from the highly conserved orthosteric sites, represents a novel approach in biased GPCR drug discovery, leading to innovative strategies for targeting GPCRs. Notably, the emergence of cryptic allosteric sites on GPCRs has expanded the repertoire of available drug targets and improved receptor subtype selectivity. Here, we conduct a summary of recent progress in the structural determination of cryptic allosteric sites on GPCRs and elucidate the biased signaling mechanisms induced by allosteric modulators. Additionally, we discuss means to identify cryptic allosteric sites and design biased allosteric modulators based on cryptic allosteric sites through structure-based drug design, which is an advanced pharmacotherapeutic approach for treating GPCR-associated diseases.
Collapse
Affiliation(s)
- Xin Qiao
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaolong Li
- Department of Orthopedics, Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Mingyang Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ning Liu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Yanmei Wu
- Department of General Surgery, Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| | - Shaoyong Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China.
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
5
|
Xiao S, Alshahrani M, Hu G, Tao P, Verkhivker G. Accurate Characterization of the Allosteric Energy Landscapes, Binding Hotspots and Long-Range Communications for KRAS Complexes with Effector Proteins : Integrative Approach Using Microsecond Molecular Dynamics, Deep Mutational Scanning of Binding Energetics and Allosteric Network Modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635141. [PMID: 39975035 PMCID: PMC11838311 DOI: 10.1101/2025.01.27.635141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
KRAS is a pivotal oncoprotein that regulates cell proliferation and survival through interactions with downstream effectors such as RAF1. Oncogenic mutations in KRAS, including G12V, G13D, and Q61R, drive constitutive activation and hyperactivation of signaling pathways, contributing to cancer progression. Despite significant advances in understanding KRAS biology, the structural and dynamic mechanisms of KRAS binding and allostery by which oncogenic mutations enhance KRAS-RAF1 binding and signaling remain incompletely understood. In this study, we employ microsecond molecular dynamics simulations, Markov State Modeling, mutational scanning and binding free energy calculations together with dynamic network modeling to elucidate the effect of KRAS mutations and characterize the thermodynamic and allosteric drivers and hotspots of KRAS binding and oncogenic activation. Our simulations revealed that oncogenic mutations stabilize the open active conformation of KRAS by differentially modulating the flexibility of the switch I and switch II regions, thereby enhancing RAF1 binding affinity. The G12V mutation rigidifies both switch I and switch II, locking KRAS in a stable, active state. In contrast, the G13D mutation moderately reduces switch I flexibility while increasing switch II dynamics, restoring a balance between stability and flexibility. The Q61R mutation induces a more complex conformational landscape, characterized by the increased switch II flexibility and expansion of functional macrostates, which promotes prolonged RAF1 binding and signaling. Mutational scanning of KRAS-RAF1 complexes identified key binding affinity hotspots, including Y40, E37, D38, and D33, and together with the MM-GBSA analysis revealed the hotspots leverage synergistic electrostatic and hydrophobic binding interactions in stabilizing the KRAS-RAF1 complexes. Network-based analysis of allosteric communication identifies critical KRAS residues (e.g., L6, E37, D57, R97) that mediate long-range interactions between the KRAS core and the RAF1 binding interface. The central β-sheet of KRAS emerges as a hub for transmitting conformational changes, linking distant functional sites and facilitating allosteric regulation. Strikingly, the predicted allosteric hotspots align with experimentally identified allosteric binding hotspots that define the energy landscape of KRAS allostery. This study highlights the power of integrating computational modeling with experimental data to unravel the complex dynamics of KRAS and its mutants. The identification of binding hotspots and allosteric communication routes offers new opportunities for developing targeted therapies to disrupt KRAS-RAF1 interactions and inhibit oncogenic signaling. Our results underscore the potential of computational approaches to guide the design of allosteric inhibitors and mutant-specific therapies for KRAS-driven cancers.
Collapse
|
6
|
Ricci CG, Philpott JM, Torgrimson MR, Freeberg AM, Narasimamurthy R, de Barros EP, Amaro R, Virshup DM, McCammon JA, Partch CL. Markovian State Models uncover Casein Kinase 1 dynamics that govern circadian period. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633651. [PMID: 39896482 PMCID: PMC11785140 DOI: 10.1101/2025.01.17.633651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Circadian rhythms in mammals are tightly regulated through phosphorylation of Period (PER) proteins by Casein Kinase 1 (CK1, subtypes δ and ε). CK1 acts on at least two different regions of PER with opposing effects: phosphorylation of phosphodegron (pD) regions leads to PER degradation, while phosphorylation of the Familial Advanced Sleep Phase (FASP) region leads to PER stabilization. To investigate how substrate selectivity is encoded by the conformational dynamics of CK1, we performed a large set of independent molecular dynamics (MD) simulations of wildtype CK1 and the tau mutant (R178C) that biases kinase activity toward a pD. We used Markovian State Models (MSMs) to integrate the simulations into a single model of the conformational landscape of CK1 and used Gaussian accelerated molecular dynamics (GaMD) to build the first molecular model of CK1 and the unphosphorylated FASP motif. Together, these findings provide a mechanistic view of CK1, establishing how the activation loop acts as a key molecular switch to control substrate selectivity. We show that the tau mutant favors an alternative conformation of the activation loop and significantly accelerates the dynamics of CK1. This reshapes the binding cleft in a way that impairs FASP binding and would ultimately lead to PER destabilization and shorter circadian periods. Finally, we identified an allosteric pocket that could be targeted to bias this molecular switch. Our integrated approach offers a detailed model of CK1's conformational landscape and its relevance to normal, mutant, and druggable circadian timekeeping.
Collapse
Affiliation(s)
- Clarisse Gravina Ricci
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California, United States
- Current address: D.E. Shaw Research, New York, New York, United States
| | - Jonathan M. Philpott
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States
| | - Megan R. Torgrimson
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States
| | - Alfred M. Freeberg
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States
| | - Rajesh Narasimamurthy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Emilia Pécora de Barros
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California, United States
| | - Rommie Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California, United States
| | - David M. Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, United States
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, California, United States
| | - Carrie L. Partch
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California, United States
- Center for Circadian Biology, University of California San Diego, San Diego, California, United States
- Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, California, United States
| |
Collapse
|
7
|
Vögele M, Thomson NJ, Truong ST, McAvity J, Zachariae U, Dror RO. Systematic analysis of biomolecular conformational ensembles with PENSA. J Chem Phys 2025; 162:014101. [PMID: 39745157 PMCID: PMC11698571 DOI: 10.1063/5.0235544] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025] Open
Abstract
Atomic-level simulations are widely used to study biomolecules and their dynamics. A common goal in such studies is to compare simulations of a molecular system under several conditions-for example, with various mutations or bound ligands-in order to identify differences between the molecular conformations adopted under these conditions. However, the large amount of data produced by simulations of ever larger and more complex systems often renders it difficult to identify the structural features that are relevant to a particular biochemical phenomenon. We present a flexible software package named Python ENSemble Analysis (PENSA) that enables a comprehensive and thorough investigation into biomolecular conformational ensembles. It provides featurization and feature transformations that allow for a complete representation of biomolecules such as proteins and nucleic acids, including water and ion binding sites, thus avoiding the bias that would come with manual feature selection. PENSA implements methods to systematically compare the distributions of molecular features across ensembles to find the significant differences between them and identify regions of interest. It also includes a novel approach to quantify the state-specific information between two regions of a biomolecule, which allows, for example, tracing information flow to identify allosteric pathways. PENSA also comes with convenient tools for loading data and visualizing results, making them quick to process and easy to interpret. PENSA is an open-source Python library maintained at https://github.com/drorlab/pensa along with an example workflow and a tutorial. We demonstrate its usefulness in real-world examples by showing how it helps us determine molecular mechanisms efficiently.
Collapse
Affiliation(s)
- Martin Vögele
- Authors to whom correspondence should be addressed: and
| | - Neil J. Thomson
- Department of Computational Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Sang T. Truong
- Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | - Jasper McAvity
- Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | | | - Ron O. Dror
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
8
|
Gao Y, Chen H, Yang W, Wang S, Gong D, Zhang X, Huang Y, Kumar V, Huang Q, Kandegama WMWW, Hao G. New avenues of combating antibiotic resistance by targeting cryptic pockets. Pharmacol Res 2024; 210:107495. [PMID: 39491636 DOI: 10.1016/j.phrs.2024.107495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/02/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Antibiotic resistance is a global health concern that is rapidly spreading among human and animal pathogens. Developing novel antibiotics is one of the most significant approaches to surmount antibiotic resistance. Given the difficult in identifying novel targets, cryptic binding sites provide new pockets for compounds design to combat antibiotic resistance. However, there exists a lack of comprehensive analysis and discussion on the successful utilization of cryptic pockets in overcoming antibiotic resistance. Here, we systematically analyze the crucial role of cryptic pockets in neutralizing antibiotic resistance. First, antibiotic resistance development and associated resistance mechanisms are summarized. Then, the advantages and mechanisms of cryptic pockets for overcoming antibiotic resistance were discussed. Specific cryptic pockets in resistant proteins and successful case studies of designed inhibitors are exemplified. This review provides insight into the discovery of cryptic pockets for drug design as an approach to overcome antibiotic resistance.
Collapse
Affiliation(s)
- Yangyang Gao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Huimin Chen
- State Key Laboratory of Green Pesticide, Central China Normal University, Wuhan 430079, PR China
| | - Weicheng Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Shuang Wang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Daohong Gong
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Xiao Zhang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Yuanqin Huang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Vinit Kumar
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - Qiuqian Huang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China
| | - W M W W Kandegama
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; Department of Horticulture and Landscape Gardening, Faculty of Agriculture and Plantation Management, Wayamba University of Sri Lanka, Makandura, Gonawila, 60170 Sri Lanka
| | - Gefei Hao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang 550025, PR China; State Key Laboratory of Green Pesticide, Central China Normal University, Wuhan 430079, PR China.
| |
Collapse
|
9
|
Lazou M, Khan O, Nguyen T, Padhorny D, Kozakov D, Joseph-McCarthy D, Vajda S. Predicting multiple conformations of ligand binding sites in proteins suggests that AlphaFold2 may remember too much. Proc Natl Acad Sci U S A 2024; 121:e2412719121. [PMID: 39565312 PMCID: PMC11621821 DOI: 10.1073/pnas.2412719121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
The goal of this paper is predicting the conformational distributions of ligand binding sites using the AlphaFold2 (AF2) protein structure prediction program with stochastic subsampling of the multiple sequence alignment (MSA). We explored the opening of cryptic ligand binding sites in 16 proteins, where the closed and open conformations define the expected extreme points of the conformational variation. Due to the many structures of these proteins in the Protein Data Bank (PDB), we were able to study whether the distribution of X-ray structures affects the distribution of AF2 models. We have found that AF2 generates both a cluster of open and a cluster of closed models for proteins that have comparable numbers of open and closed structures in the PDB and not too many other conformations. This was observed even with default MSA parameters, thus without further subsampling. In contrast, with the exception of a single protein, AF2 did not yield multiple clusters of conformations for proteins that had imbalanced numbers of open and closed structures in the PDB, or had substantial numbers of other structures. Subsampling improved the results only for a single protein, but very shallow MSA led to incorrect structures. The ability of generating both open and closed conformations for six out of the 16 proteins agrees with the success rates of similar studies reported in the literature. However, we showed that this partial success is due to AF2 "remembering" the conformational distributions in the PDB and that the approach fails to predict rarely seen conformations.
Collapse
Affiliation(s)
- Maria Lazou
- Department of Biomedical Engineering, Boston University, Boston, MA02215
| | - Omeir Khan
- Department of Chemistry, Boston University, Boston, MA02215
| | - Thu Nguyen
- Department of Computer Science, Stony Brook University, Stony Brook, NY11794
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY11794
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY11794
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
| | - Diane Joseph-McCarthy
- Department of Biomedical Engineering, Boston University, Boston, MA02215
- Department of Chemistry, Boston University, Boston, MA02215
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA02215
- Department of Chemistry, Boston University, Boston, MA02215
| |
Collapse
|
10
|
Lazou M, Kozakov D, Joseph-McCarthy D, Vajda S. Which cryptic sites are feasible drug targets? Drug Discov Today 2024; 29:104197. [PMID: 39368697 PMCID: PMC11568903 DOI: 10.1016/j.drudis.2024.104197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Cryptic sites can expand the space of druggable proteins, but the potential usefulness of such sites needs to be investigated before any major effort. Given that the binding pockets are not formed, the druggability of such sites is not well understood. The analysis of proteins and their ligands shows that cryptic sites that are formed primarily by the motion of side chains moving out of the pocket to enable ligand binding generally do not bind drug-sized molecules with sufficient potency. By contrast, sites that are formed by loop or hinge motion are potentially valuable drug targets. Arguments are provided to explain the underlying causes in terms of classical enzyme inhibition theory and the kinetics of side chain motion and ligand binding.
Collapse
Affiliation(s)
- Maria Lazou
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, USA; Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Diane Joseph-McCarthy
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston, MA 02215, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Ugurlu SY, McDonald D, He S. MEF-AlloSite: an accurate and robust Multimodel Ensemble Feature selection for the Allosteric Site identification model. J Cheminform 2024; 16:116. [PMID: 39444016 PMCID: PMC11515501 DOI: 10.1186/s13321-024-00882-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/09/2024] [Indexed: 10/25/2024] Open
Abstract
A crucial mechanism for controlling the actions of proteins is allostery. Allosteric modulators have the potential to provide many benefits compared to orthosteric ligands, such as increased selectivity and saturability of their effect. The identification of new allosteric sites presents prospects for the creation of innovative medications and enhances our comprehension of fundamental biological mechanisms. Allosteric sites are increasingly found in different protein families through various techniques, such as machine learning applications, which opens up possibilities for creating completely novel medications with a diverse variety of chemical structures. Machine learning methods, such as PASSer, exhibit limited efficacy in accurately finding allosteric binding sites when relying solely on 3D structural information.Scientific ContributionPrior to conducting feature selection for allosteric binding site identification, integration of supporting amino-acid-based information to 3D structural knowledge is advantageous. This approach can enhance performance by ensuring accuracy and robustness. Therefore, we have developed an accurate and robust model called Multimodel Ensemble Feature Selection for Allosteric Site Identification (MEF-AlloSite) after collecting 9460 relevant and diverse features from the literature to characterise pockets. The model employs an accurate and robust multimodal feature selection technique for the small training set size of only 90 proteins to improve predictive performance. This state-of-the-art technique increased the performance in allosteric binding site identification by selecting promising features from 9460 features. Also, the relationship between selected features and allosteric binding sites enlightened the understanding of complex allostery for proteins by analysing selected features. MEF-AlloSite and state-of-the-art allosteric site identification methods such as PASSer2.0 and PASSerRank have been tested on three test cases 51 times with a different split of the training set. The Student's t test and Cohen's D value have been used to evaluate the average precision and ROC AUC score distribution. On three test cases, most of the p-values ( < 0.05 ) and the majority of Cohen's D values ( > 0.5 ) showed that MEF-AlloSite's 1-6% higher mean of average precision and ROC AUC than state-of-the-art allosteric site identification methods are statistically significant.
Collapse
Affiliation(s)
- Sadettin Y Ugurlu
- School of Computer Science, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | | - Shan He
- School of Computer Science, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
- AIA Insights Ltd, Birmingham, UK.
| |
Collapse
|
12
|
Mallimadugula UL, Cruz MA, Vithani N, Zimmerman MI, Bowman GR. Opening and closing of a cryptic pocket in VP35 toggles it between two different RNA-binding modes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609218. [PMID: 39229186 PMCID: PMC11370563 DOI: 10.1101/2024.08.22.609218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Cryptic pockets are of growing interest as potential drug targets, particularly to control protein-nucleic acid interactions that often occur via flat surfaces. However, it remains unclear whether cryptic pockets contribute to protein function or if they are merely happenstantial features that can easily be evolved away to achieve drug resistance. Here, we explore whether a cryptic pocket in the Interferon Inhibitory Domain (IID) of viral protein 35 (VP35) of Zaire ebolavirus aids its ability to bind double-stranded RNA (dsRNA). We use simulations and experiments to study the relationship between cryptic pocket opening and dsRNA binding of the IIDs of two other filoviruses, Reston and Marburg. These homologs have nearly identical structures but block different interferon pathways due to different affinities for blunt ends and backbone of the dsRNA. Simulations and thiol-labeling experiments demonstrate that the homologs have varying probabilities of pocket opening. Subsequent dsRNA-binding assays suggest that closed conformations preferentially bind dsRNA blunt ends while open conformations prefer binding the backbone. Point mutations that modulate pocket opening proteins further confirm this preference. These results demonstrate the open cryptic pocket has a function, suggesting cryptic pockets are under selective pressure and may be difficult to evolve away to achieve drug resistance.
Collapse
|
13
|
Nordquist EB, Zhao M, Kumar A, MacKerell AD. Combined Physics- and Machine-Learning-Based Method to Identify Druggable Binding Sites Using SILCS-Hotspots. J Chem Inf Model 2024; 64:7743-7757. [PMID: 39283165 PMCID: PMC11473228 DOI: 10.1021/acs.jcim.4c01189] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Identifying druggable binding sites on proteins is an important and challenging problem, particularly for cryptic, allosteric binding sites that may not be obvious from X-ray, cryo-EM, or predicted structures. The Site-Identification by Ligand Competitive Saturation (SILCS) method accounts for the flexibility of the target protein using all-atom molecular simulations that include various small molecule solutes in aqueous solution. During the simulations, the combination of protein flexibility and comprehensive sampling of the water and solute spatial distributions can identify buried binding pockets absent in experimentally determined structures. Previously, we reported a method for leveraging the information in the SILCS sampling to identify binding sites (termed Hotspots) of small mono- or bicyclic compounds, a subset of which coincide with known binding sites of drug-like molecules. Here, we build on that physics-based approach and present a ML model for ranking the Hotspots according to the likelihood they can accommodate drug-like molecules (e.g., molecular weight >200 Da). In the independent validation set, which includes various enzymes and receptors, our model recalls 67% and 89% of experimentally validated ligand binding sites in the top 10 and 20 ranked Hotspots, respectively. Furthermore, we show that the model's output Decision Function is a useful metric to predict binding sites and their potential druggability in new targets. Given the utility the SILCS method for ligand discovery and optimization, the tools presented represent an important advancement in the identification of orthosteric and allosteric binding sites and the discovery of drug-like molecules targeting those sites.
Collapse
Affiliation(s)
- Erik B. Nordquist
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland 21201, United States
| | - Mingtian Zhao
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland 21201, United States
| | - Anmol Kumar
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland 21201, United States
| | - Alexander D. MacKerell
- Computer Aided Drug Design Center, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland 21201, United States
| |
Collapse
|
14
|
Yehorova D, Crean RM, Kasson PM, Kamerlin SCL. Friends and relatives: insight into conformational regulation from orthologues and evolutionary lineages using KIF and KIN. Faraday Discuss 2024; 252:341-353. [PMID: 38842247 PMCID: PMC11389856 DOI: 10.1039/d4fd00018h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Noncovalent interaction networks provide a powerful means to represent and analyze protein structure. Such networks can represent both static structures and dynamic conformational ensembles. We have recently developed two tools for analyzing such interaction networks and generating hypotheses for protein engineering. Here, we apply these tools to the conformational regulation of substrate specificity in class A β-lactamases, particularly the evolutionary development from generalist to specialist catalytic function and how that can be recapitulated or reversed by protein engineering. These tools, KIF and KIN, generate a set of prioritized residues and interactions as targets for experimental protein engineering.
Collapse
Affiliation(s)
- Dariia Yehorova
- School of Chemistry and Biochemistry, Georgia Institute of Technology, USA.
| | - Rory M Crean
- Department of Chemistry-BMC, Uppsala University, Sweden
| | - Peter M Kasson
- Department of Biomedical Engineering, University of Virginia, USA
- Department of Cell and Molecular Biology, Uppsala University, Sweden
- Departments of Chemistry & Biochemistry and Biomedical Engineering, Georgia Institute of Technology, USA.
| | | |
Collapse
|
15
|
Martino SA, Morado J, Li C, Lu Z, Rosta E. Kemeny Constant-Based Optimization of Network Clustering Using Graph Neural Networks. J Phys Chem B 2024; 128:8103-8115. [PMID: 39145603 PMCID: PMC11367579 DOI: 10.1021/acs.jpcb.3c08213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
The recent trend in using network and graph structures to represent a variety of different data types has renewed interest in the graph partitioning (GP) problem. This interest stems from the need for general methods that can both efficiently identify network communities and reduce the dimensionality of large graphs while satisfying various application-specific criteria. Traditional clustering algorithms often struggle to capture the complex relationships within graphs and generalize to arbitrary clustering criteria. The emergence of graph neural networks (GNNs) as a powerful framework for learning representations of graph data provides new approaches to solving the problem. Previous work has shown GNNs to be capable of proposing partitionings using a variety of criteria. However, these approaches have not yet been extended to Markov chains or kinetic networks. These arise frequently in the study of molecular systems and are of particular interest to the biomolecular modeling community. In this work, we propose several GNN-based architectures to tackle the GP problem for Markov Chains described as kinetic networks. This approach aims to maximize the Kemeny constant, which is a variational quantity and it represents the sum of time scales of the system. We propose using an encoder-decoder architecture and show how simple GraphSAGE-based GNNs with linear layers can outperform much larger and more expressive attention-based models in this context. As a proof of concept, we first demonstrate the method's ability to cluster randomly connected graphs. We also use a linear chain architecture corresponding to a 1D free energy profile as our kinetic network. Subsequently, we demonstrate the effectiveness of our method through experiments on a data set derived from molecular dynamics. We compare the performance of our method to other partitioning techniques, such as PCCA+. We explore the importance of feature and hyperparameter selection and propose a general strategy for large-scale parallel training of GNNs for discovering optimal graph partitionings.
Collapse
Affiliation(s)
- Sam Alexander Martino
- Department of Physics and
Astronomy, University College London, London WC1E 6BT, U.K.
| | - João Morado
- Department of Physics and
Astronomy, University College London, London WC1E 6BT, U.K.
| | - Chenghao Li
- Department of Physics and
Astronomy, University College London, London WC1E 6BT, U.K.
| | - Zhenghao Lu
- Department of Physics and
Astronomy, University College London, London WC1E 6BT, U.K.
| | - Edina Rosta
- Department of Physics and
Astronomy, University College London, London WC1E 6BT, U.K.
| |
Collapse
|
16
|
Pillai A, Idris A, Philomin A, Weidle C, Skotheim R, Leung PJY, Broerman A, Demakis C, Borst AJ, Praetorius F, Baker D. De novo design of allosterically switchable protein assemblies. Nature 2024; 632:911-920. [PMID: 39143214 PMCID: PMC11338832 DOI: 10.1038/s41586-024-07813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Allosteric modulation of protein function, wherein the binding of an effector to a protein triggers conformational changes at distant functional sites, plays a central part in the control of metabolism and cell signalling1-3. There has been considerable interest in designing allosteric systems, both to gain insight into the mechanisms underlying such 'action at a distance' modulation and to create synthetic proteins whose functions can be regulated by effectors4-7. However, emulating the subtle conformational changes distributed across many residues, characteristic of natural allosteric proteins, is a significant challenge8,9. Here, inspired by the classic Monod-Wyman-Changeux model of cooperativity10, we investigate the de novo design of allostery through rigid-body coupling of peptide-switchable hinge modules11 to protein interfaces12 that direct the formation of alternative oligomeric states. We find that this approach can be used to generate a wide variety of allosterically switchable systems, including cyclic rings that incorporate or eject subunits in response to peptide binding and dihedral cages that undergo effector-induced disassembly. Size-exclusion chromatography, mass photometry13 and electron microscopy reveal that these designed allosteric protein assemblies closely resemble the design models in both the presence and absence of peptide effectors and can have ligand-binding cooperativity comparable to classic natural systems such as haemoglobin14. Our results indicate that allostery can arise from global coupling of the energetics of protein substructures without optimized side-chain-side-chain allosteric communication pathways and provide a roadmap for generating allosterically triggerable delivery systems, protein nanomachines and cellular feedback control circuitry.
Collapse
Affiliation(s)
- Arvind Pillai
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| | - Abbas Idris
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Annika Philomin
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Connor Weidle
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Rebecca Skotheim
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Philip J Y Leung
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Program in Molecular Engineering, University of Washington, Seattle, WA, USA
| | - Adam Broerman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Cullen Demakis
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure, and Design, University of Washington, Seattle, WA, USA
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Florian Praetorius
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Ge Y, Pande V, Seierstad MJ, Damm-Ganamet KL. Exploring the Application of SiteMap and Site Finder for Focused Cryptic Pocket Identification. J Phys Chem B 2024; 128:6233-6245. [PMID: 38904218 DOI: 10.1021/acs.jpcb.4c00664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
The characterization of cryptic pockets has been elusive, despite substantial efforts. Computational modeling approaches, such as molecular dynamics (MD) simulations, can provide atomic-level details of binding site motions and binding pathways. However, the time scale that MD can achieve at a reasonable cost often limits its application for cryptic pocket identification. Enhanced sampling techniques can improve the efficiency of MD simulations by focused sampling of important regions of the protein, but prior knowledge of the simulated system is required to define the appropriate coordinates. In the case of a novel, unknown cryptic pocket, such information is not available, limiting the application of enhanced sampling techniques for cryptic pocket identification. In this work, we explore the ability of SiteMap and Site Finder, widely used commercial packages for pocket identification, to detect focus points on the protein and further apply other advanced computational methods. The information gained from this analysis enables the use of computational modeling, including enhanced MD sampling techniques, to explore potential cryptic binding pockets suggested by SiteMap and Site Finder. Here, we examined SiteMap and Site Finder results on 136 known cryptic pockets from a combination of the PocketMiner dataset (a recently curated set of cryptic pockets), the Cryptosite Set (a classic set of cryptic pockets), and Natural killer group 2D (NKG2D, a protein target where a cryptic pocket is confirmed). Our findings demonstrate the application of existing, well-studied tools in efficiently mapping potential regions harboring cryptic pockets.
Collapse
Affiliation(s)
- Yunhui Ge
- Computer-Aided Drug Design, Therapeutics Discovery, Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Vineet Pande
- Computer-Aided Drug Design, Therapeutics Discovery, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Mark J Seierstad
- Computer-Aided Drug Design, Therapeutics Discovery, Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121, United States
| | - Kelly L Damm-Ganamet
- Computer-Aided Drug Design, Therapeutics Discovery, Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121, United States
| |
Collapse
|
18
|
Borsatto A, Gianquinto E, Rizzi V, Gervasio FL. SWISH-X, an Expanded Approach to Detect Cryptic Pockets in Proteins and at Protein-Protein Interfaces. J Chem Theory Comput 2024; 20:3335-3348. [PMID: 38563746 PMCID: PMC11044271 DOI: 10.1021/acs.jctc.3c01318] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
Protein-protein interactions mediate most molecular processes in the cell, offering a significant opportunity to expand the set of known druggable targets. Unfortunately, targeting these interactions can be challenging due to their typically flat and featureless interaction surfaces, which often change as the complex forms. Such surface changes may reveal hidden (cryptic) druggable pockets. Here, we analyze a set of well-characterized protein-protein interactions harboring cryptic pockets and investigate the predictive power of current computational methods. Based on our observations, we developed a new computational strategy, SWISH-X (SWISH Expanded), which combines the established cryptic pocket identification capabilities of SWISH with the rapid temperature range exploration of OPES MultiThermal. SWISH-X is able to reliably identify cryptic pockets at protein-protein interfaces while retaining its predictive power for revealing cryptic pockets in isolated proteins, such as TEM-1 β-lactamase.
Collapse
Affiliation(s)
- Alberto Borsatto
- School
of Pharmaceutical Sciences, University of
Geneva, 1205 Geneva, Switzerland
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
- Swiss
Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Eleonora Gianquinto
- Department
of Drug Science and Technology, University
of Turin, 10125 Turin, Italy
| | - Valerio Rizzi
- School
of Pharmaceutical Sciences, University of
Geneva, 1205 Geneva, Switzerland
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
- Swiss
Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Francesco Luigi Gervasio
- School
of Pharmaceutical Sciences, University of
Geneva, 1205 Geneva, Switzerland
- Institute
of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1205 Geneva, Switzerland
- Swiss
Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Department
of Chemistry, University College London, WC1 H0AJ London, United Kingdom
- Institute
of Structural and Molecular Biology, University
College London, WC1E7JE London, United Kingdom
| |
Collapse
|
19
|
Wu Y, Cao S, Qiu Y, Huang X. Tutorial on how to build non-Markovian dynamic models from molecular dynamics simulations for studying protein conformational changes. J Chem Phys 2024; 160:121501. [PMID: 38516972 PMCID: PMC10964226 DOI: 10.1063/5.0189429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Protein conformational changes play crucial roles in their biological functions. In recent years, the Markov State Model (MSM) constructed from extensive Molecular Dynamics (MD) simulations has emerged as a powerful tool for modeling complex protein conformational changes. In MSMs, dynamics are modeled as a sequence of Markovian transitions among metastable conformational states at discrete time intervals (called lag time). A major challenge for MSMs is that the lag time must be long enough to allow transitions among states to become memoryless (or Markovian). However, this lag time is constrained by the length of individual MD simulations available to track these transitions. To address this challenge, we have recently developed Generalized Master Equation (GME)-based approaches, encoding non-Markovian dynamics using a time-dependent memory kernel. In this Tutorial, we introduce the theory behind two recently developed GME-based non-Markovian dynamic models: the quasi-Markov State Model (qMSM) and the Integrative Generalized Master Equation (IGME). We subsequently outline the procedures for constructing these models and provide a step-by-step tutorial on applying qMSM and IGME to study two peptide systems: alanine dipeptide and villin headpiece. This Tutorial is available at https://github.com/xuhuihuang/GME_tutorials. The protocols detailed in this Tutorial aim to be accessible for non-experts interested in studying the biomolecular dynamics using these non-Markovian dynamic models.
Collapse
Affiliation(s)
- Yue Wu
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Siqin Cao
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Yunrui Qiu
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Xuhui Huang
- Author to whom correspondence should be addressed:
| |
Collapse
|
20
|
Meller A, Kelly D, Smith LG, Bowman GR. Toward physics-based precision medicine: Exploiting protein dynamics to design new therapeutics and interpret variants. Protein Sci 2024; 33:e4902. [PMID: 38358129 PMCID: PMC10868452 DOI: 10.1002/pro.4902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 01/04/2024] [Indexed: 02/16/2024]
Abstract
The goal of precision medicine is to utilize our knowledge of the molecular causes of disease to better diagnose and treat patients. However, there is a substantial mismatch between the small number of food and drug administration (FDA)-approved drugs and annotated coding variants compared to the needs of precision medicine. This review introduces the concept of physics-based precision medicine, a scalable framework that promises to improve our understanding of sequence-function relationships and accelerate drug discovery. We show that accounting for the ensemble of structures a protein adopts in solution with computer simulations overcomes many of the limitations imposed by assuming a single protein structure. We highlight studies of protein dynamics and recent methods for the analysis of structural ensembles. These studies demonstrate that differences in conformational distributions predict functional differences within protein families and between variants. Thanks to new computational tools that are providing unprecedented access to protein structural ensembles, this insight may enable accurate predictions of variant pathogenicity for entire libraries of variants. We further show that explicitly accounting for protein ensembles, with methods like alchemical free energy calculations or docking to Markov state models, can uncover novel lead compounds. To conclude, we demonstrate that cryptic pockets, or cavities absent in experimental structures, provide an avenue to target proteins that are currently considered undruggable. Taken together, our review provides a roadmap for the field of protein science to accelerate precision medicine.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular BiophysicsWashington University in St. LouisSt. LouisMissouriUSA
- Medical Scientist Training ProgramWashington University in St. LouisSt. LouisMissouriUSA
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Devin Kelly
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Louis G. Smith
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Gregory R. Bowman
- Departments of Biochemistry & Biophysics and BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
21
|
Arbon R, Zhu Y, Mey ASJS. Markov State Models: To Optimize or Not to Optimize. J Chem Theory Comput 2024; 20:977-988. [PMID: 38163961 PMCID: PMC10809420 DOI: 10.1021/acs.jctc.3c01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/10/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Markov state models (MSM) are a popular statistical method for analyzing the conformational dynamics of proteins including protein folding. With all statistical and machine learning (ML) models, choices must be made about the modeling pipeline that cannot be directly learned from the data. These choices, or hyperparameters, are often evaluated by expert judgment or, in the case of MSMs, by maximizing variational scores such as the VAMP-2 score. Modern ML and statistical pipelines often use automatic hyperparameter selection techniques ranging from the simple, choosing the best score from a random selection of hyperparameters, to the complex, optimization via, e.g., Bayesian optimization. In this work, we ask whether it is possible to automatically select MSM models this way by estimating and analyzing over 16,000,000 observations from over 280,000 estimated MSMs. We find that differences in hyperparameters can change the physical interpretation of the optimization objective, making automatic selection difficult. In addition, we find that enforcing conditions of equilibrium in the VAMP scores can result in inconsistent model selection. However, other parameters that specify the VAMP-2 score (lag time and number of relaxation processes scored) have only a negligible influence on model selection. We suggest that model observables and variational scores should be only a guide to model selection and that a full investigation of the MSM properties should be undertaken when selecting hyperparameters.
Collapse
Affiliation(s)
- Robert
E. Arbon
- EaStCHEM
School of Chemistry, David Brewster Road, Joseph Black Building, The King’s Buildings, Edinburgh EH9 3FJ, United Kingdom
- Redesign
Science, 180 Varick St., New York, New York 10014, United States
| | - Yanchen Zhu
- EaStCHEM
School of Chemistry, David Brewster Road, Joseph Black Building, The King’s Buildings, Edinburgh EH9 3FJ, United Kingdom
| | - Antonia S. J. S. Mey
- EaStCHEM
School of Chemistry, David Brewster Road, Joseph Black Building, The King’s Buildings, Edinburgh EH9 3FJ, United Kingdom
| |
Collapse
|
22
|
Li M, Lan X, Lu X, Zhang J. A Structure-Based Allosteric Modulator Design Paradigm. HEALTH DATA SCIENCE 2023; 3:0094. [PMID: 38487194 PMCID: PMC10904074 DOI: 10.34133/hds.0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 10/11/2023] [Indexed: 03/17/2024]
Abstract
Importance: Allosteric drugs bound to topologically distal allosteric sites hold a substantial promise in modulating therapeutic targets deemed undruggable at their orthosteric sites. Traditionally, allosteric modulator discovery has predominantly relied on serendipitous high-throughput screening. Nevertheless, the landscape has undergone a transformative shift due to recent advancements in our understanding of allosteric modulation mechanisms, coupled with a significant increase in the accessibility of allosteric structural data. These factors have extensively promoted the development of various computational methodologies, especially for machine-learning approaches, to guide the rational design of structure-based allosteric modulators. Highlights: We here presented a comprehensive structure-based allosteric modulator design paradigm encompassing 3 critical stages: drug target acquisition, allosteric binding site, and modulator discovery. The recent advances in computational methods in each stage are encapsulated. Furthermore, we delve into analyzing the successes and obstacles encountered in the rational design of allosteric modulators. Conclusion: The structure-based allosteric modulator design paradigm holds immense potential for the rational design of allosteric modulators. We hope that this review would heighten awareness of the use of structure-based computational methodologies in advancing the field of allosteric drug discovery.
Collapse
Affiliation(s)
- Mingyu Li
- College of Pharmacy,
Ningxia Medical University, Yinchuan, NingxiaHui Autonomous Region, China
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaobin Lan
- College of Pharmacy,
Ningxia Medical University, Yinchuan, NingxiaHui Autonomous Region, China
- Medicinal Chemistry and Bioinformatics Center,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xun Lu
- College of Pharmacy,
Ningxia Medical University, Yinchuan, NingxiaHui Autonomous Region, China
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Zhang
- College of Pharmacy,
Ningxia Medical University, Yinchuan, NingxiaHui Autonomous Region, China
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
23
|
Ahmed F, Brooks CL. FASTDock: A Pipeline for Allosteric Drug Discovery. J Chem Inf Model 2023; 63:7219-7227. [PMID: 37939386 PMCID: PMC10773972 DOI: 10.1021/acs.jcim.3c00895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Allostery is involved in innumerable biological processes and plays a fundamental role in human disease. Thus, the exploration of allosteric modulation is crucial for research on biological mechanisms and in the development of novel therapeutics. The development of small-molecule allosteric effectors can be used as tools to probe biological mechanisms of interest. One of the main limitations in targeting allosteric sites is the difficulty in uncovering them for specific receptors. Furthermore, upon discovery of novel allosteric modulation, early lead generation is made more difficult as compared to that at orthosteric sites because there is likely no information about the types of molecules that can bind at the site. In the work described here, we present a novel drug discovery pipeline, FASTDock, which allows one to uncover ligandable sites as well as small molecules that target the given site without requiring pre-existing knowledge of ligands that can bind in the targeted site. By using a hierarchical screening strategy, this method has the potential to enable high-throughput screens of an exceptionally large database of targeted ligand space.
Collapse
Affiliation(s)
- Furyal Ahmed
- Biophysics Program, University of Michigan, Ann Arbor, MI 48103
| | - Charles L. Brooks
- Department of Chemistry and Biophysics Program, University of Michigan, Ann Arbor, MI 48103
| |
Collapse
|
24
|
Alshahrani M, Gupta G, Xiao S, Tao P, Verkhivker G. Comparative Analysis of Conformational Dynamics and Systematic Characterization of Cryptic Pockets in the SARS-CoV-2 Omicron BA.2, BA.2.75 and XBB.1 Spike Complexes with the ACE2 Host Receptor: Confluence of Binding and Structural Plasticity in Mediating Networks of Conserved Allosteric Sites. Viruses 2023; 15:2073. [PMID: 37896850 PMCID: PMC10612107 DOI: 10.3390/v15102073] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
In the current study, we explore coarse-grained simulations and atomistic molecular dynamics together with binding energetics scanning and cryptic pocket detection in a comparative examination of conformational landscapes and systematic characterization of allosteric binding sites in the SARS-CoV-2 Omicron BA.2, BA.2.75 and XBB.1 spike full-length trimer complexes with the host receptor ACE2. Microsecond simulations, Markov state models and mutational scanning of binding energies of the SARS-CoV-2 BA.2 and BA.2.75 receptor binding domain complexes revealed the increased thermodynamic stabilization of the BA.2.75 variant and significant dynamic differences between these Omicron variants. Molecular simulations of the SARS-CoV-2 Omicron spike full-length trimer complexes with the ACE2 receptor complemented atomistic studies and enabled an in-depth analysis of mutational and binding effects on conformational dynamic and functional adaptability of the Omicron variants. Despite considerable structural similarities, Omicron variants BA.2, BA.2.75 and XBB.1 can induce unique conformational dynamic signatures and specific distributions of the conformational states. Using conformational ensembles of the SARS-CoV-2 Omicron spike trimer complexes with ACE2, we conducted a comprehensive cryptic pocket screening to examine the role of Omicron mutations and ACE2 binding on the distribution and functional mechanisms of the emerging allosteric binding sites. This analysis captured all experimentally known allosteric sites and discovered networks of inter-connected and functionally relevant allosteric sites that are governed by variant-sensitive conformational adaptability of the SARS-CoV-2 spike structures. The results detailed how ACE2 binding and Omicron mutations in the BA.2, BA.2.75 and XBB.1 spike complexes modulate the distribution of conserved and druggable allosteric pockets harboring functionally important regions. The results are significant for understanding the functional roles of druggable cryptic pockets that can be used for allostery-mediated therapeutic intervention targeting conformational states of the Omicron variants.
Collapse
Affiliation(s)
- Mohammed Alshahrani
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
| | - Grace Gupta
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
| | - Sian Xiao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, TX 75275, USA; (S.X.); (P.T.)
| | - Peng Tao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, TX 75275, USA; (S.X.); (P.T.)
| | - Gennady Verkhivker
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA; (M.A.); (G.G.)
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
25
|
Hellemann E, Durrant JD. Worth the Weight: Sub-Pocket EXplorer (SubPEx), a Weighted Ensemble Method to Enhance Binding-Pocket Conformational Sampling. J Chem Theory Comput 2023; 19:5677-5689. [PMID: 37585617 PMCID: PMC10500992 DOI: 10.1021/acs.jctc.3c00478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Indexed: 08/18/2023]
Abstract
Structure-based virtual screening (VS) is an effective method for identifying potential small-molecule ligands, but traditional VS approaches consider only a single binding-pocket conformation. Consequently, they struggle to identify ligands that bind to alternate conformations. Ensemble docking helps address this issue by incorporating multiple conformations into the docking process, but it depends on methods that can thoroughly explore pocket flexibility. We here introduce Sub-Pocket EXplorer (SubPEx), an approach that uses weighted ensemble (WE) path sampling to accelerate binding-pocket sampling. As proof of principle, we apply SubPEx to three proteins relevant to drug discovery: heat shock protein 90, influenza neuraminidase, and yeast hexokinase 2. SubPEx is available free of charge without registration under the terms of the open-source MIT license: http://durrantlab.com/subpex/.
Collapse
Affiliation(s)
- Erich Hellemann
- Department of Biological
Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Jacob D. Durrant
- Department of Biological
Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
26
|
Alshahrani M, Gupta G, Xiao S, Tao P, Verkhivker G. Examining Functional Linkages Between Conformational Dynamics, Protein Stability and Evolution of Cryptic Binding Pockets in the SARS-CoV-2 Omicron Spike Complexes with the ACE2 Host Receptor: Recombinant Omicron Variants Mediate Variability of Conserved Allosteric Sites and Binding Epitopes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557205. [PMID: 37745525 PMCID: PMC10515794 DOI: 10.1101/2023.09.11.557205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
In the current study, we explore coarse-grained simulations and atomistic molecular dynamics together with binding energetics scanning and cryptic pocket detection in a comparative examination of conformational landscapes and systematic characterization of allosteric binding sites in the SARS-CoV-2 Omicron BA.2, BA.2.75 and XBB.1 spike full-length trimer complexes with the host receptor ACE2. Microsecond simulations, Markov state models and mutational scanning of binding energies of the SARS-CoV-2 BA.2 and BA.2.75 receptor binding domain complexes revealed the increased thermodynamic stabilization of the BA.2.75 variant and significant dynamic differences between these Omicron variants. Molecular simulations of the SARS-CoV-2 Omicron spike full length trimer complexes with the ACE2 receptor complemented atomistic studies and enabled an in-depth analysis of mutational and binding effects on conformational dynamic and functional adaptability of the Omicron variants. Despite considerable structural similarities, Omicron variants BA.2, BA.2.75 and XBB.1 can induce unique conformational dynamic signatures and specific distributions of the conformational states. Using conformational ensembles of the SARS-CoV-2 Omicron spike trimer complexes with ACE2, we conducted a comprehensive cryptic pocket screening to examine the role of Omicron mutations and ACE2 binding on the distribution and functional mechanisms of the emerging allosteric binding sites. This analysis captured all experimentally known allosteric sites and discovered networks of inter-connected and functionally relevant allosteric sites that are governed by variant-sensitive conformational adaptability of the SARS-CoV-2 spike structures. The results detailed how ACE2 binding and Omicron mutations in the BA.2, BA.2.75 and XBB.1 spike complexes modulate the distribution of conserved and druggable allosteric pockets harboring functionally important regions. The results of are significant for understanding functional roles of druggable cryptic pockets that can be used for allostery-mediated therapeutic intervention targeting conformational states of the Omicron variants.
Collapse
|
27
|
Xiao S, Alshahrani M, Gupta G, Tao P, Verkhivker G. Markov State Models and Perturbation-Based Approaches Reveal Distinct Dynamic Signatures and Hidden Allosteric Pockets in the Emerging SARS-Cov-2 Spike Omicron Variant Complexes with the Host Receptor: The Interplay of Dynamics and Convergent Evolution Modulates Allostery and Functional Mechanisms. J Chem Inf Model 2023; 63:5272-5296. [PMID: 37549201 PMCID: PMC11162552 DOI: 10.1021/acs.jcim.3c00778] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
The new generation of SARS-CoV-2 Omicron variants displayed a significant growth advantage and increased viral fitness by acquiring convergent mutations, suggesting that the immune pressure can promote convergent evolution leading to the sudden acceleration of SARS-CoV-2 evolution. In the current study, we combined structural modeling, microsecond molecular dynamics simulations, and Markov state models to characterize conformational landscapes and identify specific dynamic signatures of the SARS-CoV-2 spike complexes with the host receptor ACE2 for the recently emerged highly transmissible XBB.1, XBB.1.5, BQ.1, and BQ.1.1 Omicron variants. Microsecond simulations and Markovian modeling provided a detailed characterization of the functional conformational states and revealed the increased thermodynamic stabilization of the XBB.1.5 subvariant, which can be contrasted to more dynamic BQ.1 and BQ.1.1 subvariants. Despite considerable structural similarities, Omicron mutations can induce unique dynamic signatures and specific distributions of the conformational states. The results suggested that variant-specific changes of the conformational mobility in the functional interfacial loops of the receptor-binding domain in the SARS-CoV-2 spike protein can be fine-tuned through crosstalk between convergent mutations which could provide an evolutionary path for modulation of immune escape. By combining atomistic simulations and Markovian modeling analysis with perturbation-based approaches, we determined important complementary roles of convergent mutation sites as effectors and receivers of allosteric signaling involved in modulation of conformational plasticity and regulation of allosteric communications. This study also revealed hidden allosteric pockets and suggested that convergent mutation sites could control evolution and distribution of allosteric pockets through modulation of conformational plasticity in the flexible adaptable regions.
Collapse
Affiliation(s)
- Sian Xiao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States
| | - Mohammed Alshahrani
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
| | - Grace Gupta
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
| | - Peng Tao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States
| | - Gennady Verkhivker
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, United States
| |
Collapse
|
28
|
Voelz VA, Pande VS, Bowman GR. Folding@home: Achievements from over 20 years of citizen science herald the exascale era. Biophys J 2023; 122:2852-2863. [PMID: 36945779 PMCID: PMC10398258 DOI: 10.1016/j.bpj.2023.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/26/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Simulations of biomolecules have enormous potential to inform our understanding of biology but require extremely demanding calculations. For over 20 years, the Folding@home distributed computing project has pioneered a massively parallel approach to biomolecular simulation, harnessing the resources of citizen scientists across the globe. Here, we summarize the scientific and technical advances this perspective has enabled. As the project's name implies, the early years of Folding@home focused on driving advances in our understanding of protein folding by developing statistical methods for capturing long-timescale processes and facilitating insight into complex dynamical processes. Success laid a foundation for broadening the scope of Folding@home to address other functionally relevant conformational changes, such as receptor signaling, enzyme dynamics, and ligand binding. Continued algorithmic advances, hardware developments such as graphics processing unit (GPU)-based computing, and the growing scale of Folding@home have enabled the project to focus on new areas where massively parallel sampling can be impactful. While previous work sought to expand toward larger proteins with slower conformational changes, new work focuses on large-scale comparative studies of different protein sequences and chemical compounds to better understand biology and inform the development of small-molecule drugs. Progress on these fronts enabled the community to pivot quickly in response to the COVID-19 pandemic, expanding to become the world's first exascale computer and deploying this massive resource to provide insight into the inner workings of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus and aid the development of new antivirals. This success provides a glimpse of what is to come as exascale supercomputers come online and as Folding@home continues its work.
Collapse
Affiliation(s)
- Vincent A Voelz
- Department of Chemistry, Temple University, Philadelphia, Pennsylvania
| | | | - Gregory R Bowman
- Departments of Biochemistry & Biophysics and of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
29
|
Hardie A, Cossins BP, Lovera S, Michel J. Deconstructing allostery by computational assessment of the binding determinants of allosteric PTP1B modulators. Commun Chem 2023; 6:125. [PMID: 37322137 PMCID: PMC10272186 DOI: 10.1038/s42004-023-00926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
Fragment-based drug discovery is an established methodology for finding hit molecules that can be elaborated into lead compounds. However it is currently challenging to predict whether fragment hits that do not bind to an orthosteric site could be elaborated into allosteric modulators, as in these cases binding does not necessarily translate into a functional effect. We propose a workflow using Markov State Models (MSMs) with steered molecular dynamics (sMD) to assess the allosteric potential of known binders. sMD simulations are employed to sample protein conformational space inaccessible to routine equilibrium MD timescales. Protein conformations sampled by sMD provide starting points for seeded MD simulations, which are combined into MSMs. The methodology is demonstrated on a dataset of protein tyrosine phosphatase 1B ligands. Experimentally confirmed allosteric inhibitors are correctly classified as inhibitors, whereas the deconstructed analogues show reduced inhibitory activity. Analysis of the MSMs provide insights into preferred protein-ligand arrangements that correlate with functional outcomes. The present methodology may find applications for progressing fragments towards lead molecules in FBDD campaigns.
Collapse
Affiliation(s)
- Adele Hardie
- EaStChem School of Chemistry, Joseph Black Building, University of Edinburgh, Edinburgh, EH9 3FJ, UK
| | - Benjamin P Cossins
- UCB Pharma, 216 Bath Road, Slough, UK
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford, UK
| | - Silvia Lovera
- UCB Pharma, Chemin du Foriest 1, 1420, Braine-l'Alleud, Belgium
| | - Julien Michel
- EaStChem School of Chemistry, Joseph Black Building, University of Edinburgh, Edinburgh, EH9 3FJ, UK.
| |
Collapse
|
30
|
Xiao S, Alshahrani M, Gupta G, Tao P, Verkhivker G. Markov State Models and Perturbation-Based Approaches Reveal Distinct Dynamic Signatures and Hidden Allosteric Pockets in the Emerging SARS-Cov-2 Spike Omicron Variants Complexes with the Host Receptor: The Interplay of Dynamics and Convergent Evolution Modulates Allostery and Functional Mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.20.541592. [PMID: 37292827 PMCID: PMC10245745 DOI: 10.1101/2023.05.20.541592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The new generation of SARS-CoV-2 Omicron variants displayed a significant growth advantage and the increased viral fitness by acquiring convergent mutations, suggesting that the immune pressure can promote convergent evolution leading to the sudden acceleration of SARS-CoV-2 evolution. In the current study, we combined structural modeling, extensive microsecond MD simulations and Markov state models to characterize conformational landscapes and identify specific dynamic signatures of the SARS-CoV-2 spike complexes with the host receptor ACE2 for the recently emerged highly transmissible XBB.1, XBB.1.5, BQ.1, and BQ.1.1 Omicron variants. Microsecond simulations and Markovian modeling provided a detailed characterization of the conformational landscapes and revealed the increased thermodynamic stabilization of the XBB.1.5 subvariant which is contrasted to more dynamic BQ.1 and BQ.1.1 subvariants. Despite considerable structural similarities, Omicron mutations can induce unique dynamic signatures and specific distributions of conformational states. The results suggested that variant-specific changes of conformational mobility in the functional interfacial loops of the spike receptor binding domain can be fine-tuned through cross-talk between convergent mutations thereby providing an evolutionary path for modulation of immune escape. By combining atomistic simulations and Markovian modeling analysis with perturbation-based approaches, we determined important complementary roles of convergent mutation sites as effectors and receivers of allosteric signaling involved in modulating conformational plasticity at the binding interface and regulating allosteric responses. This study also characterized the dynamics-induced evolution of allosteric pockets in the Omicron complexes that revealed hidden allosteric pockets and suggested that convergent mutation sites could control evolution and distribution of allosteric pockets through modulation of conformational plasticity in the flexible adaptable regions. Through integrative computational approaches, this investigation provides a systematic analysis and comparison of the effects of Omicron subvariants on conformational dynamics and allosteric signaling in the complexes with the ACE2 receptor. For Table of Contents Use Only
Collapse
|
31
|
Hellemann E, Durrant JD. Worth the weight: Sub-Pocket EXplorer (SubPEx), a weighted-ensemble method to enhance binding-pocket conformational sampling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539330. [PMID: 37251500 PMCID: PMC10214482 DOI: 10.1101/2023.05.03.539330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Structure-based virtual screening (VS) is an effective method for identifying potential small-molecule ligands, but traditional VS approaches consider only a single binding-pocket conformation. Consequently, they struggle to identify ligands that bind to alternate conformations. Ensemble docking helps address this issue by incorporating multiple conformations into the docking process, but it depends on methods that can thoroughly explore pocket flexibility. We here introduce Sub-Pocket EXplorer (SubPEx), an approach that uses weighted ensemble (WE) path sampling to accelerate binding-pocket sampling. As proof of principle, we apply SubPEx to three proteins relevant to drug discovery: heat shock protein 90, influenza neuraminidase, and yeast hexokinase 2. SubPEx is available free of charge without registration under the terms of the open-source MIT license: http://durrantlab.com/subpex/.
Collapse
Affiliation(s)
- Erich Hellemann
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, United States
| | - Jacob D. Durrant
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15260, United States
| |
Collapse
|
32
|
Dominic AJ, 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; 145:9916-9927. [PMID: 37104720 DOI: 10.1021/jacs.3c01095] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Conformational changes underpin function and encode complex biomolecular mechanisms. Gaining atomic-level detail of how such changes occur has the potential to reveal these mechanisms and is of critical 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 resolution 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 biomolecular 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 molecular 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.
Collapse
Affiliation(s)
- Anthony J Dominic
- Department of Chemistry, University of Colorado Boulder, Boulder, Colorado 80309, USA
| | - Siqin Cao
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | - Xuhui Huang
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
33
|
Meller A, De Oliveira S, Davtyan A, Abramyan T, Bowman GR, van den Bedem H. Discovery of a cryptic pocket in the AI-predicted structure of PPM1D phosphatase explains the binding site and potency of its allosteric inhibitors. Front Mol Biosci 2023; 10:1171143. [PMID: 37143823 PMCID: PMC10151774 DOI: 10.3389/fmolb.2023.1171143] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/07/2023] [Indexed: 05/06/2023] Open
Abstract
Virtual screening is a widely used tool for drug discovery, but its predictive power can vary dramatically depending on how much structural data is available. In the best case, crystal structures of a ligand-bound protein can help find more potent ligands. However, virtual screens tend to be less predictive when only ligand-free crystal structures are available, and even less predictive if a homology model or other predicted structure must be used. Here, we explore the possibility that this situation can be improved by better accounting for protein dynamics, as simulations started from a single structure have a reasonable chance of sampling nearby structures that are more compatible with ligand binding. As a specific example, we consider the cancer drug target PPM1D/Wip1 phosphatase, a protein that lacks crystal structures. High-throughput screens have led to the discovery of several allosteric inhibitors of PPM1D, but their binding mode remains unknown. To enable further drug discovery efforts, we assessed the predictive power of an AlphaFold-predicted structure of PPM1D and a Markov state model (MSM) built from molecular dynamics simulations initiated from that structure. Our simulations reveal a cryptic pocket at the interface between two important structural elements, the flap and hinge regions. Using deep learning to predict the pose quality of each docked compound for the active site and cryptic pocket suggests that the inhibitors strongly prefer binding to the cryptic pocket, consistent with their allosteric effect. The predicted affinities for the dynamically uncovered cryptic pocket also recapitulate the relative potencies of the compounds (τb = 0.70) better than the predicted affinities for the static AlphaFold-predicted structure (τb = 0.42). Taken together, these results suggest that targeting the cryptic pocket is a good strategy for drugging PPM1D and, more generally, that conformations selected from simulation can improve virtual screening when limited structural data is available.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, United States
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, United States
| | | | - Aram Davtyan
- Atomwise, Inc., San Francisco, CA, United States
| | | | - Gregory R. Bowman
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, United States
| | - Henry van den Bedem
- Atomwise, Inc., San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, United States
| |
Collapse
|
34
|
Meller A, de Oliveira S, Davtyan A, Abramyan T, Bowman GR, van den Bedem H. Discovery of a cryptic pocket in the AI-predicted structure of PPM1D phosphatase explains the binding site and potency of its allosteric inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533829. [PMID: 36993233 PMCID: PMC10055338 DOI: 10.1101/2023.03.22.533829] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Virtual screening is a widely used tool for drug discovery, but its predictive power can vary dramatically depending on how much structural data is available. In the best case, crystal structures of a ligand-bound protein can help find more potent ligands. However, virtual screens tend to be less predictive when only ligand-free crystal structures are available, and even less predictive if a homology model or other predicted structure must be used. Here, we explore the possibility that this situation can be improved by better accounting for protein dynamics, as simulations started from a single structure have a reasonable chance of sampling nearby structures that are more compatible with ligand binding. As a specific example, we consider the cancer drug target PPM1D/Wip1 phosphatase, a protein that lacks crystal structures. High-throughput screens have led to the discovery of several allosteric inhibitors of PPM1D, but their binding mode remains unknown. To enable further drug discovery efforts, we assessed the predictive power of an AlphaFold-predicted structure of PPM1D and a Markov state model (MSM) built from molecular dynamics simulations initiated from that structure. Our simulations reveal a cryptic pocket at the interface between two important structural elements, the flap and hinge regions. Using deep learning to predict the pose quality of each docked compound for the active site and cryptic pocket suggests that the inhibitors strongly prefer binding to the cryptic pocket, consistent with their allosteric effect. The predicted affinities for the dynamically uncovered cryptic pocket also recapitulate the relative potencies of the compounds (τ b =0.70) better than the predicted affinities for the static AlphaFold-predicted structure (τ b =0.42). Taken together, these results suggest that targeting the cryptic pocket is a good strategy for drugging PPM1D and, more generally, that conformations selected from simulation can improve virtual screening when limited structural data is available.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S Euclid Ave, St. Louis, MO, 63110
- Medical Scientist Training Program, Washington University in St. Louis, 660 S Euclid Ave., St. Louis, MO, 63110
| | - Saulo de Oliveira
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Aram Davtyan
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Tigran Abramyan
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
| | - Gregory R. Bowman
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA, 19104
| | - Henry van den Bedem
- Atomwise, Inc., 717 Market Street, Suite 800, San Francisco, California 94103
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
35
|
Xie J, Pan G, Li Y, Lai L. How protein topology controls allosteric regulations. J Chem Phys 2023; 158:105102. [PMID: 36922138 DOI: 10.1063/5.0138279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Allostery is an important regulatory mechanism of protein functions. Among allosteric proteins, certain protein structure types are more observed. However, how allosteric regulation depends on protein topology remains elusive. In this study, we extracted protein topology graphs at the fold level and found that known allosteric proteins mainly contain multiple domains or subunits and allosteric sites reside more often between two or more domains of the same fold type. Only a small fraction of fold-fold combinations are observed in allosteric proteins, and homo-fold-fold combinations dominate. These analyses imply that the locations of allosteric sites including cryptic ones depend on protein topology. We further developed TopoAlloSite, a novel method that uses the kernel support vector machine to predict the location of allosteric sites on the overall protein topology based on the subgraph-matching kernel. TopoAlloSite successfully predicted known cryptic allosteric sites in several allosteric proteins like phosphopantothenoylcysteine synthetase, spermidine synthase, and sirtuin 6, demonstrating its power in identifying cryptic allosteric sites without performing long molecular dynamics simulations or large-scale experimental screening. Our study demonstrates that protein topology largely determines how its function can be allosterically regulated, which can be used to find new druggable targets and locate potential binding sites for rational allosteric drug design.
Collapse
Affiliation(s)
- Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Gaoxiang Pan
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yibo Li
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
36
|
Agajanian S, Alshahrani M, Bai F, Tao P, Verkhivker GM. Exploring and Learning the Universe of Protein Allostery Using Artificial Intelligence Augmented Biophysical and Computational Approaches. J Chem Inf Model 2023; 63:1413-1428. [PMID: 36827465 PMCID: PMC11162550 DOI: 10.1021/acs.jcim.2c01634] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Abstract
Allosteric mechanisms are commonly employed regulatory tools used by proteins to orchestrate complex biochemical processes and control communications in cells. The quantitative understanding and characterization of allosteric molecular events are among major challenges in modern biology and require integration of innovative computational experimental approaches to obtain atomistic-level knowledge of the allosteric states, interactions, and dynamic conformational landscapes. The growing body of computational and experimental studies empowered by emerging artificial intelligence (AI) technologies has opened up new paradigms for exploring and learning the universe of protein allostery from first principles. In this review we analyze recent developments in high-throughput deep mutational scanning of allosteric protein functions; applications and latest adaptations of Alpha-fold structural prediction methods for studies of protein dynamics and allostery; new frontiers in integrating machine learning and enhanced sampling techniques for characterization of allostery; and recent advances in structural biology approaches for studies of allosteric systems. We also highlight recent computational and experimental studies of the SARS-CoV-2 spike (S) proteins revealing an important and often hidden role of allosteric regulation driving functional conformational changes, binding interactions with the host receptor, and mutational escape mechanisms of S proteins which are critical for viral infection. We conclude with a summary and outlook of future directions suggesting that AI-augmented biophysical and computer simulation approaches are beginning to transform studies of protein allostery toward systematic characterization of allosteric landscapes, hidden allosteric states, and mechanisms which may bring about a new revolution in molecular biology and drug discovery.
Collapse
Affiliation(s)
- Steve Agajanian
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
| | - Mohammed Alshahrani
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
| | - Fang Bai
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology and Information Science and Technology, Shanghai Tech University, 393 Middle Huaxia Road, Shanghai 201210, China
| | - Peng Tao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75205, United States
| | - Gennady M Verkhivker
- Keck Center for Science and Engineering, Graduate Program in Computational and Data Sciences, Schmid College of Science and Technology, Chapman University, Orange, California 92866, United States
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, United States
| |
Collapse
|
37
|
Meller A, Ward M, Borowsky J, Kshirsagar M, Lotthammer JM, Oviedo F, Ferres JL, Bowman GR. Predicting locations of cryptic pockets from single protein structures using the PocketMiner graph neural network. Nat Commun 2023; 14:1177. [PMID: 36859488 PMCID: PMC9977097 DOI: 10.1038/s41467-023-36699-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
Cryptic pockets expand the scope of drug discovery by enabling targeting of proteins currently considered undruggable because they lack pockets in their ground state structures. However, identifying cryptic pockets is labor-intensive and slow. The ability to accurately and rapidly predict if and where cryptic pockets are likely to form from a structure would greatly accelerate the search for druggable pockets. Here, we present PocketMiner, a graph neural network trained to predict where pockets are likely to open in molecular dynamics simulations. Applying PocketMiner to single structures from a newly curated dataset of 39 experimentally confirmed cryptic pockets demonstrates that it accurately identifies cryptic pockets (ROC-AUC: 0.87) >1,000-fold faster than existing methods. We apply PocketMiner across the human proteome and show that predicted pockets open in simulations, suggesting that over half of proteins thought to lack pockets based on available structures likely contain cryptic pockets, vastly expanding the potentially druggable proteome.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S. Euclid Ave., Box 8231, St. Louis, MO, 63110, USA
- Medical Scientist Training Program, Washington University in St. Louis, 660 S. Euclid Ave., St. Louis, MO, 63110, USA
| | - Michael Ward
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S. Euclid Ave., Box 8231, St. Louis, MO, 63110, USA
| | - Jonathan Borowsky
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S. Euclid Ave., Box 8231, St. Louis, MO, 63110, USA
| | | | - Jeffrey M Lotthammer
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S. Euclid Ave., Box 8231, St. Louis, MO, 63110, USA
| | - Felipe Oviedo
- AI for Good Research Lab, Microsoft, Redmond, WA, USA
| | | | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, 660 S. Euclid Ave., Box 8231, St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
38
|
Moreno JD, Silva JR. Emerging methods to model cardiac ion channel and myocyte electrophysiology. BIOPHYSICS REVIEWS 2023; 4:011315. [PMID: 37034130 PMCID: PMC10071990 DOI: 10.1063/5.0127713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/28/2023] [Indexed: 04/03/2023]
Abstract
In the field of cardiac electrophysiology, modeling has played a central role for many decades. However, even though the effort is well-established, it has recently seen a rapid and sustained evolution in the complexity and predictive power of the models being created. In particular, new approaches to modeling have allowed the tracking of parallel and interconnected processes that span from the nanometers and femtoseconds that determine ion channel gating to the centimeters and minutes needed to describe an arrhythmia. The connection between scales has brought unprecedented insight into cardiac arrhythmia mechanisms and drug therapies. This review focuses on the generation of these models from first principles, generation of detailed models to describe ion channel kinetics, algorithms to create and numerically solve kinetic models, and new approaches toward data gathering that parameterize these models. While we focus on application of these models for cardiac arrhythmia, these concepts are widely applicable to model the physiology and pathophysiology of any excitable cell.
Collapse
Affiliation(s)
- Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | - Jonathan R. Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| |
Collapse
|
39
|
Ojha AA, Thakur S, Ahn SH, Amaro RE. DeepWEST: Deep Learning of Kinetic Models with the Weighted Ensemble Simulation Toolkit for Enhanced Sampling. J Chem Theory Comput 2023; 19:1342-1359. [PMID: 36719802 DOI: 10.1021/acs.jctc.2c00282] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Recent advances in computational power and algorithms have enabled molecular dynamics (MD) simulations to reach greater time scales. However, for observing conformational transitions associated with biomolecular processes, MD simulations still have limitations. Several enhanced sampling techniques seek to address this challenge, including the weighted ensemble (WE) method, which samples transitions between metastable states using many weighted trajectories to estimate kinetic rate constants. However, initial sampling of the potential energy surface has a significant impact on the performance of WE, i.e., convergence and efficiency. We therefore introduce deep-learned kinetic modeling approaches that extract statistically relevant information from short MD trajectories to provide a well-sampled initial state distribution for WE simulations. This hybrid approach overcomes any statistical bias to the system, as it runs short unbiased MD trajectories and identifies meaningful metastable states of the system. It is shown to provide a more refined free energy landscape closer to the steady state that could efficiently sample kinetic properties such as rate constants.
Collapse
Affiliation(s)
- Anupam Anand Ojha
- Department of Chemistry, University of California San Diego, La Jolla, California92093, United States
| | - Saumya Thakur
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra400076, India
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California Davis, Davis, California95616, United States
| | - Rommie E Amaro
- Department of Chemistry, University of California San Diego, La Jolla, California92093, United States
| |
Collapse
|
40
|
Meller A, Lotthammer JM, Smith LG, Novak B, Lee LA, Kuhn CC, Greenberg L, Leinwand LA, Greenberg MJ, Bowman GR. Drug specificity and affinity are encoded in the probability of cryptic pocket opening in myosin motor domains. eLife 2023; 12:e83602. [PMID: 36705568 PMCID: PMC9995120 DOI: 10.7554/elife.83602] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The design of compounds that can discriminate between closely related target proteins remains a central challenge in drug discovery. Specific therapeutics targeting the highly conserved myosin motor family are urgently needed as mutations in at least six of its members cause numerous diseases. Allosteric modulators, like the myosin-II inhibitor blebbistatin, are a promising means to achieve specificity. However, it remains unclear why blebbistatin inhibits myosin-II motors with different potencies given that it binds at a highly conserved pocket that is always closed in blebbistatin-free experimental structures. We hypothesized that the probability of pocket opening is an important determinant of the potency of compounds like blebbistatin. To test this hypothesis, we used Markov state models (MSMs) built from over 2 ms of aggregate molecular dynamics simulations with explicit solvent. We find that blebbistatin's binding pocket readily opens in simulations of blebbistatin-sensitive myosin isoforms. Comparing these conformational ensembles reveals that the probability of pocket opening correctly identifies which isoforms are most sensitive to blebbistatin inhibition and that docking against MSMs quantitatively predicts blebbistatin binding affinities (R2=0.82). In a blind prediction for an isoform (Myh7b) whose blebbistatin sensitivity was unknown, we find good agreement between predicted and measured IC50s (0.67 μM vs. 0.36 μM). Therefore, we expect this framework to be useful for the development of novel specific drugs across numerous protein targets.
Collapse
Affiliation(s)
- Artur Meller
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
- Medical Scientist Training Program, Washington University in St. LouisPhiladelphiaUnited States
| | - Jeffrey M Lotthammer
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
| | - Louis G Smith
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
- Department of Biochemistry and Biophysics, University of PennsylvaniaPhiladelphiaUnited States
| | - Borna Novak
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
- Medical Scientist Training Program, Washington University in St. LouisPhiladelphiaUnited States
| | - Lindsey A Lee
- Molecular, Cellular, and Developmental Biology Department, University of Colorado BoulderBoulderUnited States
- BioFrontiers InstituteBoulderUnited States
| | - Catherine C Kuhn
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
| | - Lina Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
| | - Leslie A Leinwand
- Molecular, Cellular, and Developmental Biology Department, University of Colorado BoulderBoulderUnited States
- BioFrontiers InstituteBoulderUnited States
| | - Michael J Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University in St. LouisSt LouisUnited States
- Department of Biochemistry and Biophysics, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
41
|
Clemente F, Davighi MG, Matassini C, Cardona F, Goti A, Morrone A, Paoli P, Tejero T, Merino P, Cacciarini M. Light-Triggered Control of Glucocerebrosidase Inhibitors: Towards Photoswitchable Pharmacological Chaperones. Chemistry 2023; 29:e202203841. [PMID: 36598148 DOI: 10.1002/chem.202203841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/05/2023]
Abstract
Piperidine-based photoswitchable derivatives have been developed as putative pharmacological chaperones for glucocerebrosidase (GCase), the defective enzyme in Gaucher disease (GD). The structure-activity study revealed that both the iminosugar and the light-sensitive azobenzene are essential features to exert inhibitory activity towards human GCase and a system with the correct inhibition trend (IC50 of the light-activated form lower than IC50 of the dark form) was identified. Kinetic analyses showed that all compounds are non-competitive inhibitors (mixed or pure) of GCase and the enzyme allosteric site involved in the interaction was identified by means of MD simulations. A moderate activity enhancement of mutant GCase assessed in GD patients' fibroblasts (ex vivo experiments) carrying the most common mutation was recorded. This promising observation paves the way for further studies to improve the benefit of the light-to-dark thermal conversion for chaperoning activity.
Collapse
Affiliation(s)
- Francesca Clemente
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Maria Giulia Davighi
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Camilla Matassini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy
| | - Francesca Cardona
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Associated with LENS, Via N. Carrara 1, 50019, Sesto F.no, FI, Italy
| | - Andrea Goti
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Associated with LENS, Via N. Carrara 1, 50019, Sesto F.no, FI, Italy
| | - Amelia Morrone
- Laboratory of Molecular Biology of Neurometabolic Diseases, Neuroscience Department, Meyer Children's Hospital, Viale Pieraccini 24, 50139, Firenze, Italy.,Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Viale Pieraccini 24, 50139, Firenze, Italy
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Firenze, Italy
| | - Tomás Tejero
- Institute of Chemical Synthesis and Homogeneous Catalysis. (ISQCH), University of Zaragoza, Campus San Francisco, Zaragoza, 50009, Spain
| | - Pedro Merino
- Institute of Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, Campus San Francisco, Zaragoza, 50009, Spain
| | - Martina Cacciarini
- Department of Chemistry "U. Schiff", University of Florence, Via della Lastruccia 3-13, 50019, Sesto F.no, FI, Italy.,Department of Chemistry, University of Copenhagen, Universitetsparken 5, DK-2100, Copenhagen Ø, Denmark
| |
Collapse
|
42
|
Krishnan K, Tian H, Tao P, Verkhivker GM. Probing conformational landscapes and mechanisms of allosteric communication in the functional states of the ABL kinase domain using multiscale simulations and network-based mutational profiling of allosteric residue potentials. J Chem Phys 2022; 157:245101. [PMID: 36586979 PMCID: PMC11184971 DOI: 10.1063/5.0133826] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
In the current study, multiscale simulation approaches and dynamic network methods are employed to examine the dynamic and energetic details of conformational landscapes and allosteric interactions in the ABL kinase domain that determine the kinase functions. Using a plethora of synergistic computational approaches, we elucidate how conformational transitions between the active and inactive ABL states can employ allosteric regulatory switches to modulate intramolecular communication networks between the ATP site, the substrate binding region, and the allosteric binding pocket. A perturbation-based network approach that implements mutational profiling of allosteric residue propensities and communications in the ABL states is proposed. Consistent with biophysical experiments, the results reveal functionally significant shifts of the allosteric interaction networks in which preferential communication paths between the ATP binding site and substrate regions in the active ABL state become suppressed in the closed inactive ABL form, which in turn features favorable allosteric coupling between the ATP site and the allosteric binding pocket. By integrating the results of atomistic simulations with dimensionality reduction methods and Markov state models, we analyze the mechanistic role of macrostates and characterize kinetic transitions between the ABL conformational states. Using network-based mutational scanning of allosteric residue propensities, this study provides a comprehensive computational analysis of long-range communications in the ABL kinase domain and identifies conserved regulatory hotspots that modulate kinase activity and allosteric crosstalk between the allosteric pocket, ATP binding site, and substrate binding regions.
Collapse
Affiliation(s)
| | - Hao Tian
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75205, USA
| | - Peng Tao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75205, USA
| | - Gennady M. Verkhivker
- Author to whom correspondence should be addressed: . Telephone: 714-516-4586. Fax: 714-532-6048
| |
Collapse
|
43
|
Koulgi S, Achalere A, Sonavane U, Joshi R. Markov State Modeling Analysis Captures Changes in the Temperature-Sensitive N-Terminal and β-Turn Regions of the p53 DNA-Binding Domain. J Chem Inf Model 2022; 62:6449-6461. [PMID: 35614540 DOI: 10.1021/acs.jcim.2c00380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The transcription factor p53 is one of the most widely studied cancer proteins. Its temperature-sensitive nature suggests reduction in functionality at physiological temperatures. Temperature-induced conformational variations and their impact on its functional ability still remain unexplored. A total of 20.8 μs molecular dynamics simulations of wildtype p53 in the apo and the DNA-bound states have been performed at 300 K and 310 K. Further, Markov State Modeling (MSM) analyses were performed, considering Cα-Cα distances as reaction coordinates. Filtering of these distances based on correlation with the time-independent components (tICs) resulted in 16 and 32 distances for apo and DNA-bound systems, respectively. Individual MSM analyses using these filtered distances were performed for both p53 systems. These Cα-Cα residue pairs belonged to the N-terminal, S6/7 β-turn, loop L2, loop L3, and hydrophobic core residues. At physiological temperatures, apo-p53 exhibits exposure of its hydrophobic core, where the temperature-sensitive hotspot residues were also located. This exposure was the result of the S6/7 β-turn and N-terminal moving apart. In the DNA-bound p53 system, loop L1 attains an open conformation at physiological temperatures, which weakens the DNA binding. It is already known that p53 mutants that lack DNA binding also tend to show similar conformational variations. The S6/7 β-turn along with the already known functionally important loop L2 may pose as regions to be targeted to overcome the loss in binding of temperature-sensitive wildtype p53. Rescue strategies directed toward these temperature-sensitive regions may be useful to recuperate its strong binding at physiological temperatures.
Collapse
Affiliation(s)
- Shruti Koulgi
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Archana Achalere
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Uddhavesh Sonavane
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| | - Rajendra Joshi
- High Performance Computing - Medical and Bioinformatics Applications Group, Centre for Development for Advanced Computing (C-DAC), Panchawati, Pashan, Pune 411 008, India
| |
Collapse
|
44
|
Cetin E, Atilgan AR, Atilgan C. DHFR Mutants Modulate Their Synchronized Dynamics with the Substrate by Shifting Hydrogen Bond Occupancies. J Chem Inf Model 2022; 62:6715-6726. [PMID: 35984987 PMCID: PMC9795552 DOI: 10.1021/acs.jcim.2c00507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Antibiotic resistance is a global health problem in which mutations occurring in functional proteins render drugs ineffective. The working mechanisms of the arising mutants are seldom apparent; a methodology to decipher these mechanisms systematically would render devising therapies to control the arising mutational pathways possible. Here we utilize Cα-Cβ bond vector relaxations obtained from moderate length MD trajectories to determine conduits for functionality of the resistance conferring mutants of Escherichia coli dihydrofolate reductase. We find that the whole enzyme is synchronized to the motions of the substrate, irrespective of the mutation introducing gain-of-function or loss-of function. The total coordination of the motions suggests changes in the hydrogen bond dynamics with respect to the wild type as a possible route to determine and classify the mode-of-action of individual mutants. As a result, nine trimethoprim-resistant point mutations arising frequently in evolution experiments are categorized. One group of mutants that display the largest occurrence (L28R, W30G) work directly by modifying the dihydrofolate binding region. Conversely, W30R works indirectly by the formation of the E139-R30 salt bridge which releases energy resulting from tight binding by distorting the binding cavity. A third group (D27E, F153S, I94L) arising as single, resistance invoking mutants in evolution experiment trajectories allosterically and dynamically affects a hydrogen bonding motif formed at residues 59-69-71 which in turn modifies the binding site dynamics. The final group (I5F, A26T, R98P) consists of those mutants that have properties most similar to the wild type; these only appear after one of the other mutants is fixed on the protein structure and therefore display clear epistasis. Thus, we show that the binding event is governed by the entire enzyme dynamics while the binding site residues play gating roles. The adjustments made in the total enzyme in response to point mutations are what make quantifying and pinpointing their effect a hard problem. Here, we show that hydrogen bond dynamics recorded on sub-μs time scales provide the necessary fingerprints to decipher the various mechanisms at play.
Collapse
|
45
|
Aliakbar Tehrani Z, Rulíšek L, Černý J. Molecular dynamics simulations provide structural insight into binding of cyclic dinucleotides to human STING protein. J Biomol Struct Dyn 2022; 40:10250-10264. [PMID: 34187319 DOI: 10.1080/07391102.2021.1942213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human stimulator of interferon genes (hSTING) is a signaling adaptor protein that triggers innate immune system by response to cytosolic DNA and second messenger cyclic dinucleotides (CDNs). Natural CDNs contain purine nucleobase with different phosphodiester linkage types (3'-3', 2'-2' or mixed 2'-3'-linkages) and exhibit different binding affinity towards hSTING, ranging from micromolar to nanomolar. High-affinity CDNs are considered as suitable candidates for treatment of chronic hepatitis B and cancer. We have used molecular dynamics simulations to investigate dynamical aspects of binding of natural CDNs (specifically, 2'-2'-cGAMP, 2'-3'-cGAMP, 3'-3'-cGAMP, 3'-3'-c-di-AMP, and 3'-3'-c-di-GMP) with hSTINGwt protein. Our results revealed that CDN/hSTINGwt interactions are controlled by the balance between fluctuations (conformational changes) in the CDN ligand and the protein dynamics. Binding of different CDNs induces different degrees of conformational/dynamics changes in hSTINGwt ligand binding cavity, especially in α1-helices, the so-called lid region and α2-tails. The ligand residence time in hSTINGwt protein pocket depends on different contribution of R232 and R238 residues interacting with oxygen atoms of phosphodiester groups in ligand, water distribution around interacting charged centers (in protein residues and ligand) and structural stability of closed conformation state of hSTINGwt protein. These findings may perhaps guide design of new compounds modulating hSTING activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zahra Aliakbar Tehrani
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Lubomír Rulíšek
- Theoretical Bioinorganic Chemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiří Černý
- Laboratory of Structural Bioinformatics of Proteins, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
46
|
Gorin G, Vastola JJ, Fang M, Pachter L. Interpretable and tractable models of transcriptional noise for the rational design of single-molecule quantification experiments. Nat Commun 2022; 13:7620. [PMID: 36494337 PMCID: PMC9734650 DOI: 10.1038/s41467-022-34857-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 11/09/2022] [Indexed: 12/13/2022] Open
Abstract
The question of how cell-to-cell differences in transcription rate affect RNA count distributions is fundamental for understanding biological processes underlying transcription. Answering this question requires quantitative models that are both interpretable (describing concrete biophysical phenomena) and tractable (amenable to mathematical analysis). This enables the identification of experiments which best discriminate between competing hypotheses. As a proof of principle, we introduce a simple but flexible class of models involving a continuous stochastic transcription rate driving a discrete RNA transcription and splicing process, and compare and contrast two biologically plausible hypotheses about transcription rate variation. One assumes variation is due to DNA experiencing mechanical strain, while the other assumes it is due to regulator number fluctuations. We introduce a framework for numerically and analytically studying such models, and apply Bayesian model selection to identify candidate genes that show signatures of each model in single-cell transcriptomic data from mouse glutamatergic neurons.
Collapse
Affiliation(s)
- Gennady Gorin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - John J Vastola
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Meichen Fang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
47
|
Zhao T, Karki N, Zoltowski BD, Matthews DA. Allosteric regulation in STAT3 interdomains is mediated by a rigid core: SH2 domain regulation by CCD in D170A variant. PLoS Comput Biol 2022; 18:e1010794. [PMID: 36542668 PMCID: PMC9815575 DOI: 10.1371/journal.pcbi.1010794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/05/2023] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Signal Transducer and Activator of Transcription 3 (STAT3) plays a crucial role in cancer development and thus is a viable target for cancer treatment. STAT3 functions as a dimer mediated by phosphorylation of the SRC-homology 2 (SH2) domain, a key target for therapeutic drugs. While great efforts have been employed towards the development of compounds that directly target the SH2 domain, no compound has yet been approved by the FDA due to a lack of specificity and pharmacologic efficacy. Studies have shown that allosteric regulation of SH2 via the coiled-coil domain (CCD) is an alternative drug design strategy. Several CCD effectors have been shown to modulate SH2 binding and affinity, and at the time of writing at least one drug candidate has entered phase I clinical trials. However, the mechanism for SH2 regulation via CCD is poorly understood. Here, we investigate structural and dynamic features of STAT3 and compare the wild type to the reduced function variant D170A in order to delineate mechanistic differences and propose allosteric pathways. Molecular dynamics simulations were employed to explore conformational space of STAT3 and the variant, followed by structural, conformation, and dynamic analysis. The trajectories explored show distinctive conformational changes in the SH2 domain for the D170A variant, indicating long range allosteric effects. Multiple analyses provide evidence for long range communication pathways between the two STAT3 domains, which seem to be mediated by a rigid core which connects the CCD and SH2 domains via the linker domain (LD) and transmits conformational changes through a network of short-range interactions. The proposed allosteric mechanism provides new insight into the understanding of intramolecular signaling in STAT3 and potential pharmaceutical control of STAT3 specificity and activity.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Chemistry, Southern Methodist University, Dallas, Texas, United States of America
| | - Nischal Karki
- Department of Chemistry, Southern Methodist University, Dallas, Texas, United States of America
| | - Brian D. Zoltowski
- Department of Chemistry, Southern Methodist University, Dallas, Texas, United States of America
| | - Devin A. Matthews
- Department of Chemistry, Southern Methodist University, Dallas, Texas, United States of America
| |
Collapse
|
48
|
Kochnev Y, Durrant JD. FPocketWeb: protein pocket hunting in a web browser. J Cheminform 2022; 14:58. [PMID: 36008829 PMCID: PMC9414105 DOI: 10.1186/s13321-022-00637-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/04/2022] [Indexed: 11/10/2022] Open
Abstract
Detecting macromolecular (e.g., protein) cavities where small molecules bind is an early step in computer-aided drug discovery. Multiple pocket-detection algorithms have been developed over the past several decades. Among them, fpocket, created by Schmidtke and Le Guilloux, is particularly popular. Like many programs used in computational-biology research, fpocket requires users to download and install an executable file. That file must also be run via a command-line interface, further complicating use. An existing fpocket server application effectively addresses these challenges, but it requires users to upload their possibly proprietary structures to a third-party server. The FPocketWeb web app builds on this prior work. It runs the fpocket3 executable entirely in a web browser without requiring installation. The pocket-finding calculations occur on the user's computer rather than on a remote server. A working version of the open-source FPocketWeb app can be accessed free of charge from http://durrantlab.com/fpocketweb .
Collapse
Affiliation(s)
- Yuri Kochnev
- Department of Biological Sciences, University of Pittsburgh, 15260, Pittsburgh, PA, USA
| | - Jacob D Durrant
- Department of Biological Sciences, University of Pittsburgh, 15260, Pittsburgh, PA, USA.
| |
Collapse
|
49
|
Wakefield AE, Kozakov D, Vajda S. Mapping the binding sites of challenging drug targets. Curr Opin Struct Biol 2022; 75:102396. [PMID: 35636004 PMCID: PMC9790766 DOI: 10.1016/j.sbi.2022.102396] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 02/03/2023]
Abstract
An increasing number of medically important proteins are challenging drug targets because their binding sites are too shallow or too polar, are cryptic and thus not detectable without a bound ligand or located in a protein-protein interface. While such proteins may not bind druglike small molecules with sufficiently high affinity, they are frequently druggable using novel therapeutic modalities. The need for such modalities can be determined by experimental or computational fragment based methods. Computational mapping by mixed solvent molecular dynamics simulations or the FTMap server can be used to determine binding hot spots. The strength and location of the hot spots provide very useful information for selecting potentially successful approaches to drug discovery.
Collapse
Affiliation(s)
- Amanda E. Wakefield
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215,Department of Chemistry, Boston University, Boston, Massachusetts 02215
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, USA,Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, USA NY, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215,Department of Chemistry, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
50
|
Casadó-Anguera V, Casadó V. Unmasking allosteric binding sites: Novel targets for GPCR drug discovery. Expert Opin Drug Discov 2022; 17:897-923. [PMID: 35649692 DOI: 10.1080/17460441.2022.2085684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Unexpected non-apparent and hidden allosteric binding sites are non-classical and non-apparent allosteric centers in 3-D X-ray protein structures until orthosteric or allosteric ligands bind to them. The orthosteric center of one protomer that modulates binding centers of the other protomers within an oligomer is also an unexpected allosteric site. Furthermore, another partner protein can also produce these effects, acting as an unexpected allosteric modulator. AREAS COVERED This review summarizes both classical and non-classical allosterism. The authors focus on G protein-coupled receptor (GPCR) oligomers as a paradigm of allosteric molecules. Moreover, they show several examples of unexpected allosteric sites such as hidden allosteric sites in a protomer that appear after the interaction with other molecules and the allosterism exerted between orthosteric sites within GPCR oligomer, emphasizing on the allosteric modulations that can occur between binding sites. EXPERT OPINION The study of these new non-classical allosteric sites will expand the diversity of allosteric control on the function of orthosteric sites within proteins, whether GPCRs or other receptors, enzymes or transporters. Moreover, the design of new drugs targeting these hidden allosteric sites or already known orthosteric sites acting as allosteric sites in protein homo- or hetero-oligomers will increase the therapeutic potential of allosterism.
Collapse
Affiliation(s)
- Verònica Casadó-Anguera
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain.,Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vicent Casadó
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|