1
|
Wankowicz SA, Fraser JS. Advances in uncovering the mechanisms of macromolecular conformational entropy. Nat Chem Biol 2025; 21:623-634. [PMID: 40275100 PMCID: PMC12103944 DOI: 10.1038/s41589-025-01879-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/10/2025] [Indexed: 04/26/2025]
Abstract
During protein folding, proteins transition from a disordered polymer into a globular structure, markedly decreasing their conformational degrees of freedom, leading to a substantial reduction in entropy. Nonetheless, folded proteins retain substantial entropy as they fluctuate between the conformations that make up their native state. This residual entropy contributes to crucial functions like binding and catalysis, supported by growing evidence primarily from NMR and simulation studies. Here, we propose three major ways that macromolecules use conformational entropy to perform their functions; first, prepaying entropic cost through ordering of the ground state; second, spatially redistributing entropy, in which a decrease in entropy in one area is reciprocated by an increase in entropy elsewhere; third, populating catalytically competent ensembles, in which conformational entropy within the enzymatic scaffold aids in lowering transition state barriers. We also provide our perspective on how solving the current challenge of structurally defining the ensembles encoding conformational entropy will lead to new possibilities for controlling binding, catalysis and allostery.
Collapse
Affiliation(s)
- Stephanie A Wankowicz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Ekambaram S, Arakelov G, Dokholyan NV. The Evolving Landscape of Protein Allostery: From Computational and Experimental Perspectives. J Mol Biol 2025:169060. [PMID: 40043838 DOI: 10.1016/j.jmb.2025.169060] [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: 01/31/2025] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Protein allostery is a fundamental biological regulatory mechanism that allows communication between distant locations within a protein, modifying its function in response to signals. Experimental techniques, such as NMR spectroscopy and cryo-electron microscopy (cryo-EM), are critical validation tools for computational predictions and provide valuable insights into dynamic conformational changes. Combining these approaches has greatly improved our understanding of classical conformational allostery and complex dynamic coupling mechanisms. Recent advances in machine learning and enhanced sampling methods have broadened the scope of allostery research, identifying cryptic allosteric sites and directing new drug discovery approaches. Despite progress, bridging static structural data with dynamic functional states remains challenging. This review underscores the importance of combining experimental and computational approaches to comprehensively understand protein allostery and its diverse applications in biology and medicine.
Collapse
Affiliation(s)
- Srinivasan Ekambaram
- Department of Neuroscience and Experimental Therapeutics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Grigor Arakelov
- Department of Neuroscience and Experimental Therapeutics, Penn State College of Medicine, Hershey, PA 17033, USA; Institute of Molecular Biology of the National Academy of Sciences of the Republic of Armenia, Yerevan 0014, Armenia
| | - Nikolay V Dokholyan
- Department of Neuroscience and Experimental Therapeutics, Penn State College of Medicine, Hershey, PA 17033, USA; Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA; Department of Chemistry, Penn State University, University Park, PA 16802, USA; Department of Biomedical Engineering, Penn State University, University Park, PA 16802, USA.
| |
Collapse
|
3
|
Yan F, Wang Y, Chen L, Cheng W, Oduro‐Kwateng E, Soliman MES, Yang T. Nanohydrogel of Curcumin/Berberine Co-Crystals Induces Apoptosis via Dual Covalent/Noncovalent Inhibition of Caspases in Endometrial Cancer Cell Lines: The Synergy Between Pharmacokinetics and Pharmacodynamics. J Mol Recognit 2025; 38:e70004. [PMID: 40098247 PMCID: PMC11914374 DOI: 10.1002/jmr.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/01/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Endometrial cancer remains a significant therapeutic challenge due to drug resistance and heterogeneity. This study leverages the synergistic potential of curcumin (CUR) and berberine (BBR) co-crystals encapsulated in a nanohydrogel to address these challenges through a pharmacokinetically and pharmacodynamically targeted therapeutic strategy. The nanohydrogel formulation significantly improves the solubility, stability, and bioavailability of CUR/BBR co-crystals, optimizing their therapeutic delivery and sustained release under physiological and tumor microenvironment conditions. On the other hand, the dual inhibitory mechanism of CUR and BBR, with CUR covalently binding to the active site of caspase-3 and BBR non-covalently targeting the allosteric site, achieves enhanced apoptotic activity by disrupting both the catalytic and conformational functions of caspase-3. In vitro cytotoxicity assays demonstrate remarkable efficacy of the CUR/BBR nanohydrogel, achieving an IC50 of 12.36 μg/mL against HEC-59 endometrial cancer cells, significantly outperforming the individual components and the standard drug Camptothecin (IC50: 17.27 μg/mL). Caspase-3/7 assays confirm enhanced apoptosis induction for the nanohydrogel formulation compared to co-crystals alone and Camptothecin. Molecular dynamics simulations and binding free energy analyses further validate the synergistic interaction of CUR and BBR in their dual binding mode. This study introduces a novel therapeutic approach by enhancing drug delivery and dual targeting mechanisms, demonstrating the potential of CUR-BBR nanohydrogel as a robust therapy for EC. This strategy offers a promising platform for addressing drug resistance and improving outcomes in endometrial cancer therapy.
Collapse
Affiliation(s)
- Feiyan Yan
- Department of Gynecology, The Second Affiliated Hospital of Xi'an Medical UniversityXi’anChina
| | - Yan Wang
- Department of Gynecology, The Second Affiliated Hospital of Xi'an Medical UniversityXi’anChina
| | - Lin Chen
- Department of Gynecology, The Second Affiliated Hospital of Xi'an Medical UniversityXi’anChina
| | - Wei Cheng
- Department of Gynecology, The Second Affiliated Hospital of Xi'an Medical UniversityXi’anChina
| | - Ernest Oduro‐Kwateng
- Molecular Bio‐Computation and Drug Design Research Group, School of Health SciencesUniversity of KwaZulu Natal, Westville CampusDurbanSouth Africa
| | - Mahmoud E. S. Soliman
- Molecular Bio‐Computation and Drug Design Research Group, School of Health SciencesUniversity of KwaZulu Natal, Westville CampusDurbanSouth Africa
| | - Ting Yang
- Department of Gynecology, The Second Affiliated Hospital of Xi'an Medical UniversityXi’anChina
| |
Collapse
|
4
|
Cruz-Rodriguez N, Deininger MW. Novel treatment strategies for chronic myeloid leukemia. Blood 2025; 145:931-943. [PMID: 39729529 PMCID: PMC11952011 DOI: 10.1182/blood.2024026312] [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: 10/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024] Open
Abstract
ABSTRACT Starting with imatinib, tyrosine kinase inhibitors (TKIs) have turned chronic myeloid leukemia (CML) from a lethal blood cancer into a chronic condition. As patients with access to advanced CML care have an almost normal life expectancy, there is a perception that CML is a problem of the past, and one should direct research resources elsewhere. However, a closer look at the current CML landscape reveals a more nuanced picture. Most patients still require life-long TKI therapy to avoid recurrence of active CML. Chronic TKI toxicity and the high costs of the well-tolerated agents remain challenging. Progression to blast phase still occurs, particularly in socioeconomically disadvantaged parts of the world, where high-risk CML at diagnosis is common. Here, we review the prospects of further improving TKIs to achieve optimal suppression of BCR::ABL1 kinase activity, the potential of combining different classes of TKIs, and the current state of BCR::ABL1 degraders. We cover combination therapy approaches to address TKI resistance in the setting of residual leukemia and in advanced CML. Despite the unprecedented success of TKIs in CML, more work is needed to truly finish the job, and we hope to stimulate innovative research aiming to achieve this goal.
Collapse
Affiliation(s)
| | - Michael W. Deininger
- Versiti Blood Research Institute, Milwaukee, WI
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
5
|
Wang S, Wang M, Li Z, Xu G, Wang D. Discovery of N-(2-Acetamidobenzo[ d]thiazol-6-yl)-2-phenoxyacetamide Derivatives as Novel Potential BCR-ABL1 Inhibitors Through Structure-Based Virtual Screening. Molecules 2025; 30:1065. [PMID: 40076290 PMCID: PMC11901765 DOI: 10.3390/molecules30051065] [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: 01/20/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BCR-ABL1 kinase is a critical driver of chronic myeloid leukemia (CML) pathophysiology. The approval of allosteric inhibitor asciminib brings new hope for overcoming drug resistance caused by mutations in the ATP-binding site. To expand the chemical diversity of BCR-ABL1 kinase inhibitors with positive anti-tumor effect with asciminib, structure-based virtual screening and molecular dynamics simulations were employed to discover novel scaffolds. This approach led to the identification of a series of N-(2-acetamidobenzo[d]thiazol-6-yl)-2-phenoxyacetamide derivatives as new BCR-ABL1 inhibitors. The most potent compound, 10m, demonstrated inhibition of BCR-ABL-dependent signaling and showed an anti-tumor effect against K562 cells, with an IC50 value of 0.98 μM. Compound 10m displayed powerful synergistic anti-proliferation and pro-apoptotic effects when combined with asciminib, highlighting its potential as a promising lead for the development of potential BCR-ABL inhibitors.
Collapse
Affiliation(s)
- Shuaixing Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Reference and Research on Influenza, Key Laboratory for Medical Virology and Viral Diseases, National Health Commission, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing 102206, China; (S.W.); (M.W.); (Z.L.)
| | - Minyi Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Reference and Research on Influenza, Key Laboratory for Medical Virology and Viral Diseases, National Health Commission, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing 102206, China; (S.W.); (M.W.); (Z.L.)
| | - Zi Li
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Reference and Research on Influenza, Key Laboratory for Medical Virology and Viral Diseases, National Health Commission, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing 102206, China; (S.W.); (M.W.); (Z.L.)
| | - Guofeng Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100083, China;
| | - Dayan Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Reference and Research on Influenza, Key Laboratory for Medical Virology and Viral Diseases, National Health Commission, National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Disease, Beijing 102206, China; (S.W.); (M.W.); (Z.L.)
| |
Collapse
|
6
|
Selzer AM, Gerlach G, Gonzalez-Areizaga G, Wales TE, Cui SY, Iyer P, Engen JR, Camacho C, Ishima R, Smithgall TE. An SH3-binding allosteric modulator stabilizes the global conformation of the AML-associated Src-family kinase, Hck. J Biol Chem 2025; 301:108088. [PMID: 39675702 PMCID: PMC11786751 DOI: 10.1016/j.jbc.2024.108088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
While ATP-site inhibitors for protein-tyrosine kinases are often effective drugs, their clinical utility can be limited by off-target activity and acquired resistance mutations due to the conserved nature of the ATP-binding site. However, combining ATP-site and allosteric kinase inhibitors can overcome these shortcomings in a double-drugging framework. Here we explored the allosteric effects of two pyrimidine diamines, PDA1 and PDA2, on the conformational dynamics and activity of the Src-family tyrosine kinase Hck, a promising drug target for acute myeloid leukemia. Using 1H-15N HSQC NMR, we mapped the binding site for both analogs to the SH3 domain. Despite the shared binding site, PDA1 and PDA2 had opposing effects on near-full-length Hck dynamics by hydrogen-deuterium exchange mass spectrometry, with PDA1 stabilizing and PDA2 disrupting the overall kinase conformation. Kinase activity assays were consistent with these observations, with PDA2 enhancing kinase activity while PDA1 was without effect. Molecular dynamics simulations predicted selective bridging of the kinase domain N-lobe and SH3 domain by PDA1, a mechanism of allosteric stabilization supported by site-directed mutagenesis of N-lobe contact sites. Cellular thermal shift assays confirmed SH3 domain-dependent interaction of PDA1 with WT Hck in myeloid leukemia cells and with a kinase domain gatekeeper mutant (T338M). These results identify PDA1 as a starting point for Src-family kinase allosteric inhibitor development that may work in concert with ATP-site inhibitors to suppress the evolution of resistance.
Collapse
Affiliation(s)
- Ari M Selzer
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gabriella Gerlach
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Giancarlo Gonzalez-Areizaga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas E Wales
- Department of Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Stephanie Y Cui
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Prema Iyer
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - John R Engen
- Department of Chemistry and Chemical Biology, College of Science, Northeastern University, Boston, Massachusetts, USA
| | - Carlos Camacho
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas E Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
7
|
Vaisar D, Ahn NG. Latent allosteric control of protein interactions by ATP-competitive kinase inhibitors. Curr Opin Struct Biol 2024; 89:102935. [PMID: 39395271 PMCID: PMC11884338 DOI: 10.1016/j.sbi.2024.102935] [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: 07/02/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/14/2024]
Abstract
Protein kinase inhibitors designed to compete with ATP as a primary mode of action turn out to have considerable effects that go beyond their interference of nucleotide binding. New research shows how kinase activation and sometimes noncatalytic functions of protein kinases can be controlled by allosteric properties of kinase inhibitors, communicating perturbations from the active site to distal regulatory regions.
Collapse
Affiliation(s)
- David Vaisar
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303, USA
| | - Natalie G Ahn
- Department of Biochemistry, University of Colorado Boulder, Boulder CO 80303, USA.
| |
Collapse
|
8
|
Selzer AM, Alvarado JJ, Smithgall TE. Cocrystallization of the Src-Family Kinase Hck with the ATP-Site Inhibitor A-419259 Stabilizes an Extended Activation Loop Conformation. Biochemistry 2024; 63:2594-2601. [PMID: 39315638 PMCID: PMC11483750 DOI: 10.1021/acs.biochem.4c00323] [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: 06/10/2024] [Revised: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
Hematopoietic cell kinase (Hck) is a member of the Src kinase family and is a promising drug target in myeloid leukemias. Here, we report the crystal structure of human Hck in complex with the pyrrolopyrimidine inhibitor A-419259, determined at a resolution of 1.8 Å. This structure reveals the complete Hck active site in the presence of A-419259, including the αC-helix, the DFG motif, and the activation loop. A-419259 binds at the ATP-site of Hck and induces an overall closed conformation of the kinase with the regulatory SH3 and SH2 domains bound intramolecularly to their respective internal ligands. A-419259 stabilizes the DFG-in/αC-helix-out conformation observed previously with Hck and the pyrazolopyrimidine inhibitor PP1 (PDB: 1QCF). However, the activation loop conformations are distinct, with PP1 inducing a folded loop structure with the tyrosine autophosphorylation site (Tyr416) pointing into the ATP binding site, while A-419259 stabilizes an extended loop conformation with Tyr416 facing out into the solvent. Autophosphorylation also induces activation loop extension and significantly reduces the Hck sensitivity to PP1 but not A-419259. In cancer cells where Hck is constitutively active, the extended autophosphorylation loop may render Hck more sensitive to inhibitors like A-419259 which prefer this kinase conformation. More generally, these results provide additional insight into targeted kinase inhibitor design and how conformational preferences of inhibitors may impact selectivity and potency.
Collapse
Affiliation(s)
- Ari M. Selzer
- Department of Microbiology
and Molecular Genetics, University of Pittsburgh
School of Medicine, 450 Technology Drive, Pittsburgh, Pennsylvania PA 15219, United States
| | - John J. Alvarado
- Department of Microbiology
and Molecular Genetics, University of Pittsburgh
School of Medicine, 450 Technology Drive, Pittsburgh, Pennsylvania PA 15219, United States
| | - Thomas E. Smithgall
- Department of Microbiology
and Molecular Genetics, University of Pittsburgh
School of Medicine, 450 Technology Drive, Pittsburgh, Pennsylvania PA 15219, United States
| |
Collapse
|
9
|
Raisinghani N, Alshahrani M, Gupta G, Verkhivker G. Predicting Mutation-Induced Allosteric Changes in Structures and Conformational Ensembles of the ABL Kinase Using AlphaFold2 Adaptations with Alanine Sequence Scanning. Int J Mol Sci 2024; 25:10082. [PMID: 39337567 PMCID: PMC11432724 DOI: 10.3390/ijms251810082] [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: 07/14/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Despite the success of AlphaFold2 approaches in predicting single protein structures, these methods showed intrinsic limitations in predicting multiple functional conformations of allosteric proteins and have been challenged to accurately capture the effects of single point mutations that induced significant structural changes. We examined several implementations of AlphaFold2 methods to predict conformational ensembles for state-switching mutants of the ABL kinase. The results revealed that a combination of randomized alanine sequence masking with shallow multiple sequence alignment subsampling can significantly expand the conformational diversity of the predicted structural ensembles and capture shifts in populations of the active and inactive ABL states. Consistent with the NMR experiments, the predicted conformational ensembles for M309L/L320I and M309L/H415P ABL mutants that perturb the regulatory spine networks featured the increased population of the fully closed inactive state. The proposed adaptation of AlphaFold can reproduce the experimentally observed mutation-induced redistributions in the relative populations of the active and inactive ABL states and capture the effects of regulatory mutations on allosteric structural rearrangements of the kinase domain. The ensemble-based network analysis complemented AlphaFold predictions by revealing allosteric hotspots that correspond to state-switching mutational sites which may explain the global effect of regulatory mutations on structural changes between the ABL states. This study suggested that attention-based learning of long-range dependencies between sequence positions in homologous folds and deciphering patterns of allosteric interactions may further augment the predictive abilities of AlphaFold methods for modeling of alternative protein sates, conformational ensembles and mutation-induced structural transformations.
Collapse
Affiliation(s)
- Nishank Raisinghani
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Mohammed Alshahrani
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Grace Gupta
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
| | - Gennady Verkhivker
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, Orange, CA 92866, USA
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| |
Collapse
|
10
|
Leyte-Vidal A, DeFilippis R, Outhwaite IR, Kwan I, Lee JY, Leavitt C, Miller KB, Rea D, Rangwala AM, Lou K, Patel S, Alvarez A, Shokat KM, Bahar I, Seeliger MA, Shah NP. Absence of ABL1 exon 2-encoded SH3 residues in BCR::ABL1 destabilizes the autoinhibited kinase conformation and confers resistance to asciminib. Leukemia 2024; 38:2046-2050. [PMID: 39085402 PMCID: PMC11347358 DOI: 10.1038/s41375-024-02353-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
MESH Headings
- Humans
- Fusion Proteins, bcr-abl/genetics
- Drug Resistance, Neoplasm/genetics
- Exons/genetics
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- src Homology Domains
- Proto-Oncogene Proteins c-abl/genetics
- Protein Conformation
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Niacinamide/analogs & derivatives
Collapse
Affiliation(s)
- Ariel Leyte-Vidal
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33101, USA
| | - RosaAnna DeFilippis
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
| | - Ian R Outhwaite
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Isabelle Kwan
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Ji Young Lee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Carlyn Leavitt
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
| | - Kaeli B Miller
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
| | - Delphine Rea
- Adult Hematology Department, Hôpital Saint-Louis, Paris, France
| | - Aziz M Rangwala
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Kevin Lou
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Howard Hughes Medical Institute, University of California, San Francisco, CA, 94158, USA
| | - Suhana Patel
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
| | - Ailin Alvarez
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA
| | - Kevan M Shokat
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Howard Hughes Medical Institute, University of California, San Francisco, CA, 94158, USA
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Markus A Seeliger
- Department of Pharmacological Sciences, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Neil P Shah
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
11
|
Leyte-Vidal A, Garrido Ruiz D, DeFilippis R, Leske IB, Rea D, Phan S, Miller KB, Hu F, Mase A, Shan Y, Hantschel O, Jacobson MP, Shah NP. BCR::ABL1 kinase N-lobe mutants confer moderate to high degrees of resistance to asciminib. Blood 2024; 144:639-645. [PMID: 38643492 DOI: 10.1182/blood.2023022538] [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: 09/14/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/23/2024] Open
Abstract
ABSTRACT Secondary kinase domain mutations in BCR::ABL1 represent the most common cause of resistance to tyrosine kinase inhibitor (TKI) therapy in patients with chronic myeloid leukemia. The first 5 approved BCR::ABL1 TKIs target the adenosine triphosphate (ATP)-binding pocket. Mutations confer resistance to these ATP-competitive TKIs and those approved for other malignancies by decreasing TKI affinity and/or increasing ATP affinity. Asciminib, the first highly active allosteric TKI approved for any malignancy, targets an allosteric regulatory pocket in the BCR::ABL1 kinase C-lobe. As a non-ATP-competitive inhibitor, the activity of asciminib is predicted to be impervious to increases in ATP affinity. Here, we report several known mutations that confer resistance to ATP-competitive TKIs in the BCR::ABL1 kinase N-lobe that are distant from the asciminib binding pocket yet unexpectedly confer in vitro resistance to asciminib. Among these is BCR::ABL1 M244V, which confers clinical resistance even to escalated asciminib doses. We demonstrate that BCR::ABL1 M244V does not impair asciminib binding, thereby invoking a novel mechanism of resistance. Molecular dynamic simulations of the M244V substitution implicate stabilization of an active kinase conformation through impact on the α-C helix as a mechanism of resistance. These N-lobe mutations may compromise the clinical activity of ongoing combination studies of asciminib with ATP-competitive TKIs.
Collapse
Affiliation(s)
- Ariel Leyte-Vidal
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL
| | - Diego Garrido Ruiz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA
| | - RosaAnna DeFilippis
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Inga B Leske
- Institute for Physiological Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Delphine Rea
- Adult Hematology Department, Hôpital Saint-Louis, Paris, France
| | - Stacey Phan
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Kaeli B Miller
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Feifei Hu
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Anjeli Mase
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| | - Yibing Shan
- Antidote Health Foundation for Cure of Cancer, Cambridge, MA
| | - Oliver Hantschel
- Institute for Physiological Chemistry, Philipps University of Marburg, Marburg, Germany
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA
| | - Neil P Shah
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA
| |
Collapse
|
12
|
Raisinghani N, Alshahrani M, Gupta G, Tian H, Xiao S, Tao P, Verkhivker GM. Integration of a Randomized Sequence Scanning Approach in AlphaFold2 and Local Frustration Profiling of Conformational States Enable Interpretable Atomistic Characterization of Conformational Ensembles and Detection of Hidden Allosteric States in the ABL1 Protein Kinase. J Chem Theory Comput 2024; 20:5317-5336. [PMID: 38865109 PMCID: PMC12100677 DOI: 10.1021/acs.jctc.4c00222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Despite the success of AlphaFold methods in predicting single protein structures, these methods showed intrinsic limitations in the characterization of multiple functional conformations of allosteric proteins. The recent NMR-based structural determination of the unbound ABL kinase in the active state and discovery of the inactive low-populated functional conformations that are unique for ABL kinase present an ideal challenge for the AlphaFold2 approaches. In the current study, we employ several adaptations of the AlphaFold2 methodology to predict protein conformational ensembles and allosteric states of the ABL kinase including randomized alanine sequence scanning combined with the multiple sequence alignment subsampling proposed in this study. We show that the proposed new AlphaFold2 adaptation combined with local frustration profiling of conformational states enables accurate prediction of the protein kinase structures and conformational ensembles, also offering a robust approach for interpretable characterization of the AlphaFold2 predictions and detection of hidden allosteric states. We found that the large high frustration residue clusters are uniquely characteristic of the low-populated, fully inactive ABL form and can define energetically frustrated cracking sites of conformational transitions, presenting difficult targets for AlphaFold2. The results of this study uncovered previously unappreciated fundamental connections between local frustration profiles of the functional allosteric states and the ability of AlphaFold2 methods to predict protein structural ensembles of the active and inactive states. This study showed that integration of the randomized sequence scanning adaptation of AlphaFold2 with a robust landscape-based analysis allows for interpretable atomistic predictions and characterization of protein conformational ensembles, providing a physical basis for the successes and limitations of current AlphaFold2 methods in detecting functional allosteric states that play a significant role in protein kinase regulation.
Collapse
Affiliation(s)
- Nishank Raisinghani
- 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
| | - Grace Gupta
- 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
| | - Hao Tian
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States
| | - 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
| | - 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 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
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
13
|
Raisinghani N, Alshahrani M, Gupta G, Tian H, Xiao S, Tao P, Verkhivker G. Prediction of Conformational Ensembles and Structural Effects of State-Switching Allosteric Mutants in the Protein Kinases Using Comparative Analysis of AlphaFold2 Adaptations with Sequence Masking and Shallow Subsampling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594786. [PMID: 38798650 PMCID: PMC11118581 DOI: 10.1101/2024.05.17.594786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Despite the success of AlphaFold2 approaches in predicting single protein structures, these methods showed intrinsic limitations in predicting multiple functional conformations of allosteric proteins and have been challenged to accurately capture of the effects of single point mutations that induced significant structural changes. We systematically examined several implementations of AlphaFold2 methods to predict conformational ensembles for state-switching mutants of the ABL kinase. The results revealed that a combination of randomized alanine sequence masking with shallow multiple sequence alignment subsampling can significantly expand the conformational diversity of the predicted structural ensembles and capture shifts in populations of the active and inactive ABL states. Consistent with the NMR experiments, the predicted conformational ensembles for M309L/L320I and M309L/H415P ABL mutants that perturb the regulatory spine networks featured the increased population of the fully closed inactive state. On the other hand, the predicted conformational ensembles for the G269E/M309L/T334I and M309L/L320I/T334I triple ABL mutants that share activating T334I gate-keeper substitution are dominated by the active ABL form. The proposed adaptation of AlphaFold can reproduce the experimentally observed mutation-induced redistributions in the relative populations of the active and inactive ABL states and capture the effects of regulatory mutations on allosteric structural rearrangements of the kinase domain. The ensemble-based network analysis complemented AlphaFold predictions by revealing allosteric mediating centers that often directly correspond to state-switching mutational sites or reside in their immediate local structural proximity, which may explain the global effect of regulatory mutations on structural changes between the ABL states. This study suggested that attention-based learning of long-range dependencies between sequence positions in homologous folds and deciphering patterns of allosteric interactions may further augment the predictive abilities of AlphaFold methods for modeling of alternative protein sates, conformational ensembles and mutation-induced structural transformations.
Collapse
|
14
|
Paladini J, Maier A, Habazettl JM, Hertel I, Sonti R, Grzesiek S. The molecular basis of Abelson kinase regulation by its αI-helix. eLife 2024; 12:RP92324. [PMID: 38588001 PMCID: PMC11001296 DOI: 10.7554/elife.92324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Abelson tyrosine kinase (Abl) is regulated by the arrangement of its regulatory core, consisting sequentially of the SH3, SH2, and kinase (KD) domains, where an assembled or disassembled core corresponds to low or high kinase activity, respectively. It was recently established that binding of type II ATP site inhibitors, such as imatinib, generates a force from the KD N-lobe onto the SH3 domain and in consequence disassembles the core. Here, we demonstrate that the C-terminal αI-helix exerts an additional force toward the SH2 domain, which correlates both with kinase activity and type II inhibitor-induced disassembly. The αI-helix mutation E528K, which is responsible for the ABL1 malformation syndrome, strongly activates Abl by breaking a salt bridge with the KD C-lobe and thereby increasing the force onto the SH2 domain. In contrast, the allosteric inhibitor asciminib strongly reduces Abl's activity by fixating the αI-helix and reducing the force onto the SH2 domain. These observations are explained by a simple mechanical model of Abl activation involving forces from the KD N-lobe and the αI-helix onto the KD/SH2SH3 interface.
Collapse
Affiliation(s)
- Johannes Paladini
- Structural Biology and Biophysics, Biozentrum, University of BaselBaselSwitzerland
| | - Annalena Maier
- Structural Biology and Biophysics, Biozentrum, University of BaselBaselSwitzerland
| | | | - Ines Hertel
- Structural Biology and Biophysics, Biozentrum, University of BaselBaselSwitzerland
| | - Rajesh Sonti
- Structural Biology and Biophysics, Biozentrum, University of BaselBaselSwitzerland
| | - Stephan Grzesiek
- Structural Biology and Biophysics, Biozentrum, University of BaselBaselSwitzerland
| |
Collapse
|
15
|
Stachowski TR, Fischer M. FLEXR GUI: a graphical user interface for multi-conformer modeling of proteins. J Appl Crystallogr 2024; 57:580-586. [PMID: 38596743 PMCID: PMC11001397 DOI: 10.1107/s1600576724001523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 04/11/2024] Open
Abstract
Proteins are well known 'shapeshifters' which change conformation to function. In crystallography, multiple conformational states are often present within the crystal and the resulting electron-density map. Yet, explicitly incorporating alternative states into models to disentangle multi-conformer ensembles is challenging. We previously reported the tool FLEXR, which, within a few minutes, automatically separates conformational signal from noise and builds the corresponding, often missing, structural features into a multi-conformer model. To make the method widely accessible for routine multi-conformer building as part of the computational toolkit for macromolecular crystallography, we present a graphical user interface (GUI) for FLEXR, designed as a plugin for Coot 1. The GUI implementation seamlessly connects FLEXR models with the existing suite of validation and modeling tools available in Coot. We envision that FLEXR will aid crystallographers by increasing access to a multi-conformer modeling method that will ultimately lead to a better representation of protein conformational heterogeneity in the Protein Data Bank. In turn, deeper insights into the protein conformational landscape may inform biology or provide new opportunities for ligand design. The code is open source and freely available on GitHub at https://github.com/TheFischerLab/FLEXR-GUI.
Collapse
Affiliation(s)
- Timothy R. Stachowski
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
16
|
Raisinghani N, Alshahrani M, Gupta G, Tian H, Xiao S, Tao P, Verkhivker G. Interpretable Atomistic Prediction and Functional Analysis of Conformational Ensembles and Allosteric States in Protein Kinases Using AlphaFold2 Adaptation with Randomized Sequence Scanning and Local Frustration Profiling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580591. [PMID: 38496487 PMCID: PMC10942451 DOI: 10.1101/2024.02.15.580591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The groundbreaking achievements of AlphaFold2 (AF2) approaches in protein structure modeling marked a transformative era in structural biology. Despite the success of AF2 tools in predicting single protein structures, these methods showed intrinsic limitations in predicting multiple functional conformations of allosteric proteins and fold-switching systems. The recent NMR-based structural determination of the unbound ABL kinase in the active state and two inactive low-populated functional conformations that are unique for ABL kinase presents an ideal challenge for AF2 approaches. In the current study we employ several implementations of AF2 methods to predict protein conformational ensembles and allosteric states of the ABL kinase including (a) multiple sequence alignments (MSA) subsampling approach; (b) SPEACH_AF approach in which alanine scanning is performed on generated MSAs; and (c) introduced in this study randomized full sequence mutational scanning for manipulation of sequence variations combined with the MSA subsampling. We show that the proposed AF2 adaptation combined with local frustration mapping of conformational states enable accurate prediction of the ABL active and intermediate structures and conformational ensembles, also offering a robust approach for interpretable characterization of the AF2 predictions and limitations in detecting hidden allosteric states. We found that the large high frustration residue clusters are uniquely characteristic of the low-populated, fully inactive ABL form and can define energetically frustrated cracking sites of conformational transitions, presenting difficult targets for AF2 methods. This study uncovered previously unappreciated, fundamental connections between distinct patterns of local frustration in functional kinase states and AF2 successes/limitations in detecting low-populated frustrated conformations, providing a better understanding of benefits and limitations of current AF2-based adaptations in modeling of conformational ensembles.
Collapse
|