1
|
Barlow KA, Battles MB, Brown ME, Canfield K, Lu X, Lynaugh H, Morrill M, Rappazzo CG, Reyes SP, Sandberg C, Sharkey B, Strong C, Zhao J, Sivasubramanian A. Design of orthogonal constant domain interfaces to aid proper heavy/light chain pairing of bispecific antibodies. MAbs 2025; 17:2479531. [PMID: 40126074 PMCID: PMC11934185 DOI: 10.1080/19420862.2025.2479531] [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: 12/13/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
The correct pairing of cognate heavy and light chains is critical to the efficient manufacturing of IgG-like bispecific antibodies (bsAbs) from a single host cell. We present a general solution for the elimination of heavy chain (HC):light chain (LC) mispairs in bsAbs with κ LCs via the use of two orthogonal constant domain (CH1:Cκ ) interfaces comprising computationally designed amino acid substitutions. Substitutions were designed by Rosetta to introduce novel hydrogen bond (H-bond) networks at the CH1:Cκ interface, followed by Rosetta energy calculations to identify designs with enhanced pairing specificity and interface stability. Our final design, featuring a total of 11 amino acid substitutions across two Fab constant regions, was tested on a set of six IgG-like bsAbs featuring a diverse set of unmodified human antibody variable domains. Purity assessments showed near-complete elimination of LC mispairs, including in cases with high baseline mispairing with wild-type constant domains. The engineered bsAbs broadly recapitulated the antigen-binding and biophysical developability properties of their monospecific counterparts and no adverse immunogenicity signal was identified by an in vitro assay. Fab crystal structures containing engineered constant domain interfaces revealed no major perturbations relative to the wild-type coordinates and validated the presence of the designed hydrogen bond interactions. Our work enables the facile assembly of independently discovered IgG-like bispecific antibodies in a single-cell host and demonstrates a streamlined and generalizable computational and experimental workflow for redesigning conserved protein:protein interfaces.
Collapse
Affiliation(s)
| | | | | | | | - Xiaojun Lu
- Protein Analytics, Adimab, Lebanon, NH, USA
| | | | | | | | | | | | - Beth Sharkey
- High-Throughput Expression, Adimab, Lebanon, NH, USA
| | | | | | - Arvind Sivasubramanian
- Computational Biology, Adimab, Mountain View, CA, USA
- Platform Technologies, Adimab, Lebanon, NH, USA
| |
Collapse
|
2
|
Zhang J, Chen Y, Han B, Liu Y, Li X, Yang J, Liu Y, Cao Y, Liang D, Yu B. Mitigating PCOS progression: The protective effect of C-phycocyanin on ovarian granulosa cell pyroptosis via the NRF2/NLRP3/GSDMD pathway. Int Immunopharmacol 2025; 159:114917. [PMID: 40412127 DOI: 10.1016/j.intimp.2025.114917] [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: 03/13/2025] [Revised: 05/05/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Pyroptosis is a proinflammatory cell death process that contributes to inflammatory diseases. C-Phycocyanin (C-PC) is a water-soluble protein pigment primarily derived from cyanobacteria, and it exhibits anti-inflammatory effects. However, the role of natural C-PC in pyroptosis in human ovarian granulosa cells (GCs), particularly in conditions like polycystic ovary syndrome (PCOS), remains unclear. This study investigates the effects of C-PC on pyroptosis and its relevance to PCOS. METHODS Here dehydroepiandrosterone (DHEA) was used to induce PCOS in a mouse model that was characterized by an irregular oestrous cycle, cystic follicles and an elevated serum hormone level. DHEA treatment was used to induce GC pyroptosis. Scanning electron microscopy (SEM) was used to determine cell morphology. The expression levels of key proteins for oxidative stress and pyroptosis, including nuclear factor erythroid 2-related factor 2 (NRF2), NLR family of pyrroline-containing structural domain 3 (NLRP3) inflammasomes, cleaved caspase-1, and N-GSDMD were investigated in vivo and in vitro by Western blotting and immunofluorescence staining. RESULTS C-PC restored the estrous cycle in PCOS mice, reduced testosterone levels, and decreased cystic follicles. It attenuated PCOS progression by reducing oxidative stress level and suppressing GCs pyroptosis. At the cellular level, C-PC inhibited DHEA-induced GC pyroptosis by blocking NLRP3 inflammasome activation and lowering ROS, effects reversed by the NRF2 inhibitor (ML385). Molecular docking analysis and CETSA suggested that C-PC may protect against PCOS by activating the NRF2 pathway or by indirectly binding to the Ser27 site of GSDMD. In addition, C-PC suppressed ROS/p38-MAPK activation induced by DHEA, and p38-MAPK agonists diminished its effect on NLRP3 inflammasome activity and GC pyroptosis protection. CONCLUSION C-PC exerts a protective effect on GC pyroptosis through the NRF2/NLRP3/GSDMD and ROS/p-38 MAPK pathways, highlighting its potential to mitigate inflammation and its relevance to reproductive health issues like PCOS.
Collapse
Affiliation(s)
- Jing Zhang
- College of Grain Engineering, Anhui Vocational College of Grain Engineering, Hefei 230011, Anhui, China
| | - Yaxin Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Baoqing Han
- Belgorod Institute of Food Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Yang Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xuan Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Jun Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yajing Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Dan Liang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China.
| | - Biao Yu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
3
|
Yi X, Wu X, Zang E, He X, Chang X, Liu J, Shi L. Construction of safer hirudin-derived peptides with enhanced anticoagulant properties based on C-terminal active residue adaptation and modification. J Adv Res 2025:S2090-1232(25)00289-9. [PMID: 40320167 DOI: 10.1016/j.jare.2025.04.045] [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: 02/03/2025] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
INTRODUCTION Hirudin exerts anticoagulant effects by inhibiting the binding and catalytic activity of thrombin to fibrinogen. However, its rigid N-terminal region irreversibly occupies the thrombin catalytic center, raising concerns about potential bleeding. OBJECTIVES In this study, a novel lead compound, WPHVC_V1, which is based on the competitive binding mechanism of the hirudin variant WPHV_C, was developed and validated for in vitro and in vivo activity and safety. METHODS Saturation mutagenesis, molecular dynamics simulations and mutant protein activity assays were used to elucidate the competitive anticoagulant mechanism between WPHV_C and thrombin. Next, a recombinantly expressed tyrosylprotein sulfotransferase was used to modify and confirm the sulfation site on the C-terminal tyrosine of hirudin. Finally, a multisite aromatic amino acid mutation strategy was implemented to design and synthesize the lead anticoagulant, WPHVC_V1. RESULTS The acidic amino acid cluster in WPHV_C formed strong electrostatic interactions with the positively charged thrombin exosite I, blocking fibrinogen binding. The introduction of aromatic amino acids further stabilized the complex through π-π stacking and π-cation interactions. For example, mutation of 13E to A decreased the free energy of dissociation (ΔG) from 19.27 to 10.93 kcal·mol-1 and shortened the thrombin time (TT) from 42.00 s to 30.94 s, whereas mutation of 26 K to W increased the ΔG to 24.70 kcal·mol-1 and prolonged TT to 51.92 s. In addition, the aromatic effect of 20Y, combined with sulfation, synergistically enhanced binding. Based on these findings, the newly designed WPHVC_V1 showed a ΔG of 37.24 kcal·mol-1 and, at 0.1 mg/ml, increased TT/APTT/PT from 41.72/14.38/15.86 s (WPHV_C) to 62.08/23.38/22.22 s. In in vivo studies, WPHVC_V1 achieved tail thrombus inhibition in the mouse tail by reducing the length of the thrombus from 3.562 cm in CK to 1.853 cm (1.729 cm for sodium heparin and 2.530 cm for WPHV_C), completely inhibited thrombus formation in a carotid artery model and reduced tail bleeding time by 35.2 s compared with heparin sodium. Safety evaluations revealed that WPHVC_V1 did not cause hemolysis, had no significant effect on blood pressure or cause pathological changes in major organs. CONCLUSION These findings provide an initial foundation and sequence reference for the development of safe and effective anticoagulant drugs with potential for clinical translation.
Collapse
Affiliation(s)
- Xiaozhe Yi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Xiaoli Wu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Erhuan Zang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Xiaoli He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Xinyi Chang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Jinxin Liu
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China.
| | - Linchun Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China.
| |
Collapse
|
4
|
Calvo-Barreiro L, Secor M, Damjanovic J, Abdel-Rahman SA, Lin YS, Gabr M. Computational Design of a Bicyclic Peptide Inhibitor Targeting the ICOS/ICOS-L Protein-Protein Interaction. Chem Biol Drug Des 2025; 105:e70117. [PMID: 40317592 DOI: 10.1111/cbdd.70117] [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: 12/17/2024] [Revised: 03/19/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
The interaction between the inducible T-cell costimulatory molecule (ICOS) and its ligand (ICOS-L) is a critical pathway in T-cell activation and immune regulation. We computationally designed a bicyclic peptide (CP5) that inhibits the ICOS/ICOS-L protein-protein interaction (PPI). Using the structural insights derived from the ICOS/ICOS-L co-crystal structure (PDB: 6X4G) and bias-exchange metadynamics simulations (BE-META), we first designed monocyclic peptide candidates containing the β-strand (residues 51-55 51YVYWQ55) of ICOS-L that interact with ICOS. Using Rosetta's flex ddG calculations and further disulfide-bond restraint, we arrived at CP5 (cyclo-RVY[CQPGWC]WVLpG) as a potential ICOS/ICOS-L inhibitor. Using dynamic light scattering (DLS), we examined the interaction between CP5 and ICOS. Importantly, we validated the ICOS/ICOS-L inhibitory activity of CP5 using both TR-FRET assay and ELISA. Notably, CP5 demonstrated satisfactory in vitro pharmacokinetic properties, such as metabolic stability and lipophilicity, positioning it as a promising candidate for further drug development. Our findings provide a foundation for future drug discovery efforts aiming to develop cyclic peptides that specifically target the ICOS/ICOS-L interaction.
Collapse
Affiliation(s)
- Laura Calvo-Barreiro
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
| | - Maxim Secor
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Jovan Damjanovic
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Moustafa Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
5
|
Choi YJ, Nam YA, Hyun JY, Yu J, Mun Y, Yun SH, Lee W, Park CJ, Han BW, Lee BH. Impaired chaperone-mediated autophagy leads to abnormal SORT1 (sortilin 1) turnover and CES1-dependent triglyceride hydrolysis. Autophagy 2025; 21:827-839. [PMID: 39611307 PMCID: PMC11925108 DOI: 10.1080/15548627.2024.2435234] [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: 01/22/2024] [Revised: 11/07/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
SORT1 (sortilin 1), a member of the the Vps10 (vacuolar protein sorting 10) family, is involved in hepatic lipid metabolism by regulating very low-density lipoprotein (VLDL) secretion and facilitating the lysosomal degradation of CES1 (carboxylesterase 1), crucial for triglyceride (TG) breakdown in the liver. This study explores whether SORT1 is targeted for degradation by chaperone-mediated autophagy (CMA), a selective protein degradation pathway that directs proteins containing KFERQ-like motifs to lysosomes via LAMP2A (lysosomal-associated membrane protein 2A). Silencing LAMP2A or HSPA8/Hsc70 with siRNA increased cytosolic SORT1 protein levels. Leupeptin treatment induced lysosomal accumulation of SORT1, unaffected by siLAMP2A co-treatment, indicating CMA-dependent degradation. Human SORT1 contains five KFERQ-like motifs (658VVTKQ662, 730VREVK734, 733VKDLK737, 734KDLKK738, and 735DLKKK739), crucial for HSPA8 recognition; mutating any single amino acid within these motifs decreased HSPA8 binding. Furthermore, compromised CMA activity resulted in elevated SORT1-mediated degradation of CES1, contributing to increased lipid accumulation in hepatocytes. Consistent with in vitro findings, LAMP2A knockdown in mice exacerbated high-fructose diet-induced fatty liver, marked by increased SORT1 and decreased CES1 levels. Conversely, LAMP2A overexpression promoted SORT1 degradation and CES1D accumulation, counteracting fasting-induced CES1D suppression through CMA activation. Our findings reveal that SORT1 is a substrate of CMA, highlighting its crucial role in directing CES1 to lysosomes. Consequently, disrupting CMA-mediated SORT1 degradation significantly affects CES1-dependent TG hydrolysis, thereby affecting hepatic lipid homeostasis.Abbreviations: APOB: apolipoprotein B; CES1: carboxylesterase 1; CMA: chaperone-mediated autophagy; HSPA8/Hsc70: heat shock protein family A (Hsp70) member 8; LAMP2A: lysosomal associated membrane protein 2A; LDL-C: low-density lipoprotein-cholesterol; PLIN: perilipin; SORT1: sortilin 1; TG: triglyceride; VLDL: very low-density lipoprotein; Vps10: vacuolar protein sorting 10.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Gyeongsangbuk-do, Republic of Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yoon Ah Nam
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Ye Hyun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jihyeon Yu
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yewon Mun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Ho Yun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wonseok Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Cheon Jun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byung Woo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Zhu F, Rajan S, Hayes CF, Kwong KY, Goncalves AR, Zemla AT, Lau EY, Zhang Y, Cai Y, Goforth JW, Landajuela M, Gilchuk P, Kierny M, Dippel A, Amofah B, Kaplan G, Cadevilla Peano V, Morehouse C, Sparklin B, Gopalakrishnan V, Tuffy KM, Nguyen A, Beloor J, Kijak G, Liu C, Dijokaite-Guraliuc A, Mongkolsapaya J, Screaton GR, Petersen BK, Desautels TA, Bennett D, Conti S, Segelke BW, Arrildt KT, Kaul S, Grzesiak EA, da Silva FL, Bates TW, Earnhart CG, Hopkins S, Sundaram S, Esser MT, Francica JR, Faissol DM, LLNL Generative Unconstrained Intelligent Drug Engineering (GUIDE) consortium. Preemptive optimization of a clinical antibody for broad neutralization of SARS-CoV-2 variants and robustness against viral escape. SCIENCE ADVANCES 2025; 11:eadu0718. [PMID: 40153503 PMCID: PMC11952088 DOI: 10.1126/sciadv.adu0718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025]
Abstract
Most previously authorized clinical antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have lost neutralizing activity to recent variants due to rapid viral evolution. To mitigate such escape, we preemptively enhance AZD3152, an antibody authorized for prophylaxis in immunocompromised individuals. Using deep mutational scanning (DMS) on the SARS-CoV-2 antigen, we identify AZD3152 vulnerabilities at antigen positions F456 and D420. Through two iterations of computational antibody design that integrates structure-based modeling, machine-learning, and experimental validation, we co-optimize AZD3152 against 24 contemporary and previous SARS-CoV-2 variants, as well as 20 potential future escape variants. Our top candidate, 3152-1142, restores full potency (100-fold improvement) against the more recently emerged XBB.1.5+F456L variant that escaped AZD3152, maintains potency against previous variants of concern, and shows no additional vulnerability as assessed by DMS. This preemptive mitigation demonstrates a generalizable approach for optimizing existing antibodies against potential future viral escape.
Collapse
Affiliation(s)
- Fangqiang Zhu
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Saravanan Rajan
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Conor F. Hayes
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Ka Yin Kwong
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Andre R. Goncalves
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Adam T. Zemla
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Edmond Y. Lau
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Yi Zhang
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Yingyun Cai
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - John W. Goforth
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Mikel Landajuela
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Pavlo Gilchuk
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Michael Kierny
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Andrew Dippel
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Bismark Amofah
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Gilad Kaplan
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Vanessa Cadevilla Peano
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Christopher Morehouse
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Ben Sparklin
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | | | - Kevin M. Tuffy
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Amy Nguyen
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Jagadish Beloor
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Gustavo Kijak
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Chang Liu
- Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Aiste Dijokaite-Guraliuc
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Juthathip Mongkolsapaya
- Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Gavin R. Screaton
- Chinese Academy of Medical Science (CAMS) Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Brenden K. Petersen
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Thomas A. Desautels
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Drew Bennett
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Simone Conti
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Brent W. Segelke
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Kathryn T. Arrildt
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Samantha Kaul
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Emilia A. Grzesiak
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Felipe Leno da Silva
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Thomas W. Bates
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Christopher G. Earnhart
- Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense, US Department of Defense, Frederick, MD 21703, USA
| | | | - Shivshankar Sundaram
- Center for Bioengineering, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - Mark T. Esser
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Joseph R. Francica
- Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Daniel M. Faissol
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | | |
Collapse
|
7
|
Lebedenko OO, Polovinkin MS, Kazovskaia AA, Skrynnikov NR. PCANN Program for Structure-Based Prediction of Protein-Protein Binding Affinity: Comparison With Other Neural-Network Predictors. Proteins 2025. [PMID: 40116085 DOI: 10.1002/prot.26821] [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: 11/06/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/23/2025]
Abstract
In this communication, we introduce a new structure-based affinity predictor for protein-protein complexes. This predictor, dubbed PCANN (Protein Complex Affinity by Neural Network), uses the ESM-2 language model to encode the information about protein binding interfaces and graph attention network (GAT) to parlay this information intoK d $$ {K}_{\mathrm{d}} $$ predictions. In the tests employing two previously unused literature-extracted datasets, PCANN performed better than the best of the publicly available predictors, BindPPI, with mean absolute error (MAE) of 1.3 versus 1.4 kcal/mol. Further progress in the development ofK d $$ {K}_{\mathrm{d}} $$ predictors using deep learning models is faced with two problems: (i) the amount of experimental data available to train and test new predictors is limited and (ii) the availableK d $$ {K}_{\mathrm{d}} $$ data are often not very accurate and lack internal consistency with respect to measurement conditions. These issues can be potentially addressed through an AI-leveraged literature search followed by careful human curation and by introducing additional parameters to account for variations in experimental conditions.
Collapse
Affiliation(s)
- Olga O Lebedenko
- Laboratory of Biomolecular NMR, St. Petersburg State University, St. Petersburg, Russia
| | - Mikhail S Polovinkin
- Laboratory of Biomolecular NMR, St. Petersburg State University, St. Petersburg, Russia
| | - Anastasiia A Kazovskaia
- Laboratory of Biomolecular NMR, St. Petersburg State University, St. Petersburg, Russia
- Faculty of Mathematics & Computer Science, St. Petersburg State University, St. Petersburg, Russia
| | - Nikolai R Skrynnikov
- Laboratory of Biomolecular NMR, St. Petersburg State University, St. Petersburg, Russia
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
8
|
Singh S, Gapsys V, Aldeghi M, Schaller D, Rangwala AM, White JB, Bluck JP, Scheen J, Glass WG, Guo J, Hayat S, de Groot BL, Volkamer A, Christ CD, Seeliger MA, Chodera JD. Prospective Evaluation of Structure-Based Simulations Reveal Their Ability to Predict the Impact of Kinase Mutations on Inhibitor Binding. J Phys Chem B 2025; 129:2882-2902. [PMID: 40053698 PMCID: PMC12038917 DOI: 10.1021/acs.jpcb.4c07794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Abstract
Small molecule kinase inhibitors are critical in the modern treatment of cancers, evidenced by the existence of over 80 FDA-approved small-molecule kinase inhibitors. Unfortunately, intrinsic or acquired resistance, often causing therapy discontinuation, is frequently caused by mutations in the kinase therapeutic target. The advent of clinical tumor sequencing has opened additional opportunities for precision oncology to improve patient outcomes by pairing optimal therapies with tumor mutation profiles. However, modern precision oncology efforts are hindered by lack of sufficient biochemical or clinical evidence to classify each mutation as resistant or sensitive to existing inhibitors. Structure-based methods show promising accuracy in retrospective benchmarks at predicting whether a kinase mutation will perturb inhibitor binding, but comparisons are made by pooling disparate experimental measurements across different conditions. We present the first prospective benchmark of structure-based approaches on a blinded dataset of in-cell kinase inhibitor affinities to Abl kinase mutants using a NanoBRET reporter assay. We compare NanoBRET results to structure-based methods and their ability to estimate the impact of mutations on inhibitor binding (measured as ΔΔG). Comparing physics-based simulations, Rosetta, and previous machine learning models, we find that structure-based methods accurately classify kinase mutations as inhibitor-resistant or inhibitor-sensitizing, and each approach has a similar degree of accuracy. We show that physics-based simulations are best suited to estimate ΔΔG of mutations that are distal to the kinase active site. To probe modes of failure, we retrospectively investigate two clinically significant mutations poorly predicted by our methods, T315A and L298F, and find that starting configurations and protonation states significantly alter the accuracy of our predictions. Our experimental and computational measurements provide a benchmark for estimating the impact of mutations on inhibitor binding affinity for future methods and structure-based models. These structure-based methods have potential utility in identifying optimal therapies for tumor-specific mutations, predicting resistance mutations in the absence of clinical data, and identifying potential sensitizing mutations to established inhibitors.
Collapse
Affiliation(s)
- Sukrit Singh
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Vytautas Gapsys
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Matteo Aldeghi
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for multidisciplinary sciences, D-37077 Göttingen, Germany
| | - David Schaller
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Aziz M. Rangwala
- Department of Pharmacological Sciences, Stony Brook University Medical School, Stony Brook, NY 11794, United States
| | - Jessica B. White
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, United States
| | - Joseph P. Bluck
- Structural Biology & Computational Design, Research and Development, Pharmaceuticals, Bayer AG, 13342 Berlin, Germany
| | - Jenke Scheen
- Open Molecular Software Foundation, Davis, CA 95618, USA
| | - William G. Glass
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jiaye Guo
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sikander Hayat
- Department of medicine II, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Bert L. de Groot
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for multidisciplinary sciences, D-37077 Göttingen, Germany
| | - Andrea Volkamer
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Data Driven Drug Design, Faculty of Mathematics and Computer Sciences, Saarland University, 66123 Saarbrücken, Germany
| | - Clara D. Christ
- Structural Biology & Computational Design, Research and Development, Pharmaceuticals, Bayer AG, 13342 Berlin, Germany
| | - Markus A. Seeliger
- Department of Pharmacological Sciences, Stony Brook University Medical School, Stony Brook, NY 11794, United States
| | - John D. Chodera
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
9
|
Ullanat V, Jing B, Sledzieski S, Berger B. Learning the language of protein-protein interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642188. [PMID: 40166198 PMCID: PMC11956943 DOI: 10.1101/2025.03.09.642188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Protein Language Models (PLMs) trained on large databases of protein sequences have proven effective in modeling protein biology across a wide range of applications. However, while PLMs excel at capturing individual protein properties, they face challenges in natively representing protein-protein interactions (PPIs), which are crucial to understanding cellular processes and disease mechanisms. Here, we introduce MINT, a PLM specifically designed to model sets of interacting proteins in a contextual and scalable manner. Using unsupervised training on a large curated PPI dataset derived from the STRING database, MINT outperforms existing PLMs in diverse tasks relating to protein-protein interactions, including binding affinity prediction and estimation of mutational effects. Beyond these core capabilities, it excels at modeling interactions in complex protein assemblies and surpasses specialized models in antibody-antigen modeling and T cell receptor-epitope binding prediction. MINT's predictions of mutational impacts on oncogenic PPIs align with experimental studies, and it provides reliable estimates for the potential for cross-neutralization of antibodies against SARS-CoV-2 variants of concern. These findings position MINT as a powerful tool for elucidating complex protein interactions, with significant implications for biomedical research and therapeutic discovery.
Collapse
Affiliation(s)
- Varun Ullanat
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Bowen Jing
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
| | - Samuel Sledzieski
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Center for Computational Biology, Flatiron Insitute, New York, NY
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA
- Department of Mathematics, Massachusetts Institute of Technology, MA
| |
Collapse
|
10
|
Singh S, Gapsys V, Aldeghi M, Schaller D, Rangwala AM, White JB, Bluck JP, Scheen J, Glass WG, Guo J, Hayat S, de Groot BL, Volkamer A, Christ CD, Seeliger MA, Chodera JD. Prospective evaluation of structure-based simulations reveal their ability to predict the impact of kinase mutations on inhibitor binding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.15.623861. [PMID: 40060600 PMCID: PMC11888192 DOI: 10.1101/2024.11.15.623861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Small molecule kinase inhibitors are critical in the modern treatment of cancers, evidenced by the existence of over 80 FDA-approved small-molecule kinase inhibitors. Unfortunately, intrinsic or acquired resistance, often causing therapy discontinuation, is frequently caused by mutations in the kinase therapeutic target. The advent of clinical tumor sequencing has opened additional opportunities for precision oncology to improve patient outcomes by pairing optimal therapies with tumor mutation profiles. However, modern precision oncology efforts are hindered by lack of sufficient biochemical or clinical evidence to classify each mutation as resistant or sensitive to existing inhibitors. Structure-based methods show promising accuracy in retrospective benchmarks at predicting whether a kinase mutation will perturb inhibitor binding, but comparisons are made by pooling disparate experimental measurements across different conditions. We present the first prospective benchmark of structure-based approaches on a blinded dataset of in-cell kinase inhibitor affinities to Abl kinase mutants using a NanoBRET reporter assay. We compare NanoBRET results to structure-based methods and their ability to estimate the impact of mutations on inhibitor binding (measured as ΔΔG). Comparing physics-based simulations, Rosetta, and previous machine learning models, we find that structure-based methods accurately classify kinase mutations as inhibitor-resistant or inhibitor-sensitizing, and each approach has a similar degree of accuracy. We show that physics-based simulations are best suited to estimate ΔΔG of mutations that are distal to the kinase active site. To probe modes of failure, we retrospectively investigate two clinically significant mutations poorly predicted by our methods, T315A and L298F, and find that starting configurations and protonation states significantly alter the accuracy of our predictions. Our experimental and computational measurements provide a benchmark for estimating the impact of mutations on inhibitor binding affinity for future methods and structure-based models. These structure-based methods have potential utility in identifying optimal therapies for tumor-specific mutations, predicting resistance mutations in the absence of clinical data, and identifying potential sensitizing mutations to established inhibitors.
Collapse
Affiliation(s)
- Sukrit Singh
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Vytautas Gapsys
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, Beerse 2340, Belgium
| | - Matteo Aldeghi
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for multidisciplinary sciences, D-37077 Göttingen, Germany
| | - David Schaller
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Aziz M. Rangwala
- Department of Pharmacological Sciences, Stony Brook University Medical School, Stony Brook, NY 11794, United States
| | - Jessica B. White
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, United States
| | - Joseph P. Bluck
- Structural Biology & Computational Design, Research and Development, Pharmaceuticals, Bayer AG, 13342 Berlin, Germany
| | - Jenke Scheen
- Open Molecular Software Foundation, Davis, CA 95618, USA
| | - William G. Glass
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jiaye Guo
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Sikander Hayat
- Department of medicine II, University Hospital Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Bert L. de Groot
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for multidisciplinary sciences, D-37077 Göttingen, Germany
| | - Andrea Volkamer
- In Silico Toxicology and Structural Bioinformatics, Institute of Physiology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Data Driven Drug Design, Faculty of Mathematics and Computer Sciences, Saarland University, 66123 Saarbrücken, Germany
| | - Clara D. Christ
- Structural Biology & Computational Design, Research and Development, Pharmaceuticals, Bayer AG, 13342 Berlin, Germany
| | - Markus A. Seeliger
- Department of Pharmacological Sciences, Stony Brook University Medical School, Stony Brook, NY 11794, United States
| | - John D. Chodera
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
11
|
Fu H, Mo X, Ivanov AA. Decoding the functional impact of the cancer genome through protein-protein interactions. Nat Rev Cancer 2025; 25:189-208. [PMID: 39810024 DOI: 10.1038/s41568-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/16/2025]
Abstract
Acquisition of genomic mutations enables cancer cells to gain fitness advantages under selective pressure and, ultimately, leads to oncogenic transformation. Interestingly, driver mutations, even within the same gene, can yield distinct phenotypes and clinical outcomes, necessitating a mutation-focused approach. Conversely, cellular functions are governed by molecular machines and signalling networks that are mostly controlled by protein-protein interactions (PPIs). The functional impact of individual genomic alterations could be transmitted through regulated nodes and hubs of PPIs. Oncogenic mutations may lead to modified residues of proteins, enabling interactions with other proteins that the wild-type protein does not typically interact with, or preventing interactions with proteins that the wild-type protein usually interacts with. This can result in the rewiring of molecular signalling cascades and the acquisition of an oncogenic phenotype. Here, we review the altered PPIs driven by oncogenic mutations, discuss technologies for monitoring PPIs and provide a functional analysis of mutation-directed PPIs. These driver mutation-enabled PPIs and mutation-perturbed PPIs present a new paradigm for the development of tumour-specific therapeutics. The intersection of cancer variants and altered PPI interfaces represents a new frontier for understanding oncogenic rewiring and developing tumour-selective therapeutic strategies.
Collapse
Affiliation(s)
- Haian Fu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| | - Xiulei Mo
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Andrey A Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Emory University, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
12
|
Xu W, Li A, Zhao Y, Peng Y. Decoding the effects of mutation on protein interactions using machine learning. BIOPHYSICS REVIEWS 2025; 6:011307. [PMID: 40013003 PMCID: PMC11857871 DOI: 10.1063/5.0249920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/28/2025]
Abstract
Accurately predicting mutation-caused binding free energy changes (ΔΔGs) on protein interactions is crucial for understanding how genetic variations affect interactions between proteins and other biomolecules, such as proteins, DNA/RNA, and ligands, which are vital for regulating numerous biological processes. Developing computational approaches with high accuracy and efficiency is critical for elucidating the mechanisms underlying various diseases, identifying potential biomarkers for early diagnosis, and developing targeted therapies. This review provides a comprehensive overview of recent advancements in predicting the impact of mutations on protein interactions across different interaction types, which are central to understanding biological processes and disease mechanisms, including cancer. We summarize recent progress in predictive approaches, including physicochemical-based, machine learning, and deep learning methods, evaluating the strengths and limitations of each. Additionally, we discuss the challenges related to the limitations of mutational data, including biases, data quality, and dataset size, and explore the difficulties in developing accurate prediction tools for mutation-induced effects on protein interactions. Finally, we discuss future directions for advancing these computational tools, highlighting the capabilities of advancing technologies, such as artificial intelligence to drive significant improvements in mutational effects prediction.
Collapse
Affiliation(s)
- Wang Xu
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Anbang Li
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Yunjie Zhao
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Yunhui Peng
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
13
|
Zeides P, Bellmann-Sickert K, Zhang R, Seel CJ, Most V, Schoeder CT, Groll M, Gulder T. Unraveling the molecular basis of substrate specificity and halogen activation in vanadium-dependent haloperoxidases. Nat Commun 2025; 16:2083. [PMID: 40021637 PMCID: PMC11871015 DOI: 10.1038/s41467-025-57023-1] [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: 12/19/2023] [Accepted: 02/10/2025] [Indexed: 03/03/2025] Open
Abstract
Vanadium-dependent haloperoxidases (VHPOs) are biotechnologically valuable and operationally versatile biocatalysts. VHPOs share remarkable active-site structural similarities yet display variable reactivity and selectivity. The factors dictating substrate specificity and, thus, a general understanding of VHPO reaction control still need to be discovered. This work's strategic single-point mutation in the cyanobacterial bromoperoxidase AmVHPO facilitates a selectivity switch to allow aryl chlorination. This mutation induces loop formation that interacts with the neighboring protein monomer, creating a tunnel to the active sites. Structural analysis of the substrate-R425S-mutant complex reveals a substrate-binding site at the interface of two adjacent units. There, residues Glu139 and Phe401 interact with arenes, extending the substrate residence time close to the vanadate cofactor and stabilizing intermediates. Our findings validate the long-debated existence of direct substrate binding and provide a detailed VHPO mechanistic understanding. This work will pave the way for a broader application of VHPOs in diverse chemical processes.
Collapse
Affiliation(s)
- P Zeides
- Biomimetic Catalysis, Catalysis Research Center, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
- Faculty of Chemistry and Mineralogy, Institute of Organic Chemistry, Leipzig University, Leipzig, Germany
| | - K Bellmann-Sickert
- Faculty of Chemistry and Mineralogy, Institute of Organic Chemistry, Leipzig University, Leipzig, Germany
| | - Ru Zhang
- Faculty of Chemistry and Mineralogy, Institute of Organic Chemistry, Leipzig University, Leipzig, Germany
- Organic Chemistry, Saarland University, Saarbruecken, Germany
| | - C J Seel
- Biomimetic Catalysis, Catalysis Research Center, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - V Most
- Faculty of Medicine, Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| | - C T Schoeder
- Faculty of Medicine, Institute for Drug Discovery, Leipzig University, Leipzig, Germany
| | - M Groll
- Department of Bioscience, Center for Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - T Gulder
- Biomimetic Catalysis, Catalysis Research Center, TUM School of Natural Sciences, Technical University of Munich, Garching, Germany.
- Faculty of Chemistry and Mineralogy, Institute of Organic Chemistry, Leipzig University, Leipzig, Germany.
- Organic Chemistry, Saarland University, Saarbruecken, Germany.
- Synthesis of Natural-Product Derived Drugs, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarbrücken, Germany.
| |
Collapse
|
14
|
Islam SM, Hasan M, Alam J, Dey A, Molineaux D. In Silico Screening, Molecular Dynamics Simulation and Binding Free Energy Identify Single-Point Mutations That Destabilize p53 and Reduce Binding to DNA. Proteins 2025; 93:498-514. [PMID: 39264222 PMCID: PMC11695177 DOI: 10.1002/prot.26747] [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: 06/18/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Considering p53's pivotal role as a tumor suppressor protein, proactive identification and characterization of potentially harmful p53 mutations are crucial before they appear in the population. To address this, four computational prediction tools-SIFT, Polyphen-2, PhD-SNP, and MutPred2-utilizing sequence-based and machine-learning algorithms, were employed to identify potentially deleterious p53 nsSNPs (nonsynonymous single nucleotide polymorphisms) that may impact p53 structure, dynamics, and binding with DNA. These computational methods identified three variants, namely, C141Y, C238S, and L265P, as detrimental to p53 stability. Furthermore, molecular dynamics (MD) simulations revealed that all three variants exhibited heightened structural flexibility compared to the native protein, especially the C141Y and L265P mutations. Consequently, due to the altered structure of mutant p53, the DNA-binding affinity of all three variants decreased by approximately 1.8 to 9.7 times compared to wild-type p53 binding with DNA (14 μM). Notably, the L265P mutation exhibited an approximately ten-fold greater reduction in binding affinity. Consequently, the presence of the L265P mutation in p53 could pose a substantial risk to humans. Given that p53 regulates abnormal tumor growth, this research carries significant implications for surveillance efforts and the development of anticancer therapies.
Collapse
Affiliation(s)
- Shahidul M. Islam
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Mehedi Hasan
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Jahidul Alam
- Department of Molecular Biology and Biotechnology, Queen’s University Belfast, Northern Ireland, BT7 1NN, United Kingdom
| | - Anonya Dey
- Department of Genetic Engineering and Biotechnology, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Dylan Molineaux
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| |
Collapse
|
15
|
Sumner CA, Schwedler JL, McCoy KM, Holland J, Duva V, Gelperin D, Busygina V, Stefan MA, Martinez DV, Juarros MA, Phillips AM, Weilhammer DR, Grigoryan G, Kent MS, Harmon BN. Combining computational modeling and experimental library screening to affinity-mature VEEV-neutralizing antibody F5. Protein Sci 2025; 34:e70043. [PMID: 39840828 PMCID: PMC11752144 DOI: 10.1002/pro.70043] [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: 08/27/2024] [Revised: 12/24/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
Engineered monoclonal antibodies have proven to be highly effective therapeutics in recent viral outbreaks. However, despite technical advancements, an ability to rapidly adapt or increase antibody affinity and by extension, therapeutic efficacy, has yet to be fully realized. We endeavored to stand-up such a pipeline using molecular modeling combined with experimental library screening to increase the affinity of F5, a monoclonal antibody with potent neutralizing activity against Venezuelan Equine Encephalitis Virus (VEEV), to recombinant VEEV (IAB) E1E2 antigen. We modeled the F5/E1E2 binding interface and generated predictions for mutations to improve binding using a Rosetta-based approach and dTERMen, an informatics approach. The modeling was complicated by the fact that a high-resolution structure of F5 is not available and the H3 loop of F5 exceeds the length for which current modeling approaches can determine a unique structure. A subset of the predicted mutations from both methods were incorporated into a phage display library of scFvs. This library and a library generated by error-prone PCR were screened for binding affinity to the recombinant antigen. Results from the screens identified favorable mutations which were incorporated into 12 human-IgG1 variants. The best variant, containing eight mutations, improved KD from 0.63 nM (parental) to 0.01 nM. While this did not improve neutralization or therapeutic potency of F5 against IAB, it did increase cross-reactivity to other closely related VEEV epizootic and enzootic strains, demonstrating the potential of this method to rapidly adapt existing therapeutics to emerging viral strains.
Collapse
MESH Headings
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/genetics
- Models, Molecular
- Encephalitis Virus, Venezuelan Equine/immunology
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Peptide Library
- Antibody Affinity
- Humans
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/genetics
- Single-Chain Antibodies/chemistry
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
Collapse
Affiliation(s)
- Christopher A. Sumner
- Department of Biotechnology and BioengineeringSandia National LaboratoriesLivermoreCaliforniaUSA
| | - Jennifer L. Schwedler
- Department of Biotechnology and BioengineeringSandia National LaboratoriesLivermoreCaliforniaUSA
| | - Katherine Maia McCoy
- Department of Molecular and Cell BiologyDartmouth CollegeHanoverNew HampshireUSA
| | - Jack Holland
- Department of Computer ScienceDartmouth CollegeHanoverNew HampshireUSA
| | | | | | | | - Maxwell A. Stefan
- Department of Biotechnology and BioengineeringSandia National LaboratoriesLivermoreCaliforniaUSA
| | - Daniella V. Martinez
- Department of Molecular and MicrobiologySandia National LaboratoriesAlbuquerqueNew MexicoUSA
| | - Miranda A. Juarros
- Department of Molecular and MicrobiologySandia National LaboratoriesAlbuquerqueNew MexicoUSA
| | - Ashlee M. Phillips
- Division of Biosciences and BiotechnologyLawrence Livermore National LaboratoriesLivermoreCaliforniaUSA
| | - Dina R. Weilhammer
- Division of Biosciences and BiotechnologyLawrence Livermore National LaboratoriesLivermoreCaliforniaUSA
| | - Gevorg Grigoryan
- Department of Molecular and Cell BiologyDartmouth CollegeHanoverNew HampshireUSA
- Department of Computer ScienceDartmouth CollegeHanoverNew HampshireUSA
| | - Michael S. Kent
- Department of Molecular and MicrobiologySandia National LaboratoriesAlbuquerqueNew MexicoUSA
| | - Brooke N. Harmon
- Department of Biotechnology and BioengineeringSandia National LaboratoriesLivermoreCaliforniaUSA
| |
Collapse
|
16
|
Cabrera-Alarcon JL, Cruz R, Rosa-Moreno M, Latorre-Pellicer A, de Almeida SD, Riancho JA, Rojas-Martinez A, Flores C, Lapunzina P, Sánchez-Cabo F, Carracedo Á, Enriquez JA. Shaping current European mitochondrial haplogroup frequency in response to infection: the case of SARS-CoV-2 severity. Commun Biol 2025; 8:33. [PMID: 39789223 PMCID: PMC11718132 DOI: 10.1038/s42003-024-07314-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025] Open
Abstract
The frequency of mitochondrial DNA haplogroups (mtDNA-HG) in humans is known to be shaped by migration and repopulation. Mounting evidence indicates that mtDNA-HG are not phenotypically neutral, and selection may contribute to its distribution. Haplogroup H, the most abundant in Europe, improved survival in sepsis. Here we developed a random forest trained model for mitochondrial haplogroup calling using data procured from GWAS arrays. Our results reveal that in the context of the SARS-CoV-2 pandemic, HV branch were found to represent protective factors against the development of critical SARS-CoV-2 in an analysis of 14,349 patients. These results highlight the role of mtDNA in the response to infectious diseases and support the proposal that its expansion and population proportion has been influenced by selection through successive pandemics.
Collapse
Affiliation(s)
- José Luis Cabrera-Alarcon
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, E-28029, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable.(CIBERFES) Instituto de Salud Carlos III, Madrid, E-28029, Spain
| | - Raquel Cruz
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, E-28029, Spain
- Grupo de Medicina Xenómica-CIMUS-Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marina Rosa-Moreno
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, E-28029, Spain
| | - Ana Latorre-Pellicer
- Grupo de Genética Clínica y Genómica Funcional, Facultad de Medicina, Universidad de Zaragoza, IIS Aragón, CIBERER-GCV02, E-50009, Zaragoza, Spain
| | - Silvia Diz de Almeida
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, E-28029, Spain
- Grupo de Medicina Xenómica-CIMUS-Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - José A Riancho
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable.(CIBERFES) Instituto de Salud Carlos III, Madrid, E-28029, Spain
- Universidad de Cantabria, Cantabria, Spain
- Hospital U M Valdecilla, Cantabria, Spain
- DIVAL, Cantabria, Spain
| | | | - Carlos Flores
- Centre for Biomedical Network Research on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Pablo Lapunzina
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, E-28029, Spain
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz-IDIPAZ, ERN-ITHACA-European Reference Network, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, E-28029, Spain
| | - Ángel Carracedo
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, E-28029, Spain
- Grupo de Medicina Xenómica-CIMUS-Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica (SERGAS), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, E-28029, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable.(CIBERFES) Instituto de Salud Carlos III, Madrid, E-28029, Spain.
| |
Collapse
|
17
|
Bu H, Pei C, Ouyang M, Chen Y, Yu L, Huang X, Tan Y. The antitumor peptide M1-20 induced the degradation of CDK1 through CUL4-DDB1-DCAF1-involved ubiquitination. Cancer Gene Ther 2025; 32:61-70. [PMID: 39562696 DOI: 10.1038/s41417-024-00855-8] [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: 06/14/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/21/2024]
Abstract
CDK1 is an oncogenic serine/threonine kinase known to play an important role in the regulation of the cell cycle. FOXM1, as one of the CDK1 substrates, requires binding of CDK1/CCNB1 complex for phosphorylation-dependent recruitment of p300/CBP coactivators to mediate transcriptional activity. Previous studies from our laboratory found that a novel peptide (M1-20) derived from the C-terminus of FOXM1 exhibited potent inhibitory effects for cancer cells. Based on these proofs and to explore the inhibitory mechanism of M1-20, we designed experiments and found that CDK1 served as an important target of M1-20. M1-20 enhanced the ubiquitination and degradation of CDK1 by CUL4-DDB1-DCAF1 complexes through the proteasome pathway. M1-20 could also affect the formation of CDK1/CCNB1 complexes. In addition, compared to RO3306, a CDK1 inhibitor, M1-20 exhibited excellent inhibitory effects in FVB/N MMTV-PyVT murine model of spontaneous breast cancer. These results suggested that M1-20 was a potential CDK1 inhibitor for the treatment of cancer.
Collapse
Affiliation(s)
- Huitong Bu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chaozhu Pei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
| | - Min Ouyang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
| | - Yan Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
| | - Li Yu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
| | - Xiaoqin Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan Engineering Research Center for Anticancer Targeted Protein Pharmaceuticals, Hunan University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Oh JS, Kim DS, So YS, Hong S, Yoo SH, Park CS, Park JH, Seo DH. Construction and enzymatic characterization of a monomeric variant of dimeric amylosucrase from Deinococcus geothermalis. Int J Biol Macromol 2024; 285:138249. [PMID: 39631600 DOI: 10.1016/j.ijbiomac.2024.138249] [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: 07/30/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Amylosucrase (ASase; E.C. 2.4.1.4), a member of glycoside hydrolase family 13 (GH13), produces α-1,4-glucans and sucrose isomers using sucrose as its sole substrate. This study identifies and characterizes the dimeric structure of ASase from Deinococcus geothermalis (DgAS), highlighting essential amino acid residues for maintaining the dimeric state. The monomeric form, DgAS R30A, exhibited a higher affinity for sucrose compared to the wild-type (WT), especially during the formation of the ASase-glucose intermediate complex and subsequent hydrolysis. Notably, DgAS R30A produced a higher proportion of α-glucans with a degree of polymerization (DP) of ≤20 and fewer α-glucans with a DP of ≥31. This suggested that the reduced surface area of the oligosaccharide binding site in the monomeric form led to decreased binding of longer-chain maltooligosaccharides, favoring the formation of shorter DP α-glucans. Kinetic analysis revealed significantly lower Michaelis constants (Km) for DgAS R30A's total and hydrolysis activities, with the overall performance (kcat/Km) values for DgAS R30A exceeded those of the WT at all sucrose concentrations. Here, we report the first high-resolution homodimeric DgAS structure, revealing conserved active site residues and a unique dimerization interface. This study enhances our understanding of the molecular factors influencing the oligomeric state and enzyme activities.
Collapse
Affiliation(s)
- Ju-Seon Oh
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| | - Da Som Kim
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk State, 54596 Republic of Korea
| | - Yun-Sang So
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| | - Seungpyo Hong
- Department of Molecular Biology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Sang-Ho Yoo
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| | - Cheon-Seok Park
- Department of Food Science and Biotechnology, Graduate School of Biotechnology and Institute of Life Science and Resources, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Jung Hee Park
- Division of Biotechnology, Advanced Institute of Environment and Bioscience, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk State, 54596 Republic of Korea.
| | - Dong-Ho Seo
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
19
|
Totaro MG, Vide U, Zausinger R, Winkler A, Oberdorfer G. ESM-scan-A tool to guide amino acid substitutions. Protein Sci 2024; 33:e5221. [PMID: 39565080 PMCID: PMC11577456 DOI: 10.1002/pro.5221] [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: 03/15/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/21/2024]
Abstract
Protein structure prediction and (re)design have gone through a revolution in the last 3 years. The tremendous progress in these fields has been almost exclusively driven by readily available machine learning algorithms applied to protein folding and sequence design problems. Despite these advancements, predicting site-specific mutational effects on protein stability and function remains an unsolved problem. This is a persistent challenge, mainly because the free energy of large systems is very difficult to compute with absolute accuracy and subtle changes to protein structures are hard to capture with computational models. Here, we describe the implementation and use of ESM-Scan, which uses the ESM zero-shot predictor to scan entire protein sequences for preferential amino acid changes, thus enabling in silico deep mutational scanning experiments. We benchmark ESM-Scan on its predictive capabilities for stability and functionality of sequence changes using three publicly available datasets and proceed by experimentally testing the tool's performance on a challenging test case of a blue-light-activated diguanylate cyclase from Methylotenera species (MsLadC), where it accurately predicted the importance of a highly conserved residue in a region involved in allosteric product inhibition. Our experimental results show that the ESM-zero shot model is capable of inferring the effects of a set of amino acid substitutions in their correlation between predicted fitness and experimental results. ESM-Scan is publicly available at https://huggingface.co/spaces/thaidaev/zsp.
Collapse
Affiliation(s)
| | - Uršula Vide
- Institute of BiochemistryGraz University of TechnologyGrazAustria
| | - Regina Zausinger
- Institute of BiochemistryGraz University of TechnologyGrazAustria
| | - Andreas Winkler
- Institute of BiochemistryGraz University of TechnologyGrazAustria
- BioTechMedGrazAustria
| | - Gustav Oberdorfer
- Institute of BiochemistryGraz University of TechnologyGrazAustria
- BioTechMedGrazAustria
| |
Collapse
|
20
|
Kumar A K, Rathore RS. Categorization of hotspots into three types - weak, moderate and strong to distinguish protein-protein versus protein-peptide interactions. J Biomol Struct Dyn 2024; 42:9348-9360. [PMID: 37649387 DOI: 10.1080/07391102.2023.2252077] [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: 04/15/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
Protein-protein and protein-peptide interactions (PPI and PPepI) belong to a similar category of interactions, yet seemingly subtle differences exist among them. To characterize differences between protein-protein (PP) and protein-peptide (PPep) interactions, we have focussed on two important classes of residues-hotspot and anchor residues. Using implicit solvation-based free energy calculations, a very large-scale alanine scanning has been performed on benchmark datasets, consisting of over 5700 interface residues. The differences in the two categories are more pronounced, if the data were divided into three distinct types, namely - weak hotspots (having binding free energy loss upon Ala mutation, ΔΔG, ∼2-10 kcal/mol), moderate hotspots (ΔΔG, ∼10-20 kcal/mol) and strong hotspots (ΔΔG ≥ ∼20 kcal/mol). The analysis suggests that for PPI, weak hotspots are predominantly populated by polar and hydrophobic residues. The distribution shifts towards charged and polar residues for moderate hotspot and charged residues (principally Arg) are overwhelmingly present in the strong hotspot. On the other hand, in the PPepI dataset, the distribution shifts from predominantly hydrophobic and polar (in the weak type) to almost similar preference for polar, hydrophobic and charged residues (in moderate type) and finally the charged residue (Arg) and Trp are mostly occupied in the strong type. The preferred anchor residues in both categories are Arg, Tyr and Leu, possessing bulky side chain and which also strike a delicate balance between side chain flexibility and rigidity. The present knowledge should aid in effective design of biologics, by augmentation or disruption of PPIs with peptides or peptidomimetics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kiran Kumar A
- Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - R S Rathore
- Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| |
Collapse
|
21
|
Tse AL, Acreman CM, Ricardo-Lax I, Berrigan J, Lasso G, Balogun T, Kearns FL, Casalino L, McClain GL, Chandran AM, Lemeunier C, Amaro RE, Rice CM, Jangra RK, McLellan JS, Chandran K, Miller EH. Distinct pathways for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses. PLoS Pathog 2024; 20:e1012704. [PMID: 39546542 PMCID: PMC11602109 DOI: 10.1371/journal.ppat.1012704] [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: 07/05/2024] [Revised: 11/27/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014-CoV, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the S1 N-terminal domain, uncovered through the rescue and serial passage of a virus bearing the FPPR substitution, further enhanced spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles' heels that could be targeted with countermeasures.
Collapse
Affiliation(s)
- Alexandra L. Tse
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Cory M. Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Jacob Berrigan
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Gorka Lasso
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Toheeb Balogun
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Fiona L. Kearns
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Lorenzo Casalino
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Georgia L. McClain
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Amartya Mudry Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Charlotte Lemeunier
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Rommie E. Amaro
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Rohit K. Jangra
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Present address: Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kartik Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Emily Happy Miller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| |
Collapse
|
22
|
Zhang Y, Dong M, Deng J, Wu J, Zhao Q, Gao X, Xiong D. Graph masked self-distillation learning for prediction of mutation impact on protein-protein interactions. Commun Biol 2024; 7:1400. [PMID: 39462102 PMCID: PMC11513059 DOI: 10.1038/s42003-024-07066-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Assessing mutation impact on the binding affinity change (ΔΔG) of protein-protein interactions (PPIs) plays a crucial role in unraveling structural-functional intricacies of proteins and developing innovative protein designs. In this study, we present a deep learning framework, PIANO, for improved prediction of ΔΔG in PPIs. The PIANO framework leverages a graph masked self-distillation scheme for protein structural geometric representation pre-training, which effectively captures the structural context representations surrounding mutation sites, and makes predictions using a multi-branch network consisting of multiple encoders for amino acids, atoms, and protein sequences. Extensive experiments demonstrated its superior prediction performance and the capability of pre-trained encoder in capturing meaningful representations. Compared to previous methods, PIANO can be widely applied on both holo complex structures and apo monomer structures. Moreover, we illustrated the practical applicability of PIANO in highlighting pathogenic mutations and crucial proteins, and distinguishing de novo mutations in disease cases and controls in PPI systems. Overall, PIANO offers a powerful deep learning tool, which may provide valuable insights into the study of drug design, therapeutic intervention, and protein engineering.
Collapse
Affiliation(s)
- Yuan Zhang
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Mingyuan Dong
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Junsheng Deng
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Jiafeng Wu
- Key Laboratory of Intelligent Computing and Information Processing of Ministry of Education, Xiangtan University, Xiangtan, 411105, China
| | - Qiuye Zhao
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA.
| | - Xieping Gao
- Hunan Provincial Key Laboratory of Intelligent Computing and Language Information Processing, Hunan Normal University, Changsha, 410081, China.
| | - Dapeng Xiong
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA.
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
23
|
Tydings CW, Singh B, Smith AW, Ledwitch KV, Brown BP, Lovly CM, Walker AS, Meiler J. Analysis of EGFR binding hotspots for design of new EGFR inhibitory biologics. Protein Sci 2024; 33:e5141. [PMID: 39275996 PMCID: PMC11400634 DOI: 10.1002/pro.5141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 09/16/2024]
Abstract
The epidermal growth factor (EGF) receptor (EGFR) is activated by the binding of one of seven EGF-like ligands to its ectodomain. Ligand binding results in EGFR dimerization and stabilization of the active receptor conformation subsequently leading to activation of downstream signaling. Aberrant activation of EGFR contributes to cancer progression through EGFR overexpression/amplification, modulation of its positive and negative regulators, and/or activating mutations within EGFR. EGFR targeted therapeutic antibodies prevent dimerization and interaction with endogenous ligands by binding the ectodomain of EGFR. However, these antibodies have had limited success in the clinic, partially due to EGFR ectodomain resistance mutations, and are only applicable to a subset of patients with EGFR-driven cancers. These limitations suggest that alternative EGFR targeted biologics need to be explored for EGFR-driven cancer therapy. To this end, we analyze the EGFR interfaces of known inhibitory biologics with determined structures in the context of endogenous ligands, using the Rosetta macromolecular modeling software to highlight the most important interactions on a per-residue basis. We use this analysis to identify the structural determinants of EGFR targeted biologics. We suggest that commonly observed binding motifs serve as the basis for rational design of new EGFR targeted biologics, such as peptides, antibodies, and nanobodies.
Collapse
Affiliation(s)
- Claiborne W. Tydings
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Bhuminder Singh
- Department of Medicine – Division of Gastroenterology, Hepatology, and NutritionVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Adam W. Smith
- Department of Chemistry and BiochemistryTexas Tech UniversityLubbockTexasUSA
| | - Kaitlyn V. Ledwitch
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Benjamin P. Brown
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
| | - Christine M. Lovly
- Department of Medicine – Division of Hematology and OncologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Allison S. Walker
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Department of Biological SciencesVanderbilt UniversityNashvilleTennesseeUSA
| | - Jens Meiler
- Department of ChemistryVanderbilt UniversityNashvilleTennesseeUSA
- Center for Structural BiologyVanderbilt UniversityNashvilleTennesseeUSA
- Institute for Drug DiscoveryLeipzig University Medical SchoolLeipzigSACGermany
| |
Collapse
|
24
|
Chen YC, Sargsyan K, Wright JD, Chen YH, Huang YS, Lim C. PPI-hotspot ID for detecting protein-protein interaction hot spots from the free protein structure. eLife 2024; 13:RP96643. [PMID: 39283314 PMCID: PMC11405013 DOI: 10.7554/elife.96643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Experimental detection of residues critical for protein-protein interactions (PPI) is a time-consuming, costly, and labor-intensive process. Hence, high-throughput PPI-hot spot prediction methods have been developed, but they have been validated using relatively small datasets, which may compromise their predictive reliability. Here, we introduce PPI-hotspotID, a novel method for identifying PPI-hot spots using the free protein structure, and validated it on the largest collection of experimentally confirmed PPI-hot spots to date. We explored the possibility of detecting PPI-hot spots using (i) FTMap in the PPI mode, which identifies hot spots on protein-protein interfaces from the free protein structure, and (ii) the interface residues predicted by AlphaFold-Multimer. PPI-hotspotID yielded better performance than FTMap and SPOTONE, a webserver for predicting PPI-hot spots given the protein sequence. When combined with the AlphaFold-Multimer-predicted interface residues, PPI-hotspotID yielded better performance than either method alone. Furthermore, we experimentally verified several PPI-hotspotID-predicted PPI-hot spots of eukaryotic elongation factor 2. Notably, PPI-hotspotID can reveal PPI-hot spots not obvious from complex structures, including those in indirect contact with binding partners. PPI-hotspotID serves as a valuable tool for understanding PPI mechanisms and aiding drug design. It is available as a web server (https://ppihotspotid.limlab.dnsalias.org/) and open-source code (https://github.com/wrigjz/ppihotspotid/).
Collapse
Affiliation(s)
- Yao Chi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Karen Sargsyan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jon D Wright
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Hsien Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
25
|
Cai H, Zhang Z, Wang M, Zhong B, Li Q, Zhong Y, Wu Y, Ying T, Tang J. Pretrainable geometric graph neural network for antibody affinity maturation. Nat Commun 2024; 15:7785. [PMID: 39242604 PMCID: PMC11379722 DOI: 10.1038/s41467-024-51563-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 08/13/2024] [Indexed: 09/09/2024] Open
Abstract
Increasing the binding affinity of an antibody to its target antigen is a crucial task in antibody therapeutics development. This paper presents a pretrainable geometric graph neural network, GearBind, and explores its potential in in silico affinity maturation. Leveraging multi-relational graph construction, multi-level geometric message passing and contrastive pretraining on mass-scale, unlabeled protein structural data, GearBind outperforms previous state-of-the-art approaches on SKEMPI and an independent test set. A powerful ensemble model based on GearBind is then derived and used to successfully enhance the binding of two antibodies with distinct formats and target antigens. ELISA EC50 values of the designed antibody mutants are decreased by up to 17 fold, and KD values by up to 6.1 fold. These promising results underscore the utility of geometric deep learning and effective pretraining in macromolecule interaction modeling tasks.
Collapse
Affiliation(s)
- Huiyu Cai
- BioGeometry, Beijing, China
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Zuobai Zhang
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Mingkai Wang
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bozitao Zhong
- Mila-Québec AI Institute, Montréal, QC, Canada
- Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
| | - Quanxiao Li
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuxuan Zhong
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanling Wu
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China.
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Tianlei Ying
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai, China.
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Jian Tang
- BioGeometry, Beijing, China.
- Mila-Québec AI Institute, Montréal, QC, Canada.
- Department of Decision Sciences, HEC Montréal, Montréal, QC, Canada.
| |
Collapse
|
26
|
Sampson JM, Cannon DA, Duan J, Epstein JCK, Sergeeva AP, Katsamba PS, Mannepalli SM, Bahna FA, Adihou H, Guéret SM, Gopalakrishnan R, Geschwindner S, Rees DG, Sigurdardottir A, Wilkinson T, Dodd RB, De Maria L, Mobarec JC, Shapiro L, Honig B, Buchanan A, Friesner RA, Wang L. Robust Prediction of Relative Binding Energies for Protein-Protein Complex Mutations Using Free Energy Perturbation Calculations. J Mol Biol 2024; 436:168640. [PMID: 38844044 PMCID: PMC11339910 DOI: 10.1016/j.jmb.2024.168640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how Protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.
Collapse
Affiliation(s)
- Jared M Sampson
- Schrödinger, Inc., Life Sciences Software, New York, NY, USA
| | - Daniel A Cannon
- Schrödinger, GmbH, Life Sciences Software, Mannheim, Germany
| | - Jianxin Duan
- Schrödinger, GmbH, Life Sciences Software, Mannheim, Germany
| | | | - Alina P Sergeeva
- Columbia University, Department of Systems Biology, New York, NY, USA
| | | | - Seetha M Mannepalli
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA
| | - Fabiana A Bahna
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA
| | - Hélène Adihou
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden; Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Stéphanie M Guéret
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden; Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Ranganath Gopalakrishnan
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden; Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Stefan Geschwindner
- AstraZeneca, Mechanistic and Structural Biology, Discovery Sciences, R&D, Gothenburg, Sweden
| | - D Gareth Rees
- AstraZeneca, Biologics Engineering, R&D, Cambridge, UK
| | | | | | - Roger B Dodd
- AstraZeneca, Biologics Engineering, R&D, Cambridge, UK
| | - Leonardo De Maria
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Juan Carlos Mobarec
- AstraZeneca, Mechanistic and Structural Biology, Discovery Sciences, R&D, Cambridge, UK
| | - Lawrence Shapiro
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA; Columbia University, Department of Biochemistry and Molecular Biophysics, New York, NY, USA
| | - Barry Honig
- Columbia University, Department of Systems Biology, New York, NY, USA; Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA; Columbia University, Department of Biochemistry and Molecular Biophysics, New York, NY, USA; Columbia University, Department of Medicine, New York, NY, USA
| | | | | | - Lingle Wang
- Schrödinger, Inc., Life Sciences Software, New York, NY, USA.
| |
Collapse
|
27
|
Yi X, Liu J, Zang E, Tian Y, Liu J, Shi L. Exploring a Hirudin variant from nonhematophagous leeches: Unraveling full-length sequence, alternative splicing, function, and potential as a novel anticoagulant polypeptide. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118257. [PMID: 38677578 DOI: 10.1016/j.jep.2024.118257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leeches exhibit robust anticoagulant activity, making them useful for treating cardiovascular diseases in traditional Chinese medicine. Whitmania pigra, the primary source species of leech-derived medicinal compounds in China, has been demonstrated to possess formidable anticoagulant properties. Hirudin-like peptides, recognized as potent thrombin inhibitors, are prevalent in hematophagous leeches. Considering that W. pigra is a nonhematophagic leech, the following question arises: does a hirudin variant exist in this species? AIM OF THE STUDY In this study we identified the hirudin-encoding gene (WP_HV1) in the W. pigra genome. The goal of this study was to assess its anticoagulant activity and analyze the related mechanisms. MATERIALS AND METHODS In this study, a hirudin-encoding gene, WP_HV1, was identified from the W. pigra genome, and its accurate coding sequence (CDS) was validated through cloning from cDNA extracted from fresh W. pigra specimens. The structure of WP_HV1 and the amino acids associated with its anticoagulant activity were determined by sequence and structural analysis and prediction of its binding energy to thrombin. E. coli was used for the expression of WP_HV1 and recombinant proteins with various structures and mutants. The anticoagulant activity of the synthesized recombinant proteins was then confirmed using thrombin time (TT). RESULTS Validation of the WP_HV1 gene was accomplished, and three alternative splices were discovered. The TT of the blank sample exceeded that of the recombinant WP_HV1 sample by 1.74 times (0.05 mg/ml), indicating positive anticoagulant activity. The anticoagulant activity of WP_HV1 was found to be associated with its C-terminal tyrosine, along with the presence of 9 acidic amino acids on both the left and right sides. A significant reduction in the corresponding TT was observed for the mutated amino acids compared to those of the wild type, with decreases of 4.8, 6.6, and 3.9 s, respectively. In addition, the anticoagulant activity of WP_HV1 was enhanced and prolonged for 2.7 s when the lysine-67 residue was mutated to tryptophan. CONCLUSION Only one hirudin-encoding variant was identified in W. pigra. The active amino acids associated with anticoagulation in WP_HV1 were resolved and validated, revealing a novel source for screening and developing new anticoagulant drugs.
Collapse
Affiliation(s)
- Xiaozhe Yi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Jiali Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Erhuan Zang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China
| | - Yu Tian
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Hebei Key Laboratory of Study and Exploitation of Chinese Medicine, Chengde Medical University, Chengde 067000, China
| | - Jinxin Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China.
| | - Linchun Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China; Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing 100193, China.
| |
Collapse
|
28
|
Chamness LM, Kuntz CP, McKee AG, Penn WD, Hemmerich CM, Rusch DB, Woods H, Dyotima, Meiler J, Schlebach JP. Divergent folding-mediated epistasis among unstable membrane protein variants. eLife 2024; 12:RP92406. [PMID: 39078397 PMCID: PMC11288631 DOI: 10.7554/elife.92406] [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: 07/31/2024] Open
Abstract
Many membrane proteins are prone to misfolding, which compromises their functional expression at the plasma membrane. This is particularly true for the mammalian gonadotropin-releasing hormone receptor GPCRs (GnRHR). We recently demonstrated that evolutionary GnRHR modifications appear to have coincided with adaptive changes in cotranslational folding efficiency. Though protein stability is known to shape evolution, it is unclear how cotranslational folding constraints modulate the synergistic, epistatic interactions between mutations. We therefore compared the pairwise interactions formed by mutations that disrupt the membrane topology (V276T) or tertiary structure (W107A) of GnRHR. Using deep mutational scanning, we evaluated how the plasma membrane expression of these variants is modified by hundreds of secondary mutations. An analysis of 251 mutants in three genetic backgrounds reveals that V276T and W107A form distinct epistatic interactions that depend on both the severity and the mechanism of destabilization. V276T forms predominantly negative epistatic interactions with destabilizing mutations in soluble loops. In contrast, W107A forms positive interactions with mutations in both loops and transmembrane domains that reflect the diminishing impacts of the destabilizing mutations in variants that are already unstable. These findings reveal how epistasis is remodeled by conformational defects in membrane proteins and in unstable proteins more generally.
Collapse
Affiliation(s)
- Laura M Chamness
- Department of Chemistry, Indiana UniversityBloomingtonUnited States
| | - Charles P Kuntz
- The James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue UniversityWest LafayetteUnited States
| | - Andrew G McKee
- Department of Chemistry, Indiana UniversityBloomingtonUnited States
| | - Wesley D Penn
- Department of Chemistry, Indiana UniversityBloomingtonUnited States
| | | | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana UniversityBloomingtonUnited States
| | - Hope Woods
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
- Chemical and Physical Biology Program, Vanderbilt UniversityNashvilleUnited States
| | - Dyotima
- Department of Chemistry, Indiana UniversityBloomingtonUnited States
| | - Jens Meiler
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
- Institute for Drug Discovery, Leipzig UniversityLeipzigGermany
| | - Jonathan P Schlebach
- The James Tarpo Jr. and Margaret Tarpo Department of Chemistry, Purdue UniversityWest LafayetteUnited States
| |
Collapse
|
29
|
Liang T, Sun ZY, Hines MG, Penrose KJ, Hao Y, Chu X, Mellors JW, Dimitrov DS, Xie XQ, Li W, Feng Z. AI-based IsAb2.0 for antibody design. Brief Bioinform 2024; 25:bbae445. [PMID: 39285513 PMCID: PMC11405125 DOI: 10.1093/bib/bbae445] [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: 05/31/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Therapeutic antibody design has garnered widespread attention, highlighting its interdisciplinary importance. Advancements in technology emphasize the critical role of designing nanobodies and humanized antibodies in antibody engineering. However, current experimental methods are costly and time-consuming. Computational approaches, while progressing, faced limitations due to insufficient structural data and the absence of a standardized protocol. To tackle these challenges, our lab previously developed IsAb1.0, an in silico antibody design protocol. Yet, IsAb1.0 lacked accuracy, had a complex procedure, and required extensive antibody bioinformation. Moreover, it overlooked nanobody and humanized antibody design, hindering therapeutic antibody development. Building upon IsAb1.0, we enhanced our design protocol with artificial intelligence methods to create IsAb2.0. IsAb2.0 utilized AlphaFold-Multimer (2.3/3.0) for accurate modeling and complex construction without templates and employed the precise FlexddG method for in silico antibody optimization. Validated through optimization of a humanized nanobody J3 (HuJ3) targeting HIV-1 gp120, IsAb2.0 predicted five mutations that can improve HuJ3-gp120 binding affinity. These predictions were confirmed by commercial software and validated through binding and neutralization assays. IsAb2.0 streamlined antibody design, offering insights into future techniques to accelerate immunotherapy development.
Collapse
Affiliation(s)
- Tianjian Liang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, Pittsburgh, PA 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Drug Discovery Institute, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Department of Computational Biology, School of Medicine, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15261, United States
- Department of Structural Biology, School of Medicine, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, United States
| | - Ze-Yu Sun
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, Pittsburgh, PA 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Drug Discovery Institute, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Department of Computational Biology, School of Medicine, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15261, United States
- Department of Structural Biology, School of Medicine, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, United States
| | - Margaret G Hines
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - Kerri Jo Penrose
- Division of Infectious Diseases, Department of Medicine, Center for AIDS Research, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - Yixuan Hao
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, Pittsburgh, PA 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Drug Discovery Institute, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Department of Computational Biology, School of Medicine, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15261, United States
- Department of Structural Biology, School of Medicine, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, United States
| | - Xiaojie Chu
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - John W Mellors
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, Center for AIDS Research, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - Dimiter S Dimitrov
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, Pittsburgh, PA 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Drug Discovery Institute, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Department of Computational Biology, School of Medicine, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15261, United States
- Department of Structural Biology, School of Medicine, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, United States
| | - Wei Li
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, University of Pittsburgh, 335 Sutherland Drive, Pittsburgh, PA 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Drug Discovery Institute, University of Pittsburgh, 3501 Terrace St, Pittsburgh, PA 15261, United States
- Department of Computational Biology, School of Medicine, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15261, United States
- Department of Structural Biology, School of Medicine, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA 15261, United States
| |
Collapse
|
30
|
Tse AL, Acreman CM, Ricardo-Lax I, Berrigan J, Lasso G, Balogun T, Kearns FL, Casalino L, McClain GL, Chandran AM, Lemeunier C, Amaro RE, Rice CM, Jangra RK, McLellan JS, Chandran K, Miller EH. Distinct pathway for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600393. [PMID: 38979151 PMCID: PMC11230278 DOI: 10.1101/2024.06.24.600393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the spike N-terminal domain, uncovered through forward-genetic selection, interacted epistatically with the FPPR substitution to synergistically enhance spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles' heels that could be targeted with countermeasures.
Collapse
|
31
|
Yu G, Zhao Q, Bi X, Wang J. DDAffinity: predicting the changes in binding affinity of multiple point mutations using protein 3D structure. Bioinformatics 2024; 40:i418-i427. [PMID: 38940145 PMCID: PMC11211828 DOI: 10.1093/bioinformatics/btae232] [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: 06/29/2024] Open
Abstract
MOTIVATION Mutations are the crucial driving force for biological evolution as they can disrupt protein stability and protein-protein interactions which have notable impacts on protein structure, function, and expression. However, existing computational methods for protein mutation effects prediction are generally limited to single point mutations with global dependencies, and do not systematically take into account the local and global synergistic epistasis inherent in multiple point mutations. RESULTS To this end, we propose a novel spatial and sequential message passing neural network, named DDAffinity, to predict the changes in binding affinity caused by multiple point mutations based on protein 3D structures. Specifically, instead of being on the whole protein, we perform message passing on the k-nearest neighbor residue graphs to extract pocket features of the protein 3D structures. Furthermore, to learn global topological features, a two-step additive Gaussian noising strategy during training is applied to blur out local details of protein geometry. We evaluate DDAffinity on benchmark datasets and external validation datasets. Overall, the predictive performance of DDAffinity is significantly improved compared with state-of-the-art baselines on multiple point mutations, including end-to-end and pre-training based methods. The ablation studies indicate the reasonable design of all components of DDAffinity. In addition, applications in nonredundant blind testing, predicting mutation effects of SARS-CoV-2 RBD variants, and optimizing human antibody against SARS-CoV-2 illustrate the effectiveness of DDAffinity. AVAILABILITY AND IMPLEMENTATION DDAffinity is available at https://github.com/ak422/DDAffinity.
Collapse
Affiliation(s)
- Guanglei Yu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
- Medical Engineering and Technology College, Xinjiang Medical University, Urumqi 830017, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| | - Xuehua Bi
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
- Medical Engineering and Technology College, Xinjiang Medical University, Urumqi 830017, China
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha 410083, China
| |
Collapse
|
32
|
Narkhede Y, Saxena R, Sharma T, Conarty JP, Ramirez VT, Motsa BB, Amiar S, Li S, Chapagain PP, Wiest O, Stahelin RV. Computational and experimental identification of keystone interactions in Ebola virus matrix protein VP40 dimer formation. Protein Sci 2024; 33:e4978. [PMID: 38591637 PMCID: PMC11002992 DOI: 10.1002/pro.4978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/01/2024] [Accepted: 03/17/2024] [Indexed: 04/10/2024]
Abstract
The Ebola virus (EBOV) is a lipid-enveloped virus with a negative sense RNA genome that can cause severe and often fatal viral hemorrhagic fever. The assembly and budding of EBOV is regulated by the matrix protein, VP40, which is a peripheral protein that associates with anionic lipids at the inner leaflet of the plasma membrane. VP40 is sufficient to form virus-like particles (VLPs) from cells, which are nearly indistinguishable from authentic virions. Due to the restrictions of studying EBOV in BSL-4 facilities, VP40 has served as a surrogate in cellular studies to examine the EBOV assembly and budding process from the host cell plasma membrane. VP40 is a dimer where inhibition of dimer formation halts budding and formation of new VLPs as well as VP40 localization to the plasma membrane inner leaflet. To better understand VP40 dimer stability and critical amino acids to VP40 dimer formation, we integrated computational approaches with experimental validation. Site saturation/alanine scanning calculation, combined with molecular mechanics-based generalized Born with Poisson-Boltzmann surface area (MM-GB/PBSA) method and molecular dynamics simulations were used to predict the energetic contribution of amino acids to VP40 dimer stability and the hydrogen bonding network across the dimer interface. These studies revealed several previously unknown interactions and critical residues predicted to impact VP40 dimer formation. In vitro and cellular studies were then pursued for a subset of VP40 mutations demonstrating reduction in dimer formation (in vitro) or plasma membrane localization (in cells). Together, the computational and experimental approaches revealed critical residues for VP40 dimer stability in an alpha-helical interface (between residues 106-117) as well as in a loop region (between residues 52-61) below this alpha-helical region. This study sheds light on the structural origins of VP40 dimer formation and may inform the design of a small molecule that can disrupt VP40 dimer stability.
Collapse
Affiliation(s)
- Yogesh Narkhede
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Roopashi Saxena
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
| | - Tej Sharma
- Department of PhysicsFlorida International UniversityMiamiFloridaUSA
| | - Jacob P. Conarty
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
| | - Valentina Toro Ramirez
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
- Pharmaceutical ChemistryUniversidad CESMedellínColombia
| | - Balindile B. Motsa
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
| | - Souad Amiar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
| | - Sheng Li
- Department of MedicineUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Prem P. Chapagain
- Department of PhysicsFlorida International UniversityMiamiFloridaUSA
- Biomolecular Sciences Institute, Florida International UniversityMiamiFloridaUSA
| | - Olaf Wiest
- Department of Chemistry and BiochemistryUniversity of Notre DameNotre DameIndianaUSA
| | - Robert V. Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute for Inflammation, Immunology and Infectious DiseasePurdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
33
|
Desautels TA, Arrildt KT, Zemla AT, Lau EY, Zhu F, Ricci D, Cronin S, Zost SJ, Binshtein E, Scheaffer SM, Dadonaite B, Petersen BK, Engdahl TB, Chen E, Handal LS, Hall L, Goforth JW, Vashchenko D, Nguyen S, Weilhammer DR, Lo JKY, Rubinfeld B, Saada EA, Weisenberger T, Lee TH, Whitener B, Case JB, Ladd A, Silva MS, Haluska RM, Grzesiak EA, Earnhart CG, Hopkins S, Bates TW, Thackray LB, Segelke BW, Lillo AM, Sundaram S, Bloom JD, Diamond MS, Crowe JE, Carnahan RH, Faissol DM. Computationally restoring the potency of a clinical antibody against Omicron. Nature 2024; 629:878-885. [PMID: 38720086 PMCID: PMC11111397 DOI: 10.1038/s41586-024-07385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/04/2024] [Indexed: 05/21/2024]
Abstract
The COVID-19 pandemic underscored the promise of monoclonal antibody-based prophylactic and therapeutic drugs1-3 and revealed how quickly viral escape can curtail effective options4,5. When the SARS-CoV-2 Omicron variant emerged in 2021, many antibody drug products lost potency, including Evusheld and its constituent, cilgavimab4-6. Cilgavimab, like its progenitor COV2-2130, is a class 3 antibody that is compatible with other antibodies in combination4 and is challenging to replace with existing approaches. Rapidly modifying such high-value antibodies to restore efficacy against emerging variants is a compelling mitigation strategy. We sought to redesign and renew the efficacy of COV2-2130 against Omicron BA.1 and BA.1.1 strains while maintaining efficacy against the dominant Delta variant. Here we show that our computationally redesigned antibody, 2130-1-0114-112, achieves this objective, simultaneously increases neutralization potency against Delta and subsequent variants of concern, and provides protection in vivo against the strains tested: WA1/2020, BA.1.1 and BA.5. Deep mutational scanning of tens of thousands of pseudovirus variants reveals that 2130-1-0114-112 improves broad potency without increasing escape liabilities. Our results suggest that computational approaches can optimize an antibody to target multiple escape variants, while simultaneously enriching potency. Our computational approach does not require experimental iterations or pre-existing binding data, thus enabling rapid response strategies to address escape variants or lessen escape vulnerabilities.
Collapse
Affiliation(s)
- Thomas A Desautels
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Kathryn T Arrildt
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Adam T Zemla
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Edmond Y Lau
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Fangqiang Zhu
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Dante Ricci
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Stephanie Cronin
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elad Binshtein
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Suzanne M Scheaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bernadeta Dadonaite
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Brenden K Petersen
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Taylor B Engdahl
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elaine Chen
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lynn Hall
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John W Goforth
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Denis Vashchenko
- Applications Simulations and Quality Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Sam Nguyen
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
- Google, Alphabet Inc., Mountain View, CA, USA
| | - Dina R Weilhammer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Jacky Kai-Yin Lo
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Bonnee Rubinfeld
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Edwin A Saada
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Tracy Weisenberger
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Tek-Hyung Lee
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Bradley Whitener
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Vir Biotechnology, San Francisco, CA, USA
| | - James B Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander Ladd
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Mary S Silva
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Rebecca M Haluska
- Applications Simulations and Quality Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Emilia A Grzesiak
- Global Security Computing Applications Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Christopher G Earnhart
- Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense, US Department of Defense, Frederick, MD, USA
| | | | - Thomas W Bates
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Larissa B Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brent W Segelke
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | | | - Shivshankar Sundaram
- Center for Bioengineering, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Jesse D Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel M Faissol
- Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, USA.
| |
Collapse
|
34
|
Douglas JT, Johnson DK, Roy A, Park T. Use of phosphotyrosine-containing peptides to target SH2 domains: Antagonist peptides of the Crk/CrkL-p130Cas axis. Methods Enzymol 2024; 698:301-342. [PMID: 38886037 PMCID: PMC11542726 DOI: 10.1016/bs.mie.2024.04.013] [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: 06/20/2024]
Abstract
Protein-protein interactions between SH2 domains and segments of proteins that include a post-translationally phosphorylated tyrosine residue (pY) underpin numerous signal transduction cascades that allow cells to respond to their environment. Dysregulation of the writing, erasing, and reading of these posttranslational modifications is a hallmark of human disease, notably cancer. Elucidating the precise role of the SH2 domain-containing adaptor proteins Crk and CrkL in tumor cell migration and invasion is challenging because there are no specific and potent antagonists available. Crk and CrkL SH2s interact with a region of the docking protein p130Cas containing 15 potential pY-containing tetrapeptide motifs. This chapter summarizes recent efforts toward peptide antagonists for this Crk/CrkL-p130Cas interaction. We describe our protocol for recombinant expression and purification of Crk and CrkL SH2s for functional assays and our procedure to determine the consensus binding motif from the p130Cas sequence. To develop a more potent antagonist, we employ methods often associated with structure-based drug design. Computational docking using Rosetta FlexPepDock, which accounts for peptides having a greater number of conformational degrees of freedom than small organic molecules that typically constitute libraries, provides quantitative docking metrics to prioritize candidate peptides for experimental testing. A battery of biophysical assays, including fluorescence polarization, differential scanning fluorimetry and saturation transfer difference nuclear magnetic resonance spectroscopy, were employed to assess the candidates. In parallel, GST pulldown competition assays characterized protein-protein binding in vitro. Taken together, our methodology yields peptide antagonists of the Crk/CrkL-p130Cas axis that will be used to validate targets, assess druggability, foster in vitro assay development, and potentially serve as lead compounds for therapeutic intervention.
Collapse
Affiliation(s)
- Justin T. Douglas
- Nuclear Magnetic Resonance Core Lab, University of Kansas, Lawrence, KS 66047, USA
| | - David K. Johnson
- Computational Chemical Biology Core Lab, NIH COBRE in Chemical Biology of Infectious Disease, University of Kansas, Lawrence, Kansas 66047, USA
| | - Anuradha Roy
- High Throughput Screening Laboratory, University of Kansas, Lawrence, KS 66047, USA
| | - Taeju Park
- Department of Pediatrics, Children’s Mercy Kansas City and University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
35
|
Sampson JM, Cannon DA, Duan J, Epstein JCK, Sergeeva AP, Katsamba PS, Mannepalli SM, Bahna FA, Adihou H, Guéret SM, Gopalakrishnan R, Geschwindner S, Rees DG, Sigurdardottir A, Wilkinson T, Dodd RB, De Maria L, Mobarec JC, Shapiro L, Honig B, Buchanan A, Friesner RA, Wang L. Robust prediction of relative binding energies for protein-protein complex mutations using free energy perturbation calculations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590325. [PMID: 38712280 PMCID: PMC11071377 DOI: 10.1101/2024.04.22.590325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Computational free energy-based methods have the potential to significantly improve throughput and decrease costs of protein design efforts. Such methods must reach a high level of reliability, accuracy, and automation to be effectively deployed in practical industrial settings in a way that impacts protein design projects. Here, we present a benchmark study for the calculation of relative changes in protein-protein binding affinity for single point mutations across a variety of systems from the literature, using free energy perturbation (FEP+) calculations. We describe a method for robust treatment of alternate protonation states for titratable amino acids, which yields improved correlation with and reduced error compared to experimental binding free energies. Following careful analysis of the largest outlier cases in our dataset, we assess limitations of the default FEP+ protocols and introduce an automated script which identifies probable outlier cases that may require additional scrutiny and calculates an empirical correction for a subset of charge-related outliers. Through a series of three additional case study systems, we discuss how protein FEP+ can be applied to real-world protein design projects, and suggest areas of further study.
Collapse
Affiliation(s)
| | | | - Jianxin Duan
- Schrödinger, GmbH, Life Sciences Software, Mannheim, Germany
| | | | - Alina P. Sergeeva
- Columbia University, Department of Systems Biology, New York, NY, USA
| | | | - Seetha M. Mannepalli
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA, 10027
| | - Fabiana A. Bahna
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA, 10027
| | - Hélène Adihou
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden
- Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Stéphanie M. Guéret
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden
- Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Ranganath Gopalakrishnan
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, Gothenburg, Sweden
- Max Planck Institute of Molecular Physiology, AstraZeneca-MPI Satellite Unit, Dortmund, Germany
| | - Stefan Geschwindner
- AstraZeneca, Mechanistic and Structural Biology, Discovery Sciences, R&D, Cambridge, UK
| | | | | | | | - Roger B. Dodd
- AstraZeneca, Biologics Engineering, R&D, Cambridge, UK
| | - Leonardo De Maria
- AstraZeneca, Medicinal Chemistry, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Juan Carlos Mobarec
- AstraZeneca, Mechanistic and Structural Biology, Discovery Sciences, R&D, Cambridge, UK
| | - Lawrence Shapiro
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA, 10027
- Columbia University, Department of Biochemistry and Molecular Biophysics, New York, NY, USA
| | - Barry Honig
- Columbia University, Department of Systems Biology, New York, NY, USA
- Columbia University, Zuckerman Mind Brain Behavior Institute, New York, NY, USA, 10027
- Columbia University, Department of Biochemistry and Molecular Biophysics, New York, NY, USA
- Columbia University, Department of Medicine, New York, NY, USA
| | | | | | - Lingle Wang
- Schrödinger, Inc., Life Sciences Software, New York, NY, USA
| |
Collapse
|
36
|
Rouleau FD, Dubé AK, Gagnon-Arsenault I, Dibyachintan S, Pageau A, Després PC, Lagüe P, Landry CR. Deep mutational scanning of Pneumocystis jirovecii dihydrofolate reductase reveals allosteric mechanism of resistance to an antifolate. PLoS Genet 2024; 20:e1011252. [PMID: 38683847 PMCID: PMC11125491 DOI: 10.1371/journal.pgen.1011252] [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: 10/17/2023] [Revised: 05/24/2024] [Accepted: 04/08/2024] [Indexed: 05/02/2024] Open
Abstract
Pneumocystis jirovecii is a fungal pathogen that causes pneumocystis pneumonia, a disease that mainly affects immunocompromised individuals. This fungus has historically been hard to study because of our inability to grow it in vitro. One of the main drug targets in P. jirovecii is its dihydrofolate reductase (PjDHFR). Here, by using functional complementation of the baker's yeast ortholog, we show that PjDHFR can be inhibited by the antifolate methotrexate in a dose-dependent manner. Using deep mutational scanning of PjDHFR, we identify mutations conferring resistance to methotrexate. Thirty-one sites spanning the protein have at least one mutation that leads to resistance, for a total of 355 high-confidence resistance mutations. Most resistance-inducing mutations are found inside the active site, and many are structurally equivalent to mutations known to lead to resistance to different antifolates in other organisms. Some sites show specific resistance mutations, where only a single substitution confers resistance, whereas others are more permissive, as several substitutions at these sites confer resistance. Surprisingly, one of the permissive sites (F199) is without direct contact to either ligand or cofactor, suggesting that it acts through an allosteric mechanism. Modeling changes in binding energy between F199 mutants and drug shows that most mutations destabilize interactions between the protein and the drug. This evidence points towards a more important role of this position in resistance than previously estimated and highlights potential unknown allosteric mechanisms of resistance to antifolate in DHFRs. Our results offer unprecedented resources for the interpretation of mutation effects in the main drug target of an uncultivable fungal pathogen.
Collapse
Affiliation(s)
- Francois D. Rouleau
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
| | - Alexandre K. Dubé
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
| | - Isabelle Gagnon-Arsenault
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
| | - Soham Dibyachintan
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
| | - Alicia Pageau
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
| | - Philippe C. Després
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
| | - Patrick Lagüe
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
| | - Christian R. Landry
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, Québec, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
- Regroupement Québécois de recherche sur la fonction, la structure et l’ingénierie des protéines (PROTEO), Université du Québec à Montréal, Montréal, Québec, Canada
- Centre de recherche en données massives de l’Université Laval (CRDM_UL), Québec, Québec, Canada
- Département de Biologie, Faculté des Sciences et de Génie, Université Laval, Québec, Québec, Canada
| |
Collapse
|
37
|
Yang C, Sun X, Wu G. New insights into GATOR2-dependent interactions and its conformational changes in amino acid sensing. Biosci Rep 2024; 44:BSR20240038. [PMID: 38372438 PMCID: PMC10938194 DOI: 10.1042/bsr20240038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Eukaryotic cells coordinate growth under different environmental conditions via mechanistic target of rapamycin complex 1 (mTORC1). In the amino-acid-sensing signalling pathway, the GATOR2 complex, containing five evolutionarily conserved subunits (WDR59, Mios, WDR24, Seh1L and Sec13), is required to regulate mTORC1 activity by interacting with upstream CASTOR1 (arginine sensor) and Sestrin2 (leucine sensor and downstream GATOR1 complex). GATOR2 complex utilizes β-propellers to engage with CASTOR1, Sestrin2 and GATOR1, removal of these β-propellers results in substantial loss of mTORC1 capacity. However, structural information regarding the interface between amino acid sensors and GATOR2 remains elusive. With the recent progress of the AI-based tool AlphaFold2 (AF2) for protein structure prediction, structural models were predicted for Sentrin2-WDR24-Seh1L and CASTOR1-Mios β-propeller. Furthermore, the effectiveness of relevant residues within the interface was examined using biochemical experiments combined with molecular dynamics (MD) simulations. Notably, fluorescence resonance energy transfer (FRET) analysis detected the structural transition of GATOR2 in response to amino acid signals, and the deletion of Mios β-propeller severely impeded that change at distinct arginine levels. These findings provide structural perspectives on the association between GATOR2 and amino acid sensors and can facilitate future research on structure determination and function.
Collapse
Affiliation(s)
- Can Yang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, the Joint International Research Laboratory of Metabolic and Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai, China
| | - Xuan Sun
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, the Joint International Research Laboratory of Metabolic and Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, the Joint International Research Laboratory of Metabolic and Developmental Sciences MOE, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Tandiana R, Barletta GP, Soler MA, Fortuna S, Rocchia W. Computational Mutagenesis of Antibody Fragments: Disentangling Side Chains from ΔΔ G Predictions. J Chem Theory Comput 2024; 20:2630-2642. [PMID: 38445482 DOI: 10.1021/acs.jctc.3c01225] [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: 03/07/2024]
Abstract
The development of highly potent antibodies and antibody fragments as binding agents holds significant implications in fields such as biosensing and biotherapeutics. Their binding strength is intricately linked to the arrangement and composition of residues at the binding interface. Computational techniques offer a robust means to predict the three-dimensional structure of these complexes and to assess the affinity changes resulting from mutations. Given the interdependence of structure and affinity prediction, our objective here is to disentangle their roles. We aim to evaluate independently six side-chain reconstruction methods and ten binding affinity estimation techniques. This evaluation was pivotal in predicting affinity alterations due to single mutations, a key step in computational affinity maturation protocols. Our analysis focuses on a data set comprising 27 distinct antibody/hen egg white lysozyme complexes, each with crystal structures and experimentally determined binding affinities. Using six different side-chain reconstruction methods, we transformed each structure into its corresponding mutant via in silico single-point mutations. Subsequently, these structures undergo minimization and molecular dynamics simulation. We therefore estimate ΔΔG values based on the original crystal structure, its energy-minimized form, and the ensuing molecular dynamics trajectories. Our research underscores the critical importance of selecting reliable side-chain reconstruction methods and conducting thorough molecular dynamics simulations to accurately predict the impact of mutations. In summary, our study demonstrates that the integration of conformational sampling and scoring is a potent approach to precisely characterizing mutation processes in single-point mutagenesis protocols and crucial for computational antibody design.
Collapse
Affiliation(s)
- Rika Tandiana
- Computational MOdelling of NanosCalE and BioPhysical SysTems─CONCEPT Lab Istituto Italiano di Tecnologia (IIT), Via Melen-83, B Block, 16152 Genoa, Italy
| | - German P Barletta
- Computational MOdelling of NanosCalE and BioPhysical SysTems─CONCEPT Lab Istituto Italiano di Tecnologia (IIT), Via Melen-83, B Block, 16152 Genoa, Italy
- The Abdus Salam International Centre for Theoretical Physics─ICTP, Strada Costiera 11, 34151 Trieste, Italy
| | - Miguel Angel Soler
- Dipartimento di Scienze Matematiche, Informatiche e Fisiche, Universita' di Udine, Via delle Scienze 206, 33100 Udine, Italy
| | - Sara Fortuna
- Computational MOdelling of NanosCalE and BioPhysical SysTems─CONCEPT Lab Istituto Italiano di Tecnologia (IIT), Via Melen-83, B Block, 16152 Genoa, Italy
| | - Walter Rocchia
- Computational MOdelling of NanosCalE and BioPhysical SysTems─CONCEPT Lab Istituto Italiano di Tecnologia (IIT), Via Melen-83, B Block, 16152 Genoa, Italy
| |
Collapse
|
39
|
Karnchanapandh K, Sanachai K, Poo-Arporn RP, Rungrotmongkol T. Enhancing bezlotoxumab binding to C. difficile toxin B2: insights from computational simulations and mutational analyses for antibody design. J Biomol Struct Dyn 2024:1-11. [PMID: 38511411 DOI: 10.1080/07391102.2024.2329785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
Clostridioides difficile infection (CDI) is a significant concern caused by widespread antibiotic use, resulting in diarrhea and inflammation from the gram-positive anaerobic bacterium C. difficile. Although bezlotoxumab (Bez), a monoclonal antibody (mAb), was developed to address CDI recurrences, the recurrence rate remains high, partly due to reduced neutralization efficiency against toxin B2. In this study, we aimed to enhance the binding of Bez to C. difficile toxin B2 by combining computational simulations and mutational analyses. We identified specific mutations in Bez, including S28R, S31W/K, Y32R, S56W and G103D/S in the heavy chain (Hc), and S32F/H/R/W/Y in the light chain (Lc), which significantly improved binding to toxin B2 and formed critical protein-protein interactions. Through molecular dynamics simulations, several single mutations, such as HcS28R, LcS32H, LcS32R, LcS32W and LcS32Y, exhibited superior binding affinities to toxin B2 compared to Bez wild-type (WT), primarily attributed to Coulombic interactions. Combining the HcS28R mutation with four different mutations at residue LcS32 led to even greater binding affinities in double mutants (MTs), particularly HcS28R/LcS32H, HcS28R/LcS32R and HcS28R/LcS32Y, reinforcing protein-protein binding. Analysis of per-residue decomposition free energy highlighted key residues contributing significantly to enhanced binding interactions, emphasizing the role of electrostatic interactions. These findings offer insights into rational Bez MT design for improved toxin B2 binding, providing a foundation for developing more effective antibodies to neutralize toxin B2 and combat-related infections.
Collapse
Affiliation(s)
- Kun Karnchanapandh
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Kamonpan Sanachai
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| | - Rungtiva P Poo-Arporn
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Thanyada Rungrotmongkol
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Biocatalyst and Sustainable Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
40
|
Kasap C, Izgutdina A, Patiño-Escobar B, Kang A, Chilakapati N, Akagi N, Johnson H, Rashid T, Werner J, Barpanda A, Geng H, Lin YHT, Rampersaud S, Gil-Alós D, Sobh A, Dupéré-Richer D, Wicaksono G, Kawehi Kelii K, Dalal R, Ramos E, Vijayanarayanan A, Salangsang F, Phojanakong P, Serrano JAC, Zakraoui O, Tariq I, Steri V, Shanmugam M, Boise LH, Kortemme T, Stieglitz E, Licht JD, Karlon WJ, Barwick BG, Wiita AP. Targeting high-risk multiple myeloma genotypes with optimized anti-CD70 CAR-T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.24.581875. [PMID: 38463958 PMCID: PMC10925123 DOI: 10.1101/2024.02.24.581875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Despite the success of BCMA-targeting CAR-Ts in multiple myeloma, patients with high-risk cytogenetic features still relapse most quickly and are in urgent need of additional therapeutic options. Here, we identify CD70, widely recognized as a favorable immunotherapy target in other cancers, as a specifically upregulated cell surface antigen in high risk myeloma tumors. We use a structure-guided design to define a CD27-based anti-CD70 CAR-T design that outperforms all tested scFv-based CARs, leading to >80-fold improved CAR-T expansion in vivo. Epigenetic analysis via machine learning predicts key transcription factors and transcriptional networks driving CD70 upregulation in high risk myeloma. Dual-targeting CAR-Ts against either CD70 or BCMA demonstrate a potential strategy to avoid antigen escape-mediated resistance. Together, these findings support the promise of targeting CD70 with optimized CAR-Ts in myeloma as well as future clinical translation of this approach.
Collapse
Affiliation(s)
- Corynn Kasap
- Dept. of Medicine, Division of Hematology/Oncology, University of California, San Francisco, CA
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Adila Izgutdina
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | | | - Amrik Kang
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
- Medical Scientist Training Program, University of California, San Francisco, CA
| | - Nikhil Chilakapati
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Naomi Akagi
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Haley Johnson
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Tasfia Rashid
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Juwita Werner
- Dept. of Pediatrics, Division of Oncology, University of California, San Francisco, CA
| | - Abhilash Barpanda
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Huimin Geng
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Yu-Hsiu T. Lin
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Sham Rampersaud
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Daniel Gil-Alós
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
- Dept of Hematology, Hospital 12 de Octubre, Madrid, Spain
| | - Amin Sobh
- University of Florida Health Cancer Center, The University of Florida Cancer and Genetics Research Complex, Gainesville, Florida
- Division of Hematology/Oncology, The University of Florida College of Medicine, Gainesville, Florida
| | - Daphné Dupéré-Richer
- University of Florida Health Cancer Center, The University of Florida Cancer and Genetics Research Complex, Gainesville, Florida
- Division of Hematology/Oncology, The University of Florida College of Medicine, Gainesville, Florida
| | - Gianina Wicaksono
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - K.M. Kawehi Kelii
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Radhika Dalal
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA
| | - Emilio Ramos
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | | | - Fernando Salangsang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Paul Phojanakong
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | | | - Ons Zakraoui
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Isa Tariq
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
| | - Elliot Stieglitz
- Dept. of Pediatrics, Division of Oncology, University of California, San Francisco, CA
| | - Jonathan D. Licht
- University of Florida Health Cancer Center, The University of Florida Cancer and Genetics Research Complex, Gainesville, Florida
- Division of Hematology/Oncology, The University of Florida College of Medicine, Gainesville, Florida
| | - William J. Karlon
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA
| | - Arun P. Wiita
- Dept. of Laboratory Medicine, University of California, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA
| |
Collapse
|
41
|
Li ZL, Sun CQ, Qing ZL, Li ZM, Liu HL. Engineering the thermal stability of a polyphosphate kinase by ancestral sequence reconstruction to expand the temperature boundary for an industrially applicable ATP regeneration system. Appl Environ Microbiol 2024; 90:e0157423. [PMID: 38236018 PMCID: PMC10880597 DOI: 10.1128/aem.01574-23] [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: 09/09/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024] Open
Abstract
ATP-dependent energy-consuming enzymatic reactions are widely used in cell-free biocatalysis. However, the direct addition of large amounts of expensive ATP can greatly increase cost, and enzymatic production is often difficult to achieve as a result. Although a polyphosphate kinase (PPK)-polyphosphate-based ATP regeneration system has the potential to solve this challenge, the generally poor thermal stability of PPKs limits the widespread use of this method. In this paper, we evaluated the thermal stability of a PPK from Sulfurovum lithotrophicum (SlPPK2). After directed evolution and computation-supported design, we found that SlPPK2 is very recalcitrant and cannot acquire beneficial mutations. Inspired by the usually outstanding stability of ancestral enzymes, we reconstructed the ancestral sequence of the PPK family and used it as a guide to construct three heat-stable variants of SlPPK2, of which the L35F/T144S variant has a half-life of more than 14 h at 60°C. Molecular dynamics simulations were performed on all enzymes to analyze the reasons for the increased thermal stability. The results showed that mutations at these two positions act synergistically from the interior and surface of the protein, leading to a more compact structure. Finally, the robustness of the L35F/T144S variant was verified in the synthesis of nucleotides at high temperature. In practice, the use of this high-temperature ATP regeneration system can effectively avoid byproduct accumulation. Our work extends the temperature boundary of ATP regeneration and has great potential for industrial applications.IMPORTANCEATP regeneration is an important basic applied study in the field of cell-free biocatalysis. Polyphosphate kinase (PPK) is an enzyme tool widely used for energy regeneration during enzymatic reactions. However, the thermal stability of the PPKs reported to date that can efficiently regenerate ATP is usually poor, which greatly limits their application. In this study, the thermal stability of a difficult-to-engineer PPK from Sulfurovum lithotrophicum was improved, guided by an ancestral sequence reconstruction strategy. The optimal variant has a 4.5-fold longer half-life at 60°C than the wild-type enzyme, thus enabling the extension of the temperature boundary for ATP regeneration. The ability of this variant to regenerate ATP was well demonstrated during high-temperature enzymatic production of nucleotides.
Collapse
Affiliation(s)
- Zong-Lin Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Chuan-Qi Sun
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhou-Lei Qing
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Zhi-Min Li
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology, Shanghai, China
| | - Hong-Lai Liu
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
42
|
Kim DN, McNaughton AD, Kumar N. Leveraging Artificial Intelligence to Expedite Antibody Design and Enhance Antibody-Antigen Interactions. Bioengineering (Basel) 2024; 11:185. [PMID: 38391671 PMCID: PMC10886287 DOI: 10.3390/bioengineering11020185] [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: 12/30/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
This perspective sheds light on the transformative impact of recent computational advancements in the field of protein therapeutics, with a particular focus on the design and development of antibodies. Cutting-edge computational methods have revolutionized our understanding of protein-protein interactions (PPIs), enhancing the efficacy of protein therapeutics in preclinical and clinical settings. Central to these advancements is the application of machine learning and deep learning, which offers unprecedented insights into the intricate mechanisms of PPIs and facilitates precise control over protein functions. Despite these advancements, the complex structural nuances of antibodies pose ongoing challenges in their design and optimization. Our review provides a comprehensive exploration of the latest deep learning approaches, including language models and diffusion techniques, and their role in surmounting these challenges. We also present a critical analysis of these methods, offering insights to drive further progress in this rapidly evolving field. The paper includes practical recommendations for the application of these computational techniques, supplemented with independent benchmark studies. These studies focus on key performance metrics such as accuracy and the ease of program execution, providing a valuable resource for researchers engaged in antibody design and development. Through this detailed perspective, we aim to contribute to the advancement of antibody design, equipping researchers with the tools and knowledge to navigate the complexities of this field.
Collapse
Affiliation(s)
| | | | - Neeraj Kumar
- Pacific Northwest National Laboratory, 902 Battelle Blvd., Richland, WA 99352, USA; (D.N.K.); (A.D.M.)
| |
Collapse
|
43
|
Chamness LM, Kuntz CP, McKee AG, Penn WD, Hemmerich CM, Rusch DB, Woods H, Dyotima, Meiler J, Schlebach JP. Divergent Folding-Mediated Epistasis Among Unstable Membrane Protein Variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.25.554866. [PMID: 37662415 PMCID: PMC10473758 DOI: 10.1101/2023.08.25.554866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Many membrane proteins are prone to misfolding, which compromises their functional expression at the plasma membrane. This is particularly true for the mammalian gonadotropin-releasing hormone receptor GPCRs (GnRHR). We recently demonstrated that evolutionary GnRHR modifications appear to have coincided with adaptive changes in cotranslational folding efficiency. Though protein stability is known to shape evolution, it is unclear how cotranslational folding constraints modulate the synergistic, epistatic interactions between mutations. We therefore compared the pairwise interactions formed by mutations that disrupt the membrane topology (V276T) or tertiary structure (W107A) of GnRHR. Using deep mutational scanning, we evaluated how the plasma membrane expression of these variants is modified by hundreds of secondary mutations. An analysis of 251 mutants in three genetic backgrounds reveals that V276T and W107A form distinct epistatic interactions that depend on both the severity and the mechanism of destabilization. V276T forms predominantly negative epistatic interactions with destabilizing mutations in soluble loops. In contrast, W107A forms positive interactions with mutations in both loops and transmembrane domains that reflect the diminishing impacts of the destabilizing mutations in variants that are already unstable. These findings reveal how epistasis is remodeled by conformational defects in membrane proteins and in unstable proteins more generally.
Collapse
Affiliation(s)
- Laura M. Chamness
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | - Charles P. Kuntz
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Andrew G. McKee
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | - Wesley D. Penn
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | | | - Douglas B. Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, USA
| | - Hope Woods
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Dyotima
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Institute for Drug Discovery, Leipzig University, Leipzig, SAC, Germany
| | - Jonathan P. Schlebach
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
44
|
Avila-Herrera A, Kimbrel JA, Manuel Martí J, Thissen J, Saada EA, Weisenberger T, Arrildt KT, Segelke BW, Allen JE, Zemla A, Borucki MK. Differential laboratory passaging of SARS-CoV-2 viral stocks impacts the in vitro assessment of neutralizing antibodies. PLoS One 2024; 19:e0289198. [PMID: 38271318 PMCID: PMC10810540 DOI: 10.1371/journal.pone.0289198] [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: 07/12/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Viral populations in natural infections can have a high degree of sequence diversity, which can directly impact immune escape. However, antibody potency is often tested in vitro with a relatively clonal viral populations, such as laboratory virus or pseudotyped virus stocks, which may not accurately represent the genetic diversity of circulating viral genotypes. This can affect the validity of viral phenotype assays, such as antibody neutralization assays. To address this issue, we tested whether recombinant virus carrying SARS-CoV-2 spike (VSV-SARS-CoV-2-S) stocks could be made more genetically diverse by passage, and if a stock passaged under selective pressure was more capable of escaping monoclonal antibody (mAb) neutralization than unpassaged stock or than viral stock passaged without selective pressures. We passaged VSV-SARS-CoV-2-S four times concurrently in three cell lines and then six times with or without polyclonal antiserum selection pressure. All three of the monoclonal antibodies tested neutralized the viral population present in the unpassaged stock. The viral inoculum derived from serial passage without antiserum selection pressure was neutralized by two of the three mAbs. However, the viral inoculum derived from serial passage under antiserum selection pressure escaped neutralization by all three mAbs. Deep sequencing revealed the rapid acquisition of multiple mutations associated with antibody escape in the VSV-SARS-CoV-2-S that had been passaged in the presence of antiserum, including key mutations present in currently circulating Omicron subvariants. These data indicate that viral stock that was generated under polyclonal antiserum selection pressure better reflects the natural environment of the circulating virus and may yield more biologically relevant outcomes in phenotypic assays. Thus, mAb assessment assays that utilize a more genetically diverse, biologically relevant, virus stock may yield data that are relevant for prediction of mAb efficacy and for enhancing biosurveillance.
Collapse
Affiliation(s)
- Aram Avila-Herrera
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Jeffrey A. Kimbrel
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Jose Manuel Martí
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - James Thissen
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Edwin A. Saada
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Tracy Weisenberger
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Kathryn T. Arrildt
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Brent W. Segelke
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Jonathan E. Allen
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Adam Zemla
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Monica K. Borucki
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| |
Collapse
|
45
|
Hayes RL, Nixon CF, Marqusee S, Brooks CL. Selection pressures on evolution of ribonuclease H explored with rigorous free-energy-based design. Proc Natl Acad Sci U S A 2024; 121:e2312029121. [PMID: 38194446 PMCID: PMC10801872 DOI: 10.1073/pnas.2312029121] [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: 07/14/2023] [Accepted: 11/22/2023] [Indexed: 01/11/2024] Open
Abstract
Understanding natural protein evolution and designing novel proteins are motivating interest in development of high-throughput methods to explore large sequence spaces. In this work, we demonstrate the application of multisite λ dynamics (MSλD), a rigorous free energy simulation method, and chemical denaturation experiments to quantify evolutionary selection pressure from sequence-stability relationships and to address questions of design. This study examines a mesophilic phylogenetic clade of ribonuclease H (RNase H), furthering its extensive characterization in earlier studies, focusing on E. coli RNase H (ecRNH) and a more stable consensus sequence (AncCcons) differing at 15 positions. The stabilities of 32,768 chimeras between these two sequences were computed using the MSλD framework. The most stable and least stable chimeras were predicted and tested along with several other sequences, revealing a designed chimera with approximately the same stability increase as AncCcons, but requiring only half the mutations. Comparing the computed stabilities with experiment for 12 sequences reveals a Pearson correlation of 0.86 and root mean squared error of 1.18 kcal/mol, an unprecedented level of accuracy well beyond less rigorous computational design methods. We then quantified selection pressure using a simple evolutionary model in which sequences are selected according to the Boltzmann factor of their stability. Selection temperatures from 110 to 168 K are estimated in three ways by comparing experimental and computational results to evolutionary models. These estimates indicate selection pressure is high, which has implications for evolutionary dynamics and for the accuracy required for design, and suggests accurate high-throughput computational methods like MSλD may enable more effective protein design.
Collapse
Affiliation(s)
- Ryan L. Hayes
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA92697
- Department of Chemistry, University of Michigan, Ann Arbor, MI48109
| | - Charlotte F. Nixon
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
| | - Susan Marqusee
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA94720
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Charles L. Brooks
- Department of Chemistry, University of Michigan, Ann Arbor, MI48109
- Biophysics Program, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
46
|
Jarończyk M. Software for Predicting Binding Free Energy of Protein-Protein Complexes and Their Mutants. Methods Mol Biol 2024; 2780:139-147. [PMID: 38987468 DOI: 10.1007/978-1-0716-3985-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Protein-protein binding affinity prediction is important for understanding complex biochemical pathways and to uncover protein interaction networks. Quantitative estimation of the binding affinity changes caused by mutations can provide critical information for protein function annotation and genetic disease diagnoses. The binding free energies of protein-protein complexes can be predicted using several computational tools. This chapter is a summary of software developed for the prediction of binding free energies for protein-protein complexes and their mutants.
Collapse
|
47
|
Eccleston RC, Manko E, Campino S, Clark TG, Furnham N. A computational method for predicting the most likely evolutionary trajectories in the stepwise accumulation of resistance mutations. eLife 2023; 12:e84756. [PMID: 38132182 PMCID: PMC10807863 DOI: 10.7554/elife.84756] [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: 11/07/2022] [Accepted: 12/21/2023] [Indexed: 12/23/2023] Open
Abstract
Pathogen evolution of drug resistance often occurs in a stepwise manner via the accumulation of multiple mutations that in combination have a non-additive impact on fitness, a phenomenon known as epistasis. The evolution of resistance via the accumulation of point mutations in the DHFR genes of Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) has been studied extensively and multiple studies have shown epistatic interactions between these mutations determine the accessible evolutionary trajectories to highly resistant multiple mutations. Here, we simulated these evolutionary trajectories using a model of molecular evolution, parameterised using Rosetta Flex ddG predictions, where selection acts to reduce the target-drug binding affinity. We observe strong agreement with pathways determined using experimentally measured IC50 values of pyrimethamine binding, which suggests binding affinity is strongly predictive of resistance and epistasis in binding affinity strongly influences the order of fixation of resistance mutations. We also infer pathways directly from the frequency of mutations found in isolate data, and observe remarkable agreement with the most likely pathways predicted by our mechanistic model, as well as those determined experimentally. This suggests mutation frequency data can be used to intuitively infer evolutionary pathways, provided sufficient sampling of the population.
Collapse
Affiliation(s)
- Ruth Charlotte Eccleston
- Department of Infection Biology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Emilia Manko
- Department of Infection Biology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Susana Campino
- Department of Infection Biology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Taane G Clark
- Department of Infection Biology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Nicholas Furnham
- Department of Infection Biology, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| |
Collapse
|
48
|
Sun C, Liu YT, Kang YF, Xie C, Li SX, Lu YT, Zeng MS. Elucidation of the neutralizing antibody evasion of emergent SARS-CoV-2 Omicron sub-lineages using structural analysis. SCIENCE CHINA. LIFE SCIENCES 2023; 66:2935-2938. [PMID: 37673846 DOI: 10.1007/s11427-023-2393-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/09/2023] [Indexed: 09/08/2023]
Affiliation(s)
- Cong Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Yuan-Tao Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yin-Feng Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Chu Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Shu-Xin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yu-Tong Lu
- National Supercomputer Center in Guangzhou, School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Department of Experimental Research, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
49
|
Ricotti S, Garay AS, Etcheverrigaray M, Amadeo GI, De Groot AS, Martin W, Mufarrege EF. Development of IFNβ-1a versions with reduced immunogenicity and full in vitro biological activity for the treatment of multiple sclerosis. Clin Immunol 2023; 257:109831. [PMID: 37931868 DOI: 10.1016/j.clim.2023.109831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
IFNβ (recombinant interferon Beta) has been widely used for the treatment of Multiple sclerosis for the last four decades. Despite the human origin of the IFNβ sequence, IFNβ is immunogenic, and unwanted immune responses in IFNβ-treated patients may compromise its efficacy and safety in the clinic. In this study, we applied the DeFT (De-immunization of Functional Therapeutics) approach to producing functional, de-immunized versions of IFNβ-1a. Two de-immunized versions of IFNβ-1a were produced in CHO cells and designated as IFNβ-1a(VAR1) and IFNβ-1a(VAR2). First, the secondary and tertiary protein structures were analyzed by circular dichroism spectroscopy. Then, the variants were also tested for functionality. While IFNβ-1a(VAR2) showed similar in vitro antiviral activity to the original protein, IFNβ-1a(VAR1) exhibited 40% more biological potency. Finally, in vivo assays using HLA-DR transgenic mice revealed that the de-immunized variants showed a markedly reduced immunogenicity when compared to the originator.
Collapse
Affiliation(s)
- Sonia Ricotti
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Alberto Sergio Garay
- Laboratory of Molecular Modeling, FBCB (School of Biochemistry and Biological Sciences), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Marina Etcheverrigaray
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Gabriel Ignacio Amadeo
- Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI 02903, USA; Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States of America
| | | | - Eduardo Federico Mufarrege
- UNL, CONICET, FBCB (School of Biochemistry and Biological Sciences), CBL (Biotechnological Center of Litoral), Ciudad Universitaria, Ruta Nacional 168, Km 472.4, C.C. 242, Santa Fe S3000ZAA, Argentina.
| |
Collapse
|
50
|
Hernández González JE, de Araujo AS. Alchemical Calculation of Relative Free Energies for Charge-Changing Mutations at Protein-Protein Interfaces Considering Fixed and Variable Protonation States. J Chem Inf Model 2023; 63:6807-6822. [PMID: 37851531 DOI: 10.1021/acs.jcim.3c00972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
The calculation of relative free energies (ΔΔG) for charge-changing mutations at protein-protein interfaces through alchemical methods remains challenging due to variations in the system's net charge during charging steps, the possibility of mutated and contacting ionizable residues occurring in various protonation states, and undersampling issues. In this study, we present a set of strategies, collectively termed TIRST/TIRST-H+, to address some of these challenges. Our approaches combine thermodynamic integration (TI) with the prediction of pKa shifts to calculate ΔΔG values. Moreover, special sets of restraints are employed to keep the alchemically transformed molecules separated. The accuracy of the devised approaches was assessed on a large and diverse data set comprising 164 point mutations of charged residues (Asp, Glu, Lys, and Arg) to Ala at the protein-protein interfaces of complexes with known three-dimensional structures. Mean absolute and root-mean-square errors ranging from 1.38 to 1.66 and 1.89 to 2.44 kcal/mol, respectively, and Pearson correlation coefficients of ∼0.6 were obtained when testing the approaches on the selected data set using the GPU-TI module of Amber18 suite and the ff14SB force field. Furthermore, the inclusion of variable protonation states for the mutated acid residues improved the accuracy of the predicted ΔΔG values. Therefore, our results validate the use of TIRST/TIRST-H+ in prospective studies aimed at evaluating the impact of charge-changing mutations to Ala on the stability of protein-protein complexes.
Collapse
|