1
|
Ramon A, Ni M, Predeina O, Gaffey R, Kunz P, Onuoha S, Sormanni P. Prediction of protein biophysical traits from limited data: a case study on nanobody thermostability through NanoMelt. MAbs 2025; 17:2442750. [PMID: 39772905 PMCID: PMC11730357 DOI: 10.1080/19420862.2024.2442750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
In-silico prediction of protein biophysical traits is often hindered by the limited availability of experimental data and their heterogeneity. Training on limited data can lead to overfitting and poor generalizability to sequences distant from those in the training set. Additionally, inadequate use of scarce and disparate data can introduce biases during evaluation, leading to unreliable model performances being reported. Here, we present a comprehensive study exploring various approaches for protein fitness prediction from limited data, leveraging pre-trained embeddings, repeated stratified nested cross-validation, and ensemble learning to ensure an unbiased assessment of the performances. We applied our framework to introduce NanoMelt, a predictor of nanobody thermostability trained with a dataset of 640 measurements of apparent melting temperature, obtained by integrating data from the literature with 129 new measurements from this study. We find that an ensemble model stacking multiple regression using diverse sequence embeddings achieves state-of-the-art accuracy in predicting nanobody thermostability. We further demonstrate NanoMelt's potential to streamline nanobody development by guiding the selection of highly stable nanobodies. We make the curated dataset of nanobody thermostability freely available and NanoMelt accessible as a downloadable software and webserver.
Collapse
Affiliation(s)
- Aubin Ramon
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Mingyang Ni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Olga Predeina
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Rebecca Gaffey
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Patrick Kunz
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Pietro Sormanni
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
2
|
Lin W, Zheng Y, Zhang J, Zhou Y, Wang M, You S, Su R, Qi W. Enhanced catalytic activity of polyethylene terephthalate hydrolase by structure-guided loop-focused iterative mutagenesis strategy. JOURNAL OF HAZARDOUS MATERIALS 2025; 490:137837. [PMID: 40054191 DOI: 10.1016/j.jhazmat.2025.137837] [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: 01/03/2025] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 04/16/2025]
Abstract
The accumulation of polyethylene terephthalate (PET) waste has caused significant environmental pollution. Although biological depolymerization offers a promising solution, its efficiency remains constrained by the limited activity of PET-degrading enzymes. In this study, we designed a Structure-guided Loop-focused Iterative Mutagenesis (SLIM) strategy and rationally engineered the PET degradation enzyme ICCG for higher activity. The strategy was designed by demonstrating the critical role of the β8-α6 loop in type Ⅰ enzymes, which has currently not been reported. The best variant obtained, YITA (H183Y/L202I/I208T/T153A), exhibited 4.46-fold higher hydrolytic activity on amorphous PET at 72 °C compared to ICCG, outperforming other PET hydrolases, and exhibited superior degradation activity on real substrates such as cake containers and PET fibers. Conformational analysis revealed the key role of the remodeled β8-α6 loop in substrate binding and overall stability. Collectively, this study explores a promising approach to modifying PET hydrolase and lays a theoretical foundation for advancing bio-circular economy.
Collapse
Affiliation(s)
- Wei Lin
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China
| | - Yunxin Zheng
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China
| | - Jiaxing Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China
| | - Yu Zhou
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China
| | - Mengfan Wang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, PR China; Yuantian Biotechnology (Tianjin) Co., Ltd, PR China
| | - Shengping You
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China; Beijing Meihao Biotechnology Co., Ltd, PR China.
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China; State Key Laboratory of Chemical Engineering, Tianjin 300072, PR China; Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, PR China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, PR China; State Key Laboratory of Chemical Engineering, Tianjin 300072, PR China; Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, PR China
| |
Collapse
|
3
|
Hossain M, Dodda SR, Das S, Aikat K, Mukhopadhyay SS. Catalytic tunnel engineering of thermostable endoglucanase of GH7 family (W356C) from Aspergillus fumigatus gains catalytic rate. Enzyme Microb Technol 2025; 187:110632. [PMID: 40139016 DOI: 10.1016/j.enzmictec.2025.110632] [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: 12/10/2024] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
Tunnel engineering targets the access tunnels in enzymes, which is crucial for substrate binding and product release. Modifying the tunnels can lead to better biomass-degrading abilities of the lignocellulolytic enzymes. In this report, we have engineered the thermostable GH7 family endoglucanase from Aspergillus fumigatus (AfEgl7). The residues in the open tunnel having the highest bottleneck radius are mutated. Mutations are created (T229F, W356C) in the non-conserved region. The mutant W356C showed a 2-fold increase in product release rate (Vmax = 375.8 µM/min) and 2.5-fold higher catalytic activity (Kcat = 75.1 min-1) compared to wild-type (Vmax= 232 µM/min; Kcat = 30.9 min-1) using CM cellulose as substrate. The mutant T229F lost both catalytic activity and thermostability. Molecular dynamic simulations and docking studies of W356C revealed a change in structure near the product exit region, which may facilitate faster product release and account for the increased catalytic efficiency of the mutant. This study showed how redesigning the access pathways can be a promising strategy for protein engineering and de novo protein design by tailoring the open tunnel geometry to a ligand-specific manner.
Collapse
Affiliation(s)
- Musaddique Hossain
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Subba Reddy Dodda
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Shalini Das
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Kaustav Aikat
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India
| | - Sudit S Mukhopadhyay
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209, India.
| |
Collapse
|
4
|
Su T, Xia Y. A quantitative comparison of the deleteriousness of missense and nonsense mutations using the structurally resolved human protein interactome. Protein Sci 2025; 34:e70155. [PMID: 40384578 PMCID: PMC12086521 DOI: 10.1002/pro.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
The complex genotype-to-phenotype relationships in Mendelian diseases can be elucidated by mutation-induced disturbances to the networks of molecular interactions (interactomes) in human cells. Missense and nonsense mutations cause distinct perturbations within the human protein interactome, leading to functional and phenotypic effects with varying degrees of severity. Here, we structurally resolve the human protein interactome at atomic-level resolutions and perform structural and thermodynamic calculations to assess the biophysical implications of these mutations. We focus on a specific type of missense mutation, known as "quasi-null" mutations, which destabilize proteins and cause similar functional consequences (node removal) to nonsense mutations. We propose a "fold difference" quantification of deleteriousness, which measures the ratio between the fractions of node-removal mutations in datasets of Mendelian disease-causing and non-pathogenic mutations. We estimate the fold differences of node-removal mutations to range from 3 (for quasi-null mutations with folding ΔΔG ≥2 kcal/mol) to 20 (for nonsense mutations). We observe a strong positive correlation between biophysical destabilization and phenotypic deleteriousness, demonstrating that the deleteriousness of quasi-null mutations spans a continuous spectrum, with nonsense mutations at the extreme (highly deleterious) end. Our findings substantiate the disparity in phenotypic severity between missense and nonsense mutations and suggest that mutation-induced protein destabilization is indicative of the phenotypic outcomes of missense mutations. Our analyses of node-removal mutations allow for the potential identification of proteins whose removal or destabilization lead to harmful phenotypes, enabling the development of targeted therapeutic approaches, and enhancing comprehension of the intricate mechanisms governing genotype-to-phenotype relationships in clinically relevant diseases.
Collapse
Affiliation(s)
- Ting‐Yi Su
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
| | - Yu Xia
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
- Department of BioengineeringMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
5
|
Wei Y, Li F, Zheng Y, Liang Y, Du Y, Yu H. Strengthening core-region hydrogen-bond networks and rigidifying surface loop to enhance thermostability of an (R)-selective transaminase converting chiral hydroxyl amines. J Biotechnol 2025; 402:39-50. [PMID: 40058651 DOI: 10.1016/j.jbiotec.2025.03.006] [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: 12/06/2024] [Revised: 02/16/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Transaminases have important applications in the synthesis of drug intermediates such as chiral amines. However, natural transaminases exhibit suboptimal thermal stability, limiting their further applications. Building upon an Rhodobacter sp.-derived (R)-selective transaminase (RbTA), we report a dual-region coupling engineering approach to improve thermostability of RbTA by strengthening the core hydrogen-bond networks and rigidifying the flexible surface loop. Through single strategy, we identified 4 thermostability improved single mutations, among which I249Q demonstrated the most substantial improvement, achieving a 18-fold increase in half-life (t1/240) and a 11.2 ℃ increase in T5010. Then in strategic coupling, the synergistic effect of dual-region modification was observed in both thermal stability and activity enhancement, as mutant with the best high-temperature catalytic performance, R136P/F228Y, had its T5010 improved by 7.1℃ and exhibited a 4.2-fold increase in kcat/Km towards (R)-3-amino-1-butanol. Finally, R136P/F228Y achieved a 20.5 % improvement in conversion over WT in an analytical-scale synthesis in 72 h at a 5 ℃ elevated catalytic temperature. Molecular dynamics simulations demonstrated that the synergy of the formation of new hydrogen bonds and decrease in flexibility accounted for the thermostability improvements. This study provides guidance for enhancing thermostability of similar fold-type enzymes without impairing enzymatic activity in an efficient manner.
Collapse
Affiliation(s)
- Yuwen Wei
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China
| | - Fulong Li
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China
| | - Yukun Zheng
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China
| | - Youxiang Liang
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China
| | - Yan Du
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China
| | - Huimin Yu
- Department of Chemical Engineering, Tsinghua University, Beijing 100084, China; Key Laboratory of Industrial Biocatalysis (Tsinghua University), the Ministry of Education, Beijing 100084, China; State Key Laboratory of Green Biomanufacturing, Beijing, China; Beijing Key Laboratory of Recombinant Protein Synthetic Biomanufacturing, Beijing, China; Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
6
|
Gruca-Stryjak K, Maciak K, Winiewska-Szajewska M, Jurkiewicz A, Gora M, Kacprzak MM, Drgas O, Bialek-Proscinska A, Sobczynska-Tomaszewska A, Pluta KD, Jamsheer A, Markwitz W, Poznanski J, Burzynska B. A novel NEK1 variant disturbs the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel, causing lethal short-rib polydactyly syndrome. Bone 2025; 195:117471. [PMID: 40147672 DOI: 10.1016/j.bone.2025.117471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The NIMA-related kinase 1 (NEK1) gene belongs to the Never in Mitosis Gene A (NIMA) kinase family, a group whose members play essential roles in cell cycle regulation, specifically in cell division and ciliogenesis. Mutations in the NEK1 gene have been implicated in several diseases, including short-rib polydactyly syndrome (SRPS). SRPS is a bone growth disorder characterized by severe congenital anomalies. Here, we describe a family with a lethal form of SRPS due to a novel intronic variant in the NEK1 gene. Basing on whole-exome sequencing of fetuses with SRPS we identified a homozygous variant of the NEK1 gene at position c.3584-10T>A as the causative mutation. Bioinformatic methods and minigene splicing assays were then used to assess the harmfulness and functional impact of the variant. We found that the identified mutation leads to the synthesis of the NEK1 protein lacking 90C-terminal residues following the last coiled-coil region. Additional experiments were performed to identify proteins that interact with the C-terminal fragment of NEK1 absent in the mutated protein. We suggest that the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel is essential for the VDAC1 phosphorylation, the absence of which is likely to affect ciliogenesis.
Collapse
Affiliation(s)
- Karolina Gruca-Stryjak
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland; Centers of Medical Genetics GENESIS, Poznan, Poland
| | - Karolina Maciak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Aneta Jurkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | | - Krzysztof D Pluta
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Wieslaw Markwitz
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jaroslaw Poznanski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
7
|
Lan SC, Perng MD, Chang YY, Chen YF, Lan MY. Phenotypic and molecular characterization of a recurrent SPTAN1 mutation causing SPG91. Mol Biol Rep 2025; 52:476. [PMID: 40397273 DOI: 10.1007/s11033-025-10582-4] [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: 03/29/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Spectrins are ubiquitous cytoskeleton proteins found in all metazoan cells. αII-spectrin, encoded by SPTAN1, is the pivotal protein responsible for organization of the axonal cytoskeleton. Monoallelic SPTAN1 mutations cause various inherited neurological diseases, including spastic paraplegia 91 (SPG91), a type of hereditary spastic paraplegia (HSP). METHODS AND RESULTS We reported two patients with SPG91 caused by the SPTAN1 mutation c.55 C > T (p.Arg19Trp), who presented with lower limb spasticity and polyneuropathy. An analysis of the patients reported in the literature in addition to the present patients revealed that SPTAN1 p.Arg19Trp was specific for an HSP phenotype, with 35% of the combined patients with sensory‒motor polyneuropathy and 30% with cerebellar ataxia. In computational simulations, this variant was predicted to perturb the stability of αII/β spectrin heterotetramerization but did not destabilize the tetramerization domain of αII-spectrin. CONCLUSIONS Our findings on genotype‒phenotype correlations and genetic effects on molecular characteristics may provide important insights into the exploration of αII-spectrin-related neurological diseases.
Collapse
Affiliation(s)
- Shih-Chun Lan
- School of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ming-Der Perng
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Yee Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Center for Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ying-Fa Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Center for Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Min-Yu Lan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
- Center for Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
8
|
Tang W, Kim J, Lee RT, Maurer-Stroh S, Renia L, Tay MZ. SARS-CoV-2: lessons in virus mutation prediction and pandemic preparedness. Curr Opin Immunol 2025; 95:102560. [PMID: 40378522 DOI: 10.1016/j.coi.2025.102560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/19/2025]
Abstract
The COVID-19 pandemic has prompted an unprecedented global response. In particular, extraordinary efforts have been dedicated toward monitoring and predicting variant emergence due to its huge impact, particularly for vaccine escape. Broadly, we classify such methods into two categories: forward mutation prediction, where phenotypes are first observed and the responsible genotypes traced, and reverse mutation prediction, which starts with selected pathogen genetic profiles and characterizes their associated phenotypes. Reverse mutation prediction strategies have advantages in being able to sample a more complete evolutionary space since sequences that do not yet exist can be sampled. The rapid improvement in the maturity and scale of reverse mutation prediction strategies, such as deep mutational scanning, has led to significant amounts of data for machine learning, with concomitant improvement in the prediction results from computational tools. Such integrated prediction approaches are generalizable and offer significant opportunities for anticipating viral evolution and for pandemic preparedness.
Collapse
Affiliation(s)
- Weiyi Tang
- A*STAR Infectious Diseases Labs (AIDL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jenna Kim
- A*STAR Infectious Diseases Labs (AIDL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Raphael Tc Lee
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore; GISAID Global Data Science Initiative (GISAID), Munich, Germany
| | - Sebastian Maurer-Stroh
- A*STAR Infectious Diseases Labs (AIDL), Agency for Science, Technology and Research (A*STAR), Singapore; Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore; GISAID Global Data Science Initiative (GISAID), Munich, Germany; National Public Health Laboratory, Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Singapore; Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (AIDL), Agency for Science, Technology and Research (A*STAR), Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore
| | - Matthew Z Tay
- A*STAR Infectious Diseases Labs (AIDL), Agency for Science, Technology and Research (A*STAR), Singapore; Department of Biochemistry, National University of Singapore, Singapore.
| |
Collapse
|
9
|
Wang Y, Zhu Y, Shi X, Wang L. 3D-ΔΔG: A Dual-Channel Prediction Model for Protein-Protein Binding Affinity Changes Following Mutation Based on Protein 3D Structures. Proteins 2025. [PMID: 40375059 DOI: 10.1002/prot.26837] [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: 02/16/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/18/2025]
Abstract
Protein-protein interactions are crucial for cellular regulation, antigen-antibody interactions, and other vital processes within living organisms. However, mutations in amino acid residues have the potential to induce changes in protein-protein binding affinity (ΔΔG), which may contribute to the onset and progression of disease. Existing methods for predicting ΔΔG use either protein sequence information or structural data. Furthermore, some methods are only applicable to single-point mutation cases. To address these limitations, we introduce a ΔΔG predictor that can handle complex scenarios involving multipoint mutations. In this investigation, a dual-channel deep learning model three-dimensional (3D)-ΔΔG is introduced, which is designed to predict ΔΔG by combining mutation information from side chain sequences and 3D structures. The proposed model employs a pre-trained protein language model to encode the side-chain amino acid sequence. A graph attention network is deployed to handle the graph representation of proteins simultaneously. Finally, a dual-channel processing module is implemented to facilitate depth fusion and extraction of both sequence and structural features. The model effectively captures the intricate alterations occurring pre- and post-protein mutation by integrating both sequence and 3D structural information. Results on the single-point mutation data set demonstrate a substantial improvement compared to state-of-the-art models. More significantly, 3D-ΔΔG exhibits superior performance when evaluated on the mixed mutation data sets, SKEMPIv1 and SKEMPIv2. The high level of agreement between the computationally predicted ΔΔG values and the experimentally determined values illustrates the potential of the 3D-ΔΔG model as an effective pre-screening tool in protein design and engineering.
Collapse
Affiliation(s)
- Yuxiang Wang
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Yibo Zhu
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Xiumin Shi
- School of Information and Electronics, Beijing Institute of Technology, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Feng T, Huang H, Xu Y, Che X, Wang M, Tao X, Feng Y, Xue S. Multisite synergistic evolution of oleate hydratase via DCCM and the orthogonal location of distal sites. Int J Biol Macromol 2025; 312:144001. [PMID: 40379171 DOI: 10.1016/j.ijbiomac.2025.144001] [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: 03/04/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The evolution of enzymes through multisite combinations is often constrained by the dynamic correlations among residues, resulting in the challenge of achieving additive effects when combining single-site positive mutants. Nevertheless, the incorporation of distal-site residue mutants offers a promising avenue to unlock synergistic interactions, particularly in enzymes with channels that mediate substrate and product transport. While combinatorial mutagenesis typically requires exhaustive screening of extensive mutant libraries to identify superior variants. In this study, a multisite combination strategy using dynamic cross-correlation matrices (DCCM) iterative analysis, coupled with location orthogonal filtration (DCCM/OL) was established based on single-site mutants enhancing enzyme performance by engineering distal residues using the "Structure, SCANEER, and Sequence (3S)" approach. Thirteen beneficial single mutants of oleate hydratase from Staphylococcus aureus (SaOhy), which catalyzes the hydration of linoleic acid, were targeted using the "3S" approach. By employing the DCCM/OL strategy, the number of candidates in the combination mutation library for experimental screening were reduced from 60 to 5. And a triple-site mutant, SaOhy_L151V/I411L/V135A, was ultimately identified, which increased the catalytic efficiencies with linoleic acid and oleic acid by a 4- and 2.3-fold, respectively. The philosophy of multisite mutation based on distal residues using the DCCM/OL strategy effectively achieves an amplification of enzymatic activity with distinct substrates simultaneously. This study offers a promising strategy for the construction of a mutant smart library and multisite combinations to efficiently increase enzymatic performance.
Collapse
Affiliation(s)
- Ting Feng
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Haoxian Huang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Ying Xu
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Xinyu Che
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Mingdong Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Xiangyu Tao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Yanbin Feng
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China.
| | - Song Xue
- MOE Key Laboratory of Bio-Intelligent Manufacturing, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China.
| |
Collapse
|
11
|
Cui XC, Zheng Y, Liu Y, Yuchi Z, Yuan YJ. AI-driven de novo enzyme design: Strategies, applications, and future prospects. Biotechnol Adv 2025; 82:108603. [PMID: 40368118 DOI: 10.1016/j.biotechadv.2025.108603] [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: 02/02/2025] [Revised: 04/22/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Enzymes are indispensable for biological processes and diverse applications across industries. While top-down modification strategies, such as directed evolution, have achieved remarkable success in optimizing existing enzymes, bottom-up de novo enzyme design has emerged as a transformative approach for engineering novel enzymes with customized catalytic functions, independent of natural templates. Recent advancements in artificial intelligence (AI) and computational power have significantly accelerated this field, enabling breakthroughs in enzyme engineering. These technologies facilitate the rapid generation of enzyme structures and amino acid sequences optimized for specific functions, thereby enhancing design efficiency. They also support functional validation and activity optimization, improving the catalytic performance, stability, and robustness of de novo designed enzymes. This review highlights recent advancements in AI-driven de novo enzyme design, discusses strategies for validation and optimization, and examines the challenges and future prospects of integrating these technologies into enzyme development.
Collapse
Affiliation(s)
- Xi-Chen Cui
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 30072, PR China; Frontiers Science Center for Synthetic Biology(Ministry of Education), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300072, PR China
| | - Yan Zheng
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 30072, PR China; Frontiers Science Center for Synthetic Biology(Ministry of Education), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300072, PR China
| | - Ye Liu
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 30072, PR China; Frontiers Science Center for Synthetic Biology(Ministry of Education), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300072, PR China; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Zhiguang Yuchi
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 30072, PR China; Frontiers Science Center for Synthetic Biology(Ministry of Education), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300072, PR China; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| | - Ying-Jin Yuan
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 30072, PR China; Frontiers Science Center for Synthetic Biology(Ministry of Education), School of Synthetic Biology and Biomanufacturing, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
12
|
Banerjee A, Bogetti AT, Bahar I. Accurate identification and mechanistic evaluation of pathogenic missense variants with Rhapsody-2. Proc Natl Acad Sci U S A 2025; 122:e2418100122. [PMID: 40314982 PMCID: PMC12067267 DOI: 10.1073/pnas.2418100122] [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: 09/05/2024] [Accepted: 04/06/2025] [Indexed: 05/03/2025] Open
Abstract
Understanding the effects of missense mutations or single amino acid variants (SAVs) on protein function is crucial for elucidating the molecular basis of diseases/disorders and designing rational therapies. We introduce here Rhapsody-2, a machine learning tool for discriminating pathogenic and neutral SAVs, significantly expanding on a precursor limited by the availability of structural data. With the advent of AlphaFold2 as a powerful tool for structure prediction, Rhapsody-2 is trained on a significantly expanded dataset of 117,525 SAVs corresponding to 12,094 human proteins reported in the ClinVar database. Adopting a broad set of descriptors composed of sequence evolutionary, structural, dynamic, and energetics features in the training algorithm, Rhapsody-2 achieved an AUROC of 0.94 in 10-fold cross-validation when all SAVs of a particular test protein (mutant) were excluded from the training set. Benchmarking against a variety of testing datasets demonstrated the high performance of Rhapsody-2. While sequence evolutionary descriptors play a dominant role in pathogenicity prediction, those based on structural dynamics provide a mechanistic interpretation. Notably, residues involved in allosteric communication and those distinguished by pronounced fluctuations in the high-frequency modes of motion or subject to spatial constraints in soft modes usually give rise to pathogenicity when mutated. Overall, Rhapsody-2 provides an efficient and transparent tool for accurately predicting the pathogenicity of SAVs and unraveling the mechanistic basis of the observed behavior, thus advancing our understanding of genotype-to-phenotype relations.
Collapse
Affiliation(s)
- Anupam Banerjee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| | - Anthony T. Bogetti
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY11794
- Department of Biochemistry and Cell Biology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY11794
| |
Collapse
|
13
|
Zhang L, Wang E, Wu L, Zhang J, You S, Su R, Qi W. Rational Design of UvsX Recombinase Variants for Enhanced Performance in Recombinase Polymerase Amplification Applications. Biochemistry 2025; 64:2025-2038. [PMID: 40261914 DOI: 10.1021/acs.biochem.5c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Homologous recombination is a vital biological process for DNA repair, genomic stability, and genetic diversity, driven by the RecA/Rad51 recombinase family. However, as a T4 bacteriophage recombinase homologous to RecA/Rad51, UvsX has limited in vitro performance during recombinase polymerase amplification (RPA) due to ATP utilization and DNA affinity. In this study, UvsX was rationally engineered to enhance these properties through homology modeling, virtual saturation mutations, and consensus mutation strategies. Targeted mutagenesis produced UvsX variants (E198N, E198R, E198K, and K35G) with a 16 ± 4% to 39 ± 6% improvement in RPA activity, while the double mutant K35G/E198R showed an increase of up to 43 ± 4%. Structural analysis revealed that the K35G/E198R mutation enlarged ATP-binding pockets and increased the positive surface potential of DNA-binding sites, resulting in a 12 ± 4% improvement in ATP utilization and more ADP and less AMP generated, a 10 ± 2% enhancement in DNA interaction compared to the wild-type, and better inhibitor tolerance. These findings establish a foundation for the rational optimization of recombinases in nucleic acid amplification and promote their potential for industrial RPA applications.
Collapse
Affiliation(s)
- Lin Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Enjie Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Lvping Wu
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Jiaxing Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
| | - Shengping You
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
14
|
Nath VR, Krishnan H, Mishra S, Raghu P. Ca2+ binding to Esyt modulates membrane contact site density in Drosophila photoreceptors. J Cell Biol 2025; 224:e202407190. [PMID: 40042442 PMCID: PMC11893162 DOI: 10.1083/jcb.202407190] [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: 07/29/2024] [Revised: 12/09/2024] [Accepted: 01/29/2025] [Indexed: 03/12/2025] Open
Abstract
Membrane contact sites (MCS) between the plasma membrane (PM) and endoplasmic reticulum (ER) regulate Ca2+ influx. However, the mechanisms by which cells modulate ER-PM MCS density are not understood, and the role of Ca2+, if any, in regulating these is unknown. We report that in Drosophila photoreceptors, MCS density is regulated by the Ca2+ channels, TRP and TRPL. Regulation of MCS density by Ca2+ is mediated by Drosophila extended synaptotagmin (dEsyt), a protein localized to ER-PM MCS and previously shown to regulate MCS density. We find that the Ca2+-binding activity of dEsyt is required for its function in vivo. dEsytCaBM, a Ca2+ non-binding mutant of dEsyt is unable to modulate MCS structure. Further, reconstitution of dEsyt null photoreceptors with dEsytCaBM is unable to rescue ER-PM MCS density and other key phenotypes. Thus, our data supports a role for Ca2+ binding to dEsyt in regulating ER-PM MCS density in photoreceptors thus tuning signal transduction during light-activated Ca2+ influx.
Collapse
Affiliation(s)
- Vaisaly R. Nath
- National Centre for Biological Sciences-TIFR, Bangalore, India
- School of Biotechnology, Amrita University, Kollam, India
| | - Harini Krishnan
- National Centre for Biological Sciences-TIFR, Bangalore, India
| | - Shirish Mishra
- National Centre for Biological Sciences-TIFR, Bangalore, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR, Bangalore, India
| |
Collapse
|
15
|
Bergson S, Sarig O, Giladi M, Mohamad J, Mogezel-Salem M, Smorodinsky-Atias K, Sade O, Manori B, Assaf S, Malovitski K, Feller Y, Pavlovsky M, Hainzl S, Kocher T, Hummel JI, Eretz Kdosha N, Khair LG, Zauner R, Pinon Hofbauer J, Shalom-Feuerstein R, Wally V, Koller U, Samuelov L, Haitin Y, Ashery U, Rubinstein R, Sprecher E. HMCN1 variants aggravate epidermolysis bullosa simplex phenotype. J Exp Med 2025; 222:e20240827. [PMID: 39976600 PMCID: PMC11841684 DOI: 10.1084/jem.20240827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/16/2024] [Accepted: 01/08/2025] [Indexed: 02/23/2025] Open
Abstract
Epidermolysis bullosa simplex (EBS) refers to a heterogeneous group of inherited skin disorders characterized by blister formation within the basal cell layer. The disease is characterized by marked variations in phenotype severity, suggesting co-inheritance of genetic modifiers. We identified three deleterious variants in HMCN1 that co-segregated with a more severe phenotype in a group of 20 individuals with EBS caused by mutations in KRT14, encoding keratin 14 (K14). HMCN1 codes for hemicentin-1. Protein modeling, molecular dynamics simulations, and functional experiments showed that all three HMCN1 variants disrupt protein stability. Hemicentin-1 was found to be expressed in human skin above the BMZ. Using yeast-2-hybrid, co-immunoprecipitation, and proximity ligation assays, we found that hemicentin-1 binds K14. Three-dimensional skin equivalents grown from hemicentin-1-deficient cells were found to spontaneously develop subepidermal blisters, and HMCN1 downregulation was found to reduce keratin intermediate filament formation. In conclusion, hemicentin-1 binds K14 and contributes to BMZ stability, which explains the fact that deleterious HMCN1 variants co-segregate with a more severe phenotype in KRT14-associated EBS.
Collapse
Affiliation(s)
- Shir Bergson
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Sarig
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Moshe Giladi
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Internal Medicine D, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Janan Mohamad
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Mariana Mogezel-Salem
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Karina Smorodinsky-Atias
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Ofir Sade
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Bar Manori
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sari Assaf
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Kiril Malovitski
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yarden Feller
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Mor Pavlovsky
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Stefan Hainzl
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Thomas Kocher
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Julia I. Hummel
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Noy Eretz Kdosha
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Lubna Gazi Khair
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Roland Zauner
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Josefina Pinon Hofbauer
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ruby Shalom-Feuerstein
- The Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Verena Wally
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ulrich Koller
- Department of Dermatology and Allergology, EB House Austria, Research Program for Molecular Therapy of Genodermatoses, University Hospital of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Liat Samuelov
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoni Haitin
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Rotem Rubinstein
- Faculty of Life Sciences, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Eli Sprecher
- Division of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
Liu YA, Lee CC, Górecki K, Stiebritz MT, Duffin C, Solomon JB, Ribbe MW, Hu Y. Heterologous synthesis of a simplified nitrogenase analog in Escherichia coli. SCIENCE ADVANCES 2025; 11:eadw6785. [PMID: 40315313 PMCID: PMC12047441 DOI: 10.1126/sciadv.adw6785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
The heterologous synthesis of a nitrogen-fixing system in a non-diazotrophic organism is a long-sought-after goal because of the crucial importance of nitrogenase for agronomy, energy, and the environment. Here, we report the heterologous synthesis of a two-component nitrogenase analog from Azotobacter vinelandii, which consists of the reductase component (NifH) and the cofactor maturase (NifEN), in Escherichia coli. Metal, electron paramagnetic resonance, and activity analyses verify the cluster composition and functional competence of the heterologously expressed NifH and NifEN. Nuclear magnetic resonance, nanoscale secondary ion mass spectrometry, and growth experiments further illustrate the ability of the NifH/NifEN system to reduce N2 and incorporate the reduced N into the cellular mass. These results establish NifEN/NifH as a simplified nitrogenase analog that could be optimized and engineered to facilitate transgenic expression and biotechnological adaptations of this important metalloenzyme.
Collapse
Affiliation(s)
- Yiling A. Liu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
| | - Chi Chung Lee
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
| | - Kamil Górecki
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
| | - Martin T. Stiebritz
- Department of Biology, Friedrich-Alexander University Erlangen-Nuremberg, D-91052 Erlangen, Germany
| | - Calder Duffin
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
- Department of Chemistry, University of California, Irvine, CA 92697-2025, USA
| | - Joseph B. Solomon
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
- Department of Chemistry, University of California, Irvine, CA 92697-2025, USA
| | - Markus W. Ribbe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
- Department of Chemistry, University of California, Irvine, CA 92697-2025, USA
| | - Yilin Hu
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697-3900, USA
| |
Collapse
|
17
|
Ramadane-Morchadi L, Rotenberg N, Esteban-Sánchez A, Fortuno C, Gómez-Sanz A, Varga MJ, Chamberlin A, Richardson ME, Michailidou K, Pérez-Segura P, Spurdle AB, de la Hoya M. ACMG/AMP interpretation of BRCA1 missense variants: Structure-informed scores add evidence strength granularity to the PP3/BP4 computational evidence. Am J Hum Genet 2025; 112:993-1002. [PMID: 40233743 DOI: 10.1016/j.ajhg.2024.12.011] [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: 05/29/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 04/17/2025] Open
Abstract
Classification of missense variants is challenging. Lacking compelling clinical and/or functional data, ACMG/AMP lines of evidence are restricted to PM2 (rarity code applied at supporting level) and PP3/BP4 (computational evidence based mostly on multiple-sequence-alignment conservation tools). Currently, the ClinGen ENIGMA BRCA1/2 Variant Curation Expert Panel uses BayesDel to apply PP3/BP4 to missense variants located in the BRCA1 RING/BRCT domains. The ACMG/AMP framework does not refer explicitly to protein structure as a putative source of pathogenic/benign evidence. Here, we tested the value of incorporating structure-based evidence such as relative solvent accessibility (RSA), folding stability (ΔΔG), and/or AlphaMissense pathogenicity to the classification of BRCA1 missense variants. We used MAVE functional scores as proxies for pathogenicity/benignity. We computed RSA and FoldX5.0 ΔΔG predictions using as alternative input templates for either PDB files or AlphaFold2 models, and we retrieved pre-computed AlphaMissense and BayesDel scores. We calculated likelihood ratios toward pathogenicity/benignity provided by the tools (individually or combined). We performed a clinical validation of major findings using the large-scale BRIDGES case-control dataset. AlphaMissense outperforms ΔΔG and BayesDel, providing similar PP3/BP4 evidence strengths with lower rate of variants in the uninformative score range. AlphaMissense combined with ΔΔG increases evidence strength granularity. AlphaFold2 models perform well as input templates for ΔΔG predictions. Regardless of the tool, BP4 (but not PP3) is highly dependent on RSA, with benignity evidence provided only to variants targeting buried or partially buried residues (RSA ≤ 60%). Stratification by functional domain did not reveal major differences. In brief, structure-based analysis improves PP3/BP4 assessment, uncovering a relevant role for RSA.
Collapse
Affiliation(s)
- Lobna Ramadane-Morchadi
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Nitsan Rotenberg
- University of Queensland, Brisbane, QLD, Australia; Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Ada Esteban-Sánchez
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Cristina Fortuno
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Alicia Gómez-Sanz
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | | | | | | | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology & Genetics, 2371 Nicosia, Cyprus
| | - Pedro Pérez-Segura
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Amanda B Spurdle
- University of Queensland, Brisbane, QLD, Australia; Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain.
| |
Collapse
|
18
|
Zhang J, Xiong Y. PackPPI: An integrated framework for protein-protein complex side-chain packing and ΔΔG prediction based on diffusion model. Protein Sci 2025; 34:e70110. [PMID: 40260988 PMCID: PMC12012842 DOI: 10.1002/pro.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
Deep learning methods have played an increasingly pivotal role in advancing side-chain packing and mutation effect prediction (ΔΔG) for protein complexes. Although these two tasks are inherently closely related, they are typically treated separately in practice. Furthermore, the lack of effective post-processing in most approaches results in sub-optimal refinement of generated conformations, limiting the plausibility of the predicted conformations. In this study, we introduce an integrated framework, PackPPI, which employs a diffusion model and a proximal optimization algorithm to improve side-chain prediction for protein complexes while using learned representations to predict ΔΔG. The results demonstrate that PackPPI achieved the lowest atom RMSD (0.9822) on the CASP15 dataset. The proximal optimization algorithm effectively reduces spatial clashes between side-chain atoms while maintaining a low-energy landscape. Furthermore, PackPPI achieves state-of-the-art performance in predicting binding affinity changes induced by multi-point mutations on the SKEMPI v2.0 dataset. These findings underscore the potential of PackPPI as a robust and versatile computational tool for protein design and engineering. The implementation of PackPPI is available at https://github.com/Jackz915/PackPPI.
Collapse
Affiliation(s)
- Jingkai Zhang
- State Key Laboratory of BiocontrolSchool of Life Sciences, Sun Yat‐sen UniversityGuangzhouChina
| | - Yuanyan Xiong
- State Key Laboratory of BiocontrolSchool of Life Sciences, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
19
|
Zhao YF, Zhang Y, Peng YZ, Khurshid M, Herman RA, Zhu XL, Lv X, Li J, Zhao WG, Wang J, You S. Enzymolysis for effective grain processing: Computer-aided optimization of a 1,3-1,4-β-glucanase with improved thermostability and catalytic activity. Int J Biol Macromol 2025; 309:143038. [PMID: 40220841 DOI: 10.1016/j.ijbiomac.2025.143038] [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: 11/27/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
β-Glucanases, widely applied in grain processing, are commonly restricted for efficient industrial application due to the limited thermostability. In this study, a 1,3-1,4-β-glucanase (BisGlu16B_ΔC) was optimized for thermostability through a computer-aided design of energy optimization. Three variants (T40K, Q53L, and S311Y) were selected and generated a combined mutant T40K/Q53L/S311Y (M3). Comparing with the WT, M3 exhibited better thermostability (with t1/2 at 60 °C extend by 126 min), higher specific activity (1.24 folds; 69,700 vs. 56,200 U/mg), higher catalytic effciency (1.18 folds; 14,100 vs. 11,900 mL‧s-1‧mg-1), and improved protease resistance. For mechanism, more hydrogen bonds, salt bridges, and rigid secondary components in M3 led to an enhanced overall rigidity, boosting the thermostability. While enhanced long-range negative interactions affected some key residues in the catalytic channels, improving the catalytic efficiency. For application, M3 showed superiority with higher dry matter digestibility (1.49 folds; 80.3 % vs. 53.9 %) in simulated gastrointestinal system, together with more reduction of filtration time (1.55 folds; 22.2 % vs. 14.3 %) and viscosity (2.37 folds; 10.2 % vs. 4.3 %) during malting, comparing with the WT. Furthermore, the strongest synergistic effects were found between xylanase and M3, among all β-glucanases tested. All results verified M3 as an efficient β-glucanase for grain processing industry.
Collapse
Affiliation(s)
- Yi-Fan Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Ying Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Ying-Zhi Peng
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Marriam Khurshid
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Richard-Ansah Herman
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Xiao-Lu Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Xiang Lv
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jing Li
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Wei-Guo Zhao
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Jun Wang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China
| | - Shuai You
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu 212100, China.
| |
Collapse
|
20
|
Küng C, Protsenko O, Vanella R, Nash MA. Deep mutational scanning reveals a de novo disulfide bond and combinatorial mutations for engineering thermostable myoglobin. Protein Sci 2025; 34:e70112. [PMID: 40247745 PMCID: PMC12006728 DOI: 10.1002/pro.70112] [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: 11/02/2024] [Revised: 02/18/2025] [Accepted: 03/13/2025] [Indexed: 04/19/2025]
Abstract
Engineering protein stability is a critical challenge in biotechnology. Here, we used massively parallel deep mutational scanning (DMS) to comprehensively explore the mutational stability landscape of human myoglobin (hMb) and identify key mutations that enhance stability. Our DMS approach involved screening over 10,000 hMb variants by yeast surface display, single-cell sorting, and high-throughput DNA sequencing. We show how surface display levels serve as a proxy for thermostability of soluble hMb variants and report strong correlations between DMS-derived display levels and top-performing machine learning stability prediction algorithms. This approach led to the discovery of a variant with a de novo disulfide bond between residues R32C and C111, which increased thermostability by >12°C compared with wild-type hMb. By combining single stabilizing mutations with R32C, we engineered combinatorial variants that exhibited predominantly additive effects on stability with minimal epistasis. The most stable combinatorial variant exhibited a denaturation temperature exceeding 89°C, representing a >17°C improvement over wild-type hMb. Our findings demonstrate the capabilities in DMS-assisted combinatorial protein engineering to guide the discovery of thermostable variants and highlight the potential of massively parallel mutational analysis for the development of proteins for industrial and biomedical applications.
Collapse
Affiliation(s)
- Christoph Küng
- Department of Chemistry, Institute of Physical ChemistryUniversity of BaselBaselSwitzerland
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
| | - Olena Protsenko
- Department of Chemistry, Institute of Physical ChemistryUniversity of BaselBaselSwitzerland
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
| | - Rosario Vanella
- Department of Chemistry, Institute of Physical ChemistryUniversity of BaselBaselSwitzerland
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
| | - Michael A. Nash
- Department of Chemistry, Institute of Physical ChemistryUniversity of BaselBaselSwitzerland
- Department of Biosystems Science and EngineeringETH ZurichBaselSwitzerland
| |
Collapse
|
21
|
Rossi I, Barducci G, Sanavia T, Turina P, Capriotti E, Fariselli P. Mass balance approximation of unfolding boosts potential-based protein stability predictions. Protein Sci 2025; 34:e70134. [PMID: 40277391 PMCID: PMC12023412 DOI: 10.1002/pro.70134] [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: 12/05/2024] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Predicting protein stability changes upon single-point mutations is crucial in computational biology, with applications in drug design, enzyme engineering, and understanding disease mechanisms. While deep-learning approaches have emerged, many remain inaccessible for routine use. In contrast, potential-like methods, including deep-learning-based ones, are faster, user-friendly, and effective in estimating stability changes. However, most of them approximate Gibbs free-energy differences without accounting for the free-energy changes of the unfolded state, violating mass balance and potentially reducing accuracy. Here, we show that incorporating mass balance as a first approximation of the unfolded state significantly improves potential-like methods. While many machine-learning models implicitly or explicitly use mass balance, our findings suggest that a more accurate unfolded-state representation could further enhance stability change predictions.
Collapse
Affiliation(s)
- Ivan Rossi
- Department of Medical SciencesUniversity of TorinoTorinoItaly
| | - Guido Barducci
- Department of Medical SciencesUniversity of TorinoTorinoItaly
| | - Tiziana Sanavia
- Department of Medical SciencesUniversity of TorinoTorinoItaly
| | - Paola Turina
- Department of Pharmacy and Biotechnology (FaBiT)University of BolognaBolognaItaly
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology (FaBiT)University of BolognaBolognaItaly
- Computational Genomics Platform, IRCCS University Hospital of BolognaBolognaItaly
| | - Piero Fariselli
- Department of Medical SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
22
|
Varga MJ, Richardson ME, Chamberlin A. Structural biology in variant interpretation: Perspectives and practices from two studies. Am J Hum Genet 2025; 112:984-992. [PMID: 40233741 DOI: 10.1016/j.ajhg.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Structural biology offers a powerful lens through which to assess genetic variants by providing insights into their impact on clinically relevant protein structure and function. Due to the availability of new, user-friendly, web-based tools, structural analyses by wider audiences have become more mainstream. These new tools, including AlphaMissense and AlphaFold, have recently been in the limelight due to their initial success and projected future promise; however, the intricacies and limitations of using these tools still need to be disseminated to the more general audience that is likely to use them in variant analysis. Here, we expound on frameworks applying structural biology to variant interpretation by examining two accompanying articles. To this end, we explore the nuances of choosing the correct protein model, compare and contrast various structural approaches, and highlight both the advantages and limitations of employing structural biology in variant interpretation. Using two articles published in this issue of The American Journal of Human Genetics as a baseline, we focus on case studies in TP53 and BRCA1 to illuminate gene-specific differences in the applications of structural information, which illustrate the complexities inherent in this field. Additionally, we discuss the implications of recent advancements, such as AlphaFold, and provide practical guidance for researchers navigating variant interpretation using structural biology.
Collapse
|
23
|
Rotenberg N, Fortuno C, Varga MJ, Chamberlin AC, Ramadane-Morchadi L, Feng BJ, de la Hoya M, Richardson ME, Spurdle AB. Integration of protein stability and AlphaMissense scores improves bioinformatic impact prediction for p53 missense and in-frame amino acid deletion variants. Am J Hum Genet 2025; 112:1003-1014. [PMID: 40233742 DOI: 10.1016/j.ajhg.2025.01.012] [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: 05/28/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 04/17/2025] Open
Abstract
The clinical classification of germline missense variants and single-amino-acid deletions is challenging. The BayesDel and Align-GVGD bioinformatic prediction tools currently used for ClinGen TP53 variant curation expert panel (VCEP) classification do not directly capture changes in protein folding stability, measured using computed destabilization energies (ΔΔG scores). The AlphaMissense tool recently developed by Google DeepMind to predict pathogenicity for all human proteome missense variants is trained in part using AlphaFold2 architecture. Our study investigated whether protein folding stability and/or AlphaMissense scores could improve impact prediction for p53 missense and single-amino-acid deletion variants. ΔΔG scores were calculated for missense variants using FoldX and for single-amino-acid deletions using an AlphaFold2/RosettaRelax protocol. Residue surface exposure was categorized using relative solvent accessibility (RSA) measures. The predictive values of ΔΔG scores, AlphaMissense, BayesDel, and Align-GVGD were examined using Boruta and binary logistic regression based on functionally defined reference sets. The likelihood ratio (LR) toward pathogenicity was estimated and used to refine optimal categories for predicting variant pathogenicity for different RSA values. We showed that current VCEP predictive approaches for missense variants were improved by integrating ΔΔG scores ≥2.5 kcal/mol for partially buried and buried residues, but better performance was achieved using AlphaMissense with ΔΔG and RSA. For deletion variants, ΔΔG scores ≥4.8 Rosetta energy unit (REU) in buried residues outperformed currently used predictive approaches. Future TP53 VCEP specifications for p53 missense impact prediction may consider AlphaMissense, ΔΔG score, and RSA combined for substitution variants and ΔΔG score alone for deletion variants.
Collapse
Affiliation(s)
- Nitsan Rotenberg
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia
| | - Cristina Fortuno
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | | | | | - Lobna Ramadane-Morchadi
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Bing-Jian Feng
- University of Utah Department of Dermatology, Salt Lake City, UT, USA; University of Utah Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | | | - Amanda B Spurdle
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
24
|
Lu C, Fang R, Tian S, Hu M, Wang J, Ding J. Integrating protein contact networks for the engineering of thermostable lipase A. Int J Biol Macromol 2025; 306:141725. [PMID: 40044005 DOI: 10.1016/j.ijbiomac.2025.141725] [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: 01/02/2025] [Revised: 03/01/2025] [Accepted: 03/02/2025] [Indexed: 05/03/2025]
Abstract
In the field of industrial biocatalysis, the rapid advancement of enzyme functional evolution necessitates new theories and computational methods to achieve target functions with fewer iterations. This study identified key residues affecting enzyme stability by constructing the protein contact network (PCN) of Lipase A. Comparing the PCNs of the wild-type (WT) and the 6B variant revealed that changes in residue interactions and node properties (e.g., degree and betweenness centrality (BC)) positively impacted stability. Using thresholds for degree and BC, 25 candidate sites were screened, and 11 out of 18 single-point mutation designs improved thermal stability. Mutations were divided into three groups (M1, M2, M3) based on network communities and contributions, followed by iterative combinations. M1, containing five mutations distributed across four communities, increased the melting temperature (Tm) by 14.61 °C, close to the predicted 13.97 °C, demonstrating a linear additive effect. In M2, three new mutations resulted in a non-linear additive effect, with a ΔTm of 17.58 °C (Expected ΔTm = 18.93 °C). In contrast, the three new mutations in M3 destabilized the enzyme (Observed ΔTm = 15.94 °C vs Expected ΔTm = 19.92 °C). Molecular dynamics simulations showed that polar edge nodes enhanced network connectivity, while proline mutations rigidified flexible regions, improving stability. Conversely, M3 mutations disrupted α-helix stability by increasing the dihedral angle fluctuations of residue Y161, might to a stability-activity trade-off. The PCN provides valuable insights for developing efficient and precise design strategies.
Collapse
Affiliation(s)
- Cheng Lu
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China
| | - Ruijie Fang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China
| | - Siyuan Tian
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China
| | - Mingzhu Hu
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China
| | - Jianan Wang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China
| | - Jian Ding
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, China.
| |
Collapse
|
25
|
Yang M, Wang J, Zhou Z, Li W, Verkhivker G, Xiao F, Hu G. Machine Learning and Structural Dynamics-Based Approach to Reveal Molecular Mechanism of PTEN Missense Mutations Shared by Cancer and Autism Spectrum Disorder. J Chem Inf Model 2025; 65:4173-4188. [PMID: 40228162 DOI: 10.1021/acs.jcim.5c00134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Missense mutations in oncogenic proteins that are concurrently associated with neurodevelopmental disorders have garnered significant attention. Phosphatase and tensin homologue (PTEN) serves as a paradigmatic model for mapping its mutational landscape and identifying genotypic predictors of distinct phenotypic outcomes, including cancer and autism spectrum disorder (ASD). Despite extensive research into the genotype-phenotype correlations of PTEN mutations, the mechanisms underlying the dual association of specific PTEN mutations with both cancer and ASD (PTEN-cancer/ASD mutations) remain elusive. This study introduces an integrative approach that combines machine learning (ML) with structural dynamics to elucidate the molecular effects of PTEN-cancer/ASD mutations. Analysis of biophysical and network-biology-based signatures reveals a complex energetic and functional landscape. Subsequently, an ML model and corresponding integrated score were developed to classify and predict PTEN-cancer/ASD mutations, underscoring the significance of protein dynamics in predicting cellular phenotypes. Further molecular dynamics simulations demonstrated that PTEN-cancer/ASD mutations induce dynamic alterations characterized by open conformational changes restricted to the P loop and coupled with interdomain allosteric regulation. This research aims to enhance the genotypic and phenotypic understanding of PTEN-cancer/ASD mutations through an interpretable ML model integrated with structural dynamics analysis. By identifying shared mechanisms between cancer and ASD, the findings pave the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Miao Yang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jingran Wang
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Ziyun Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Wentian Li
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Gennady Verkhivker
- Keck Center for Science and Engineering, Schmid College of Science and Technology, Chapman University, One University Drive, Orange, California 92866, United States
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California 92618, United States
| | - Fei Xiao
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Guang Hu
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-infective Medicine, Department of Bioinformatics and Computational Biology, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, Soochow University, Suzhou 215123, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| |
Collapse
|
26
|
Drotarova M, Asselta R, Caccia S, Skornova I, Zolkova J, Kolkova Z, Loderer D, Podusel V, Stasko J, Simurda T. A novel pathogenic variant in the fibrinogen gamma chain gene p.Glu275Lys causes congenital hypofibrinogenemia. Blood Coagul Fibrinolysis 2025:00001721-990000000-00200. [PMID: 40310436 DOI: 10.1097/mbc.0000000000001362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/27/2025] [Indexed: 05/02/2025]
Abstract
Congenital hypofibrinogenemia presents not only with bleeding, but also paradoxically with thrombosis. This heterogeneity of clinical phenotype complicates both diagnosis and management. The thrombotic phenotype is thought to arise from alterations in fibrin structure and stability, leading to abnormal clot formation and an increased risk of thrombosis. Coagulation assays, gene analysis, and protein modeling were utilized to elucidate the pathogenic variant. We highlight the pathophysiology of the novel missense variant in the FGG gene (c.823G/A, p.Glu275Lys), which causes mild hypofibrinogenemia and clinically manifests as an ischemic stroke. Protein modeling displays that the amino-acid substitution of glutamine with lysine at position 275 in mentioned missense variant causes local changes in the fibrinogen structure. The structural changes are mainly minor surface alterations and changes in physicochemical properties, which could potentially affect the recruitment of other proteins or lead to abnormal fibrin polymerization. This study provides novel insights into the pathophysiological mechanism, emphasizing the importance of molecular and structural analyses in understanding and managing atypical presentations of fibrinogen disorders.
Collapse
Affiliation(s)
- Miroslava Drotarova
- National Centre of Hemostasis and Thrombosis, Department of Hematology and Transfusiology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital Martin, Slovakia
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- IRCCS Humanitas Research Hospital, Rozzano
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Ingrid Skornova
- National Centre of Hemostasis and Thrombosis, Department of Hematology and Transfusiology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital Martin, Slovakia
| | - Jana Zolkova
- National Centre of Hemostasis and Thrombosis, Department of Hematology and Transfusiology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital Martin, Slovakia
| | | | | | - Vladimir Podusel
- Department of Internal Medicine I., Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia
| | - Jan Stasko
- National Centre of Hemostasis and Thrombosis, Department of Hematology and Transfusiology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital Martin, Slovakia
| | - Tomas Simurda
- National Centre of Hemostasis and Thrombosis, Department of Hematology and Transfusiology, Comenius University in Bratislava, Jessenius Faculty of Medicine and University Hospital Martin, Slovakia
| |
Collapse
|
27
|
Gajardo M, Guerrero JL, Poblete B, Bayyad E, Castro I, Maturana J, Tobar J, Faúndes V, Krall P. Systematic use of protein free energy changes for classifying variants of uncertain significance: the case of IFT140 in Mainzer-Saldino Syndrome. Front Mol Biosci 2025; 12:1561380. [PMID: 40337643 PMCID: PMC12055525 DOI: 10.3389/fmolb.2025.1561380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/06/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Advanced genetic strategies have transformed our understanding of the genetic basis and diagnosis of many phenotypes, including rare diseases. However, missense variants (MVs) are frequently identified and often classified as variants of uncertain significance (VUS). Although changes in protein free energy (ΔΔG) were recently proposed as a tool for VUS classification, no objective cut-offs exist to distinguish between benign and pathogenic variants. Methods We utilized the computational tool mCSM to calculate ΔΔG and predict the impact of MVs on protein stability. Specifically, we systematically analyzed the ΔΔG of MVs in IFT140 to identify those potentially pathogenic and associated with Mainzer-Saldino syndrome (MSS). To this end, we evaluated ΔΔG in IFT140 MVs sourced from ClinVar, gnomAD, and MSS patients, aiming to resolve the diagnosis of MSS in a child with a novel homozygous IFT140 variant, initially reported as a VUS. Results IFT140 MVs from MSS patients showed lower ΔΔG values than those reported in gnomAD individuals (-1.389 vs. -0.681 kcal/mol; p = 0.0031). A ROC curve demonstrated strong discriminative ability (AUC = 0.8488; p = 0.0002), and a ΔΔG cut-off of -1.3 kcal/mol achieving 50% sensibility and 90% specificity. The analysis of ClinVar IFT140 variants classified as VUS, showed that 75/323 (23%) presented ΔΔG values below the cut-off. In the child clinically suspicious of MSS, this cut-off allowed the reclassification of the VUS (IFT140:p.W80C; ΔΔG = -1.745 kcal/mol) as likely pathogenic, which confirmed the diagnosis molecularly. Conclusion Our findings demonstrate that ΔΔG analysis can effectively distinguish potentially pathogenic variants in IFT140, enabling confirmation of MSS. The established cut-off of -1.3 kcal/mol showed strong discriminative power, aiding in the reclassification of VUS identified in IFT140. This approach highlights the utility of protein stability predictions in resolving diagnostic uncertainty in rare diseases.
Collapse
Affiliation(s)
| | | | - Bárbara Poblete
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Esperanza Bayyad
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Ignacio Castro
- Instituto de Informática, Facultad de Ciencias e Ingeniería, Universidad Austral de Chile, Valdivia, Chile
| | - Jorge Maturana
- Instituto de Informática, Facultad de Ciencias e Ingeniería, Universidad Austral de Chile, Valdivia, Chile
| | - Jaime Tobar
- Servicio de Pediatría, Hospital de Arica, Arica, Chile
| | - Víctor Faúndes
- Laboratorio de Genética y Enfermedades Metabólicas, Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| | - Paola Krall
- Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Centro de Investigación Clínica Avanzada (CICA)-Hospital Luis Calvo Mackenna, Santiago, Chile
| |
Collapse
|
28
|
Medina FE, Coloma J, Oviedo C. Theoretical conformational analysis of cross-linking bonds in fungal hydrophobin from Aspergillus fumigatus. J Biomol Struct Dyn 2025:1-10. [PMID: 40265330 DOI: 10.1080/07391102.2025.2496289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
Aspergillus fumigatus is a common saprophytic filamentous fungus that plays a crucial role in nutrient cycling but can become an opportunistic pathogen, posing a significant threat to immunocompromised individuals by causing invasive aspergillosis. A key feature of A. fumigatus is the presence of hydrophobins-small amphipathic proteins that form a protective rodlet layer on conidial surfaces, facilitating biofilm formation and immune evasion. This rodlet structure, stabilized by cross-linking disulfide bonds, provides resistance to desiccation, oxidative stress, and immune defenses, making these cross-links a compelling target for study. In this work, we employ all-atom simulations, incorporating quantum mechanics/molecular mechanics (QM/MM) calculations, to evaluate the energy and conformational effects of cross-linking disulfide bonds (CL1, CL2, CL3, and CL4) in the rodlet assembly. By integrating QM/MM approaches, we achieve a detailed representation of the electronic and structural properties of these bonds within the complex rodlet layer, gaining deeper insights into their essential role in maintaining the stability and integrity of the RodA hydrophobin protein from A. fumigatus conidial surface. We identify a group of ten residues that influence directly in the cross-linking, with Gln23 and Lys17 emerging as key candidates for experimental mutation to control rodlet assembly. Our findings shed light on the molecular mechanisms underlying rodlet formation and highlight potential targets for disrupting this protective layer, offering promising avenues for antifungal strategies.
Collapse
Affiliation(s)
- Fabiola E Medina
- Departamento de Química, Facultad de Ciencias, Universidad del Bío-Bío, Concepción, Chile
| | - Juana Coloma
- Departamento de Ingeniería de Maderas, Facultad de Ingeniería, Universidad del Bío-Bío, Concepción, Chile
| | - Claudia Oviedo
- Departamento de Química, Facultad de Ciencias, Universidad del Bío-Bío, Concepción, Chile
| |
Collapse
|
29
|
Kaczmarska A, Christen M, Del Caño-Ochoa F, Ramon-Maiques S, Miro AC, Rupp A, Jagannathan V, Leeb T, Gutierrez-Quintana R. Epileptic encephalopathy in a young Bengal cat caused by CAD deficiency. Sci Rep 2025; 15:13506. [PMID: 40251393 PMCID: PMC12008240 DOI: 10.1038/s41598-025-98414-0] [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: 10/21/2024] [Accepted: 04/11/2025] [Indexed: 04/20/2025] Open
Abstract
Developmental and epileptic encephalopathy type 50 (DEE50) in humans is a severe early-onset neurometabolic disorder caused by biallelic loss-of-function variants in the CAD gene encoding a key multi-enzymatic protein for de novo pyrimidine nucleotide synthesis. Untreated, the condition is often fatal, but patients respond to uridine supplementation, which fuels nucleotide synthesis through CAD-independent salvage pathways. Here, we report a novel variant in the feline CAD gene in a 4-month-old Bengal kitten with intractable seizures and abnormal behavior. The variant, XP_011279586.1:p.(Ser2015Asn), was predicted to affect the oligomerization of the C-terminal aspartate transcarbamylase (ATCase) domain of CAD. Genotyping of 110 unaffected Bengal cats revealed four additional carriers of the mutant allele, confirming its presence in the breed. In a CAD-knockout human cell line dependent on uridine, the recombinant expression of human wildtype CAD, but not of the Asn2015 mutant, restored cell growth without uridine, demonstrating that the p.Ser2015Asn variant disrupts CAD function and is pathogenic. This study facilitates genetic testing of carriers and affected cats and suggests that uridine supplementation could be a potential treatment. Furthermore, CAD-deficient Bengal cats might serve as a valuable spontaneous large animal model to further investigate the pathogenic mechanisms of this rare epileptic encephalopathy in humans.
Collapse
Affiliation(s)
- Adriana Kaczmarska
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK
| | - Matthias Christen
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bremgartenstrasse 109a, 3001, Bern, Switzerland
| | | | - Santiago Ramon-Maiques
- Instituto de Biomedicina de Valencia (IBV), CSIC, Valencia, Spain
- Group CB06/07/0077, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-Instituto de Salud Carlos III, Valencia, Spain
- Valencia Biomedical Research Foundation, Centro de Investigación Príncipe Felipe (CIPF) - Associated Unit to the Instituto de Biomedicina de Valencia (IBV), Valencia, Spain
| | - Ana Cloquell Miro
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK
| | - Angie Rupp
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK
| | - Vidhya Jagannathan
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bremgartenstrasse 109a, 3001, Bern, Switzerland
| | - Tosso Leeb
- Vetsuisse Faculty, Institute of Genetics, University of Bern, Bremgartenstrasse 109a, 3001, Bern, Switzerland.
| | | |
Collapse
|
30
|
Liang T, Sun ZY, Ishima R, Xie XQ, Xue Y, Li W, Feng Z. ProstaNet: A Novel Geometric Vector Perceptrons-Graph Neural Network Algorithm for Protein Stability Prediction in Single- and Multiple-Point Mutations with Experimental Validation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0674. [PMID: 40235597 PMCID: PMC11997553 DOI: 10.34133/research.0674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/21/2025] [Accepted: 03/23/2025] [Indexed: 04/17/2025]
Abstract
Proteins play a critical role in biology and biopharma due to their specificity and minimal side effects. Predicting the effects of mutations on protein stability is vital but experimentally challenging. Deep learning offers an efficient solution to this problem. In the present work, we introduced ProstaNet, a deep learning framework that predicts stability changes resulting from single- and multiple-point mutations using geometric vector perceptrons-graph neural network for 3-dimensional feature processing. For training ProstaNet, we meticulously crafted ProstaDB, a comprehensive and pristine thermodynamics repository, including 3,784 single-point mutations and 1,642 multiple-point mutations. We also created thermodynamic looping for enlarging the limited data size of multiple-point mutation and applied an innovative clustering method to generate a standard testing set of multiple-point mutation. Besides, we identified residue scoring as the most important encoding method in protein properties prediction. With these innovations, ProstaNet accurately predicts thermostability changes for both single-point and multiple-point mutations without showing any bias. ProstaNet achieves an accuracy of 0.75, outperforming existing methods for single-point mutation prediction, including ThermoMPNN (0.63), PoPMuSiCsym (0.66), MUPRO (0.52), and FoldX (0.71). ProstaNet also achieves a 1.3-fold increase in accuracy compared to FoldX for multiple-point mutation predictions. Validated by experiment, 4 out of 5 single-point mutation predictions (80%) and all multiple-point mutation predictions (100%) for HuJ3 mutants were accurate, demonstrating the potential benefits of ProstaNet for protein engineering and drug development.
Collapse
Affiliation(s)
- Tianjian Liang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics and System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research,
University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ze-Yu Sun
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics and System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research,
University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Rieko Ishima
- Department of Structural Biology, School of Medicine,
University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics and System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research,
University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Ying Xue
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics and System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research,
University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Wei Li
- Department of Medicine, Center for Antibody Therapeutics, Division of Infectious Diseases, School of Medicine,
University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics and System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research,
University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
31
|
Liu C, Cai S, Pan T, Ogata H, Song J, Akutsu T. SFM-Net: Selective Fusion of Multiway Protein Feature Network for Predicting Binding Affinity Changes upon Mutations. J Chem Inf Model 2025; 65:3854-3865. [PMID: 40111004 DOI: 10.1021/acs.jcim.5c00130] [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: 03/22/2025]
Abstract
Accurately predicting the effect of mutations on protein-protein interactions (PPIs) is essential for understanding the protein structure and function, as well as providing insights into disease-causing mechanisms. Many recent popular approaches based on the three-dimensional structure of proteins have been proposed to predict the changes in binding affinity caused by mutations, i.e. ΔΔG. However, how to effectively use the structural information to comprehensively exploit complex interactions within proteins and integrate multisource features remains a significant challenge. In this study, we propose SFM-Net, a powerful deep learning model constructed with GNN-based multiway feature extractors and a new context-aware selective fusion module that jointly leverages the sequence, structural, and evolutionary information. Such design enables SFM-Net to effectively and selectively use features from different sources to facilitate binding affinity change prediction. Benchmarking experiments and targeted ablation studies illustrate the effectiveness and robustness of our method for improving the binding affinity change prediction.
Collapse
Affiliation(s)
- Chunting Liu
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan
| | - Sudong Cai
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
| | - Tong Pan
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne 3800, Australia
| | - Hiroyuki Ogata
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne 3800, Australia
- Monash Data Futures Institute, Monash University, Melbourne 3800, Australia
| | - Tatsuya Akutsu
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Uji 611-0011, Japan
| |
Collapse
|
32
|
Lian E, Belardinelli JM, De K, Pandurangan AP, Angala SK, Palčeková Z, Grzegorzewicz AE, Bryant JM, Blundell TL, Parkhill J, Floto RA, Wheat WH, Jackson M. Cell envelope polysaccharide modifications alter the surface properties and interactions of Mycobacterium abscessus with innate immune cells in a morphotype-dependent manner. mBio 2025; 16:e0032225. [PMID: 40084888 PMCID: PMC11980365 DOI: 10.1128/mbio.00322-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025] Open
Abstract
Mycobacterium abscessus is one of the leading causes of pulmonary infections caused by non-tuberculous mycobacteria. The ability of M. abscessus to establish a chronic infection in the lung relies on a series of adaptive mutations impacting, in part, global regulators and cell envelope biosynthetic enzymes. One of the genes under strong evolutionary pressure during host adaptation is ubiA, which participates in the elaboration of the arabinan domains of two major cell envelope polysaccharides: arabinogalactan (AG) and lipoarabinomannan (LAM). We here show that patient-derived UbiA mutations not only cause alterations in the AG, LAM, and mycolic acid contents of M. abscessus but also tend to render the bacterium more prone to forming biofilms while evading uptake by innate immune cells and enhancing their pro-inflammatory properties. The fact that the effects of UbiA mutations on the physiology and pathogenicity of M. abscessus were impacted by the rough or smooth morphotype of the strain suggests that the timing of their selection relative to morphotype switching may be key to their ability to promote chronic persistence in the host.IMPORTANCEMultidrug-resistant pulmonary infections caused by Mycobacterium abscessus and subspecies are increasing in the U.S.A. and globally. Little is known of the mechanisms of pathogenicity of these microorganisms. We have identified single-nucleotide polymorphisms (SNPs) in a gene involved in the biosynthesis of two major cell envelope polysaccharides, arabinogalactan and lipoarabinomannan, in lung-adapted isolates from 13 patients. Introduction of these individual SNPs in a reference M. abscessus strain allowed us to study their impact on the physiology of the bacterium and its interactions with immune cells. The significance of our work is in identifying some of the mechanisms used by M. abscessus to colonize and persist in the human lung, which will facilitate the early detection of potentially more virulent clinical isolates and lead to new therapeutic strategies. Our findings may further have broader biomedical impacts, as the ubiA gene is conserved in other tuberculous and non-tuberculous mycobacterial pathogens.
Collapse
Affiliation(s)
- Elena Lian
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Juan M. Belardinelli
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Kavita De
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Arun Prasad Pandurangan
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Trumpington, Cambridge, UK
- Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
| | - Shiva K. Angala
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Zuzana Palčeková
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Anna E. Grzegorzewicz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | | | - Tom L. Blundell
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Trumpington, Cambridge, UK
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - R. Andres Floto
- Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge, UK
- Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology, Trumpington, Cambridge, UK
| | - William H. Wheat
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
33
|
Zhang Y, Deng J, Dong M, Wu J, Zhao Q, Gao X, Xiong D. PILOT: Deep Siamese network with hybrid attention improves prediction of mutation impact on protein stability. Neural Netw 2025; 188:107476. [PMID: 40252373 DOI: 10.1016/j.neunet.2025.107476] [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: 10/05/2024] [Revised: 02/13/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
Evaluating the mutation impact on protein stability (ΔΔG) is essential in the study of protein engineering and understanding molecular mechanisms of disease-associated mutations. Here, we propose a novel deep learning framework, PILOT, for improved prediction of ΔΔG using a Siamese network with hybrid attention mechanism. The PILOT framework leverages multiple attention modules to effectively extract representations for amino acids, atoms, and protein sequences, respectively. This approach significantly ensures the deep fusion of structural information at both residue and atom levels, the seamless integration of structural and sequence representations, and the effective capture of both long-range and short-range dependencies among amino acids. Our extensive evaluations demonstrate that PILOT greatly outperforms other state-of-the-art methods. We also showcase that PILOT identifies exceptional patterns for different mutation types. Moreover, we illustrate the clinical applicability of PILOT in highlighting pathogenic variants from benign variants and VUS (variants of uncertain significance), and distinguishing de novo mutations in disease cases and controls. In summary, PILOT presents a robust deep learning tool that could offer significant insights into drug design, medical applications, and protein engineering studies.
Collapse
Affiliation(s)
- Yuan Zhang
- 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
| | - Mingyuan Dong
- 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
|
34
|
Reddy PR, Kulandaisamy A, Gromiha MM. TMB Stab-pred: Predicting the stability of transmembrane β-barrel proteins using their sequence and structural signatures. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141070. [PMID: 40189175 DOI: 10.1016/j.bbapap.2025.141070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/03/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Understanding the folding and stability of transmembrane β-barrel proteins (TMBs) provides insights into their structural integrity, functional mechanisms, and implications for disease states. In this work, we have characterized the important features that influence the folding and stability of TMBs. Our results showed that lipid accessible surface area and transition energy are important for understanding the stability of TMBs. Further, this information was utilized to develop a linear regression-based method for predicting the stability of TMBs. Our method achieved a correlation and mean absolute error (MAE) of 0.96 and 0.94 kcal/mol on the jack-knife test. Moreover, we compared the stability of TMBs with globular all-β proteins and observed that long-range interactions and energetic properties are crucial for maintaining the stability of both β-barrel membrane and all-β globular proteins. On the other hand, side-chain - side-chain hydrogen bonds and lipid accessible surface area are specific to membrane proteins. These features are critical for membrane proteins because they influence a protein to embed within the membrane environment. Further, we have developed a web server, TMB Stab-pred for predicting the stability of TMBs, and it is accessible at https://web.iitm.ac.in/bioinfo2/TMBB/index.html.
Collapse
Affiliation(s)
- P Ramakrishna Reddy
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - A Kulandaisamy
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India.
| |
Collapse
|
35
|
Xia Y, Wang Z, Huang F, Xiong Z, Wang Y, Qiu M, Zhang W. DeepInterAware: Deep Interaction Interface-Aware Network for Improving Antigen-Antibody Interaction Prediction from Sequence Data. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412533. [PMID: 39932383 PMCID: PMC11967782 DOI: 10.1002/advs.202412533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/06/2025] [Indexed: 04/05/2025]
Abstract
Identifying interactions between candidate antibodies and target antigens is a key step in developing effective human therapeutics. The antigen-antibody interaction (AAI) occurs at the structural level, but the limited structure data poses a significant challenge. However, recent studies revealed that structural information can be learned from the vast amount of sequence data, indicating that the interaction prediction can benefit from the abundance of antigen and antibody sequences. In this study, DeepInterAware (deep interaction interface-aware network) is proposed, a framework dynamically incorporating interaction interface information directly learned from sequence data, along with the inherent specificity information of the sequences. Experimental results in interaction prediction demonstrate that DeepInterAware outperforms existing methods and exhibits promising inductive capabilities for predicting interactions involving unseen antigens or antibodies, and transfer capabilities for similar tasks. More notably, DeepInterAware has unique advantages that existing methods lack. First, DeepInterAware can dive into the underlying mechanisms of AAIs, offering the ability to identify potential binding sites. Second, it is proficient in detecting mutations within antigens or antibodies, and can be extended for precise predictions of the binding free energy changes upon mutations. The HER2-targeting antibody screening experiment further underscores DeepInterAware's exceptional capability in identifying binding antibodies for target antigens, establishing it as an important tool for antibody screening.
Collapse
Affiliation(s)
- Yuhang Xia
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Zhiwei Wang
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Feng Huang
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Zhankun Xiong
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Yongkang Wang
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Minyao Qiu
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| | - Wen Zhang
- College of InformaticsHuazhong Agricultural UniversityWuhan430070China
| |
Collapse
|
36
|
Arnaudi M, Utichi M, Tiberti M, Papaleo E. Predicting the structure-altering mechanisms of disease variants. Curr Opin Struct Biol 2025; 91:102994. [PMID: 40020537 DOI: 10.1016/j.sbi.2025.102994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 03/03/2025]
Abstract
Missense variants can affect the severity of disease, choice of treatment, and treatment outcomes. While the number of known variants has been increasing at a rapid pace, available evidence of their clinical effect has been lagging behind, constituting a challenge for clinicians and researchers. Multiplexed assays of variant effects (MAVEs) are important to close the gap; nonetheless, computational predictions of pathogenicity are still often the only available data for scoring variants. Such methods are not designed to provide a mechanistic explanation for the effect of amino acid substitutions. To this purpose, we propose structure-based frameworks as ensemble methodologies, with each method tailored to predict a different aspect among those exerted by amino acid substitutions to link predicted pathogenicity to mechanistic indicators. We review available frameworks, as well as advancements in underlying structure-based methods that predict variant effects on several protein features, such as protein stability, biomolecular interactions, allostery, post-translational modifications, and more.
Collapse
Affiliation(s)
- Matteo Arnaudi
- Cancer Structural Biology, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark; Cancer Systems Biology, Section of Bioinformatics, Health and Technology Department, Technical University of Denmark, Lyngby, Denmark
| | - Mattia Utichi
- Cancer Structural Biology, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark; Cancer Systems Biology, Section of Bioinformatics, Health and Technology Department, Technical University of Denmark, Lyngby, Denmark
| | - Matteo Tiberti
- Cancer Structural Biology, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark.
| | - Elena Papaleo
- Cancer Structural Biology, Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark; Cancer Systems Biology, Section of Bioinformatics, Health and Technology Department, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
37
|
Rajendra D, Maroli N, Dixit NM, Maiti PK. Molecular dynamics simulations show how antibodies may rescue HIV-1 mutants incapable of infecting host cells. J Biomol Struct Dyn 2025; 43:2982-2992. [PMID: 38111161 DOI: 10.1080/07391102.2023.2294835] [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: 08/10/2023] [Accepted: 12/01/2023] [Indexed: 12/20/2023]
Abstract
High mutation and replication rates of HIV-1 result in the continuous generation of variants, allowing it to adapt to changing host environments. Mutations often have deleterious effects, but variants carrying them are rapidly purged. Surprisingly, a particular variant incapable of entering host cells was found to be rescued by host antibodies targeting HIV-1. Understanding the molecular mechanism of this rescue is important to develop and improve antibody-based therapies. To unravel the underlying mechanisms, we performed fully atomistic molecular dynamics simulations of the HIV-1 gp41 trimer responsible for viral entry into host cells, its entry-deficient variant, and its complex with the rescuing antibody. We find that the Q563R mutation, which the entry-deficient variant carries, prevents the native conformation of the gp41 6-helix bundle required for entry and stabilizes an alternative conformation instead. This is the consequence of substantial changes in the secondary structure and interactions between the domains of gp41. Binding of the antibody F240 to gp41 reverses these changes and re-establishes the native conformation, resulting in rescue. To test the generality of this mechanism, we performed simulations with the entry-deficient L565A variant and antibody 3D6. We find that 3D6 binding was able to reverse structural and interaction changes introduced by the mutation and restore the native gp41 conformation. Viral variants may not only escape antibodies but be aided by them in their survival, potentially compromising antibody-based therapies, including vaccination and passive immunization. Our simulation framework could serve as a tool to assess the likelihood of such resistance against specific antibodies.
Collapse
Affiliation(s)
- Dharanish Rajendra
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bengaluru, India
| | - Nikhil Maroli
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bengaluru, India
| | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bengaluru, India
- Department of Bioengineering, Indian Institute of Science, Bengaluru, India
| | - Prabal K Maiti
- Centre for Condensed Matter Theory, Department of Physics, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
38
|
Barroso da Silva FL, Paco K, Laaksonen A, Ray A. Biophysics of SARS-CoV-2 spike protein's receptor-binding domain interaction with ACE2 and neutralizing antibodies: from computation to functional insights. Biophys Rev 2025; 17:309-333. [PMID: 40376405 PMCID: PMC12075047 DOI: 10.1007/s12551-025-01276-z] [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: 12/01/2024] [Accepted: 01/24/2025] [Indexed: 05/18/2025] Open
Abstract
The spike protein encoded by the SARS-CoV-2 has become one of the most studied macromolecules in recent years due to its central role in COVID-19 pathogenesis. The spike protein's receptor-binding domain (RBD) directly interacts with the host-encoded receptor protein, ACE2. This review critically examines computational insights into RBD's interaction with ACE2 and with therapeutic antibodies designed to interfere with this interaction. We begin by summarizing insights from early computational studies on pre-pandemic SARS-CoV-1 RBD interactions and how these early studies shaped the understanding of SARS-CoV-2. Next, we highlight key theoretical contributions that revealed the molecular mechanisms behind the binding affinity of SARS-CoV-2 RBD against ACE2, and the structural changes that have enhanced the infectivity of emerging variants. Special attention is given to the "RBD charge rule", a predictive framework for determining variant infectivity based on the electrostatic properties of the RBD. Towards applying the computational insights to therapy, we discuss a multiscale computational protocol for optimizing monoclonal antibodies to improve binding affinity across multiple spike protein variants, including representatives from the Omicron family. Finally, we explore how these insights can inform the development of future vaccines and therapeutic interventions for combating future coronavirus diseases.
Collapse
Affiliation(s)
- Fernando Luís Barroso da Silva
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av Prof Zeferino Vaz, S/no, Ribeirão Preto, São Paulo BR-14040-903 Brazil
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Engineering Building I (EB1), Raleigh, NC 27695-7905 USA
| | - Karen Paco
- Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr., Claremont, CA 91711 USA
| | - Aatto Laaksonen
- Department of Chemistry, Arrhenius Laboratory, Stockholm University, Svante Arrhenius Väg 8, 106 91 Stockholm, Sweden
- State Key Laboratory of Materials-Oriented and Chemical Engineering, Nanjing Tech University, NO.30 Puzhu Road(S), Nanjing, 210009 People’s Republic of China
- Department of Engineering Sciences and Mathematics, Division of Energy Science, Luleå University of Technology, Laboratorievägen 14, 97187 Luleå, Sweden
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, Petru Poni Institute of Macromolecular Chemistry, Aleea Grigore Ghica-Voda, 41A, 700487 Iasi, Romania
| | - Animesh Ray
- Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr., Claremont, CA 91711 USA
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125 USA
| |
Collapse
|
39
|
Mandl Š, Di Geronimo B, Alonso‐Gil S, Grininger C, George G, Ferstl U, Herzog SA, Žagrović B, Nusshold C, Pavkov‐Keller T, Sánchez‐Murcia PA. A new view of missense mutations in α-mannosidosis using molecular dynamics conformational ensembles. Protein Sci 2025; 34:e70080. [PMID: 40126164 PMCID: PMC11931667 DOI: 10.1002/pro.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 03/25/2025]
Abstract
The mutation of remote positions on enzyme scaffolds and how these residue changes can affect enzyme catalysis is still far from being fully understood. One paradigmatic example is the group of lysosomal storage disorders, where the enzyme activity of a lysosomal enzyme is abolished or severely reduced. In this work, we analyze molecular dynamics simulation conformational ensembles to unveil the molecular features controlling the deleterious effects of the 43 reported missense mutations in the human lysosomal α-mannosidase. Using residue descriptors for protein dynamics, their coupling with the active site, and their impact on protein stability, we have assigned the contribution of each of the missense mutations into protein stability, protein dynamics, and their connectivity with the active site. We demonstrate here that the use of conformational ensembles is a powerful approach not only to better understand missense mutations at the molecular level but also to revisit the missense mutations reported in lysosomal storage disorders in order to aid the treatment of these diseases.
Collapse
Affiliation(s)
- Špela Mandl
- Laboratory of Computer‐Aided Molecular Design, Division of Medicinal Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
| | - Bruno Di Geronimo
- Laboratory of Computer‐Aided Molecular Design, Division of Medicinal Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
- Present address:
School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Santiago Alonso‐Gil
- Max Perutz LabsVienna Biocenter Campus (VBC)ViennaAustria
- Department of Structural and Computational BiologyVienna BioCenter University of Vienna Campus‐Vienna‐Biocenter 5ViennaAustria
| | | | - Gibu George
- Institut de Química Computacional i Catàlisi and Departament de QuímicaUniversitat de GironaGironaCataloniaSpain
| | - Ulrika Ferstl
- Laboratory of Computer‐Aided Molecular Design, Division of Medicinal Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
| | - Sereina Annik Herzog
- Institute for Medical Informatics, Statistics and DocumentationMedical University of GrazGrazAustria
| | - Bojan Žagrović
- Max Perutz LabsVienna Biocenter Campus (VBC)ViennaAustria
- Department of Structural and Computational BiologyVienna BioCenter University of Vienna Campus‐Vienna‐Biocenter 5ViennaAustria
| | - Christoph Nusshold
- Laboratory of Computer‐Aided Molecular Design, Division of Medicinal Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
| | - Tea Pavkov‐Keller
- Institute of Molecular Biosciences, NAWI GrazUniversity of GrazGrazAustria
- Field of Excellence BioHealthUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| | - Pedro A. Sánchez‐Murcia
- Laboratory of Computer‐Aided Molecular Design, Division of Medicinal Chemistry, Otto‐Loewi Research CenterMedical University of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
| |
Collapse
|
40
|
Allman BE, Vieira L, Diaz DJ, Wilke CO. A systematic evaluation of the language-of-viral-escape model using multiple machine learning frameworks. J R Soc Interface 2025; 22:20240598. [PMID: 40300635 PMCID: PMC12040448 DOI: 10.1098/rsif.2024.0598] [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: 08/29/2024] [Revised: 01/02/2025] [Accepted: 02/18/2025] [Indexed: 05/01/2025] Open
Abstract
Predicting the evolutionary patterns of emerging and endemic viruses is key for mitigating their spread. In particular, it is critical to rapidly identify mutations with the potential for immune escape or increased disease burden. Knowing which circulating mutations pose a concern can inform treatment or mitigation strategies such as alternative vaccines or targeted social distancing. In 2021, Hie B, Zhong ED, Berger B, Bryson B. 2021 Learning the language of viral evolution and escape. Science 371, 284-288. (doi:10.1126/science.abd7331) proposed that variants of concern can be identified using two quantities extracted from protein language models, grammaticality and semantic change. These quantities are defined by analogy to concepts from natural language processing. Grammaticality is intended to be a measure of whether a variant viral protein is viable, and semantic change is intended to be a measure of potential for immune escape. Here, we systematically test this hypothesis, taking advantage of several high-throughput datasets that have become available, and also comparing this model with several more recently published machine learning models. We find that grammaticality can be a measure of protein viability, though methods that are trained explicitly to predict mutational effects appear to be more effective. By contrast, we do not find compelling evidence that semantic change is a useful tool for identifying immune escape mutations.
Collapse
Affiliation(s)
- Brent E. Allman
- Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Luiz Vieira
- Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel J. Diaz
- Institute for Foundations of Machine Learning, The University of Texas at Austin, Austin, Texas, USA
| | - Claus O. Wilke
- Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
41
|
Sugawara R, Hamada K, Ito H, Scala M, Ueda H, Tabata H, Ogata K, Nagata KI. A p.N92K variant of the GTPase RAC3 disrupts cortical neuron migration and axon elongation. J Biol Chem 2025; 301:108346. [PMID: 40015633 PMCID: PMC11968283 DOI: 10.1016/j.jbc.2025.108346] [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: 11/14/2024] [Revised: 02/05/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
RAC3 encodes a small GTPase of the Rho family, crucial for actin cytoskeleton organization and signaling pathways. De novo deleterious variants in RAC3 cause neurodevelopmental disorder with structural brain anomalies and dysmorphic facies (NEDBAF). Disease-causing variants thus far reported are thought to impact key conserved regions within RAC3, such as the P-loop, switch I/II, and G boxes, which are essential for the interaction with regulatory proteins and effectors. Recently, however, a novel variant, c.276T > A, p.N92K, was identified in a prenatal case with complex brain malformations. This variant, located outside the core functional regions, represents a unique class of RAC3 pathogenic mutations. We investigated the variant's effects using in vitro, in silico, and in vivo approaches. Overexpression of RAC3-N92K in primary hippocampal neurons impaired differentiation, leading to round cell shape with lamellipodia, suggesting that RAC3-N92K is active. Biochemical studies showed that RAC3-N92K is (1) resistant to GAP-mediated inactivation, (2) responsive to GEF activation, and (3) capable of interacting with RAC effectors PAK1 and MLK2, as well as Rho-kinase 1, activating gene expression through SRF, NFκB, and AP1 pathways. Structural analyses suggest that N92K disrupts GAP interactions but preserves interactions with GEF, PAK1, and MLK2. In vivo, RAC3-N92K expression in embryonic mouse cortical neurons led to migration defects and periventricular clustering during corticogenesis, along with impaired axon elongation. These findings indicate that RAC3-N92K's activated state significantly disrupts cortical development, expanding the genetic and pathophysiological spectrum of NEDBAF.
Collapse
Affiliation(s)
- Ryota Sugawara
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| | - Keisuke Hamada
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy; Unit of Medical Genetics, IRCCS Giannina Gaslini Institute, Genova, Italy
| | - Hiroshi Ueda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, Japan
| | - Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Japan; Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
42
|
Dewaker V, Morya VK, Kim YH, Park ST, Kim HS, Koh YH. Revolutionizing oncology: the role of Artificial Intelligence (AI) as an antibody design, and optimization tools. Biomark Res 2025; 13:52. [PMID: 40155973 PMCID: PMC11954232 DOI: 10.1186/s40364-025-00764-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
Antibodies play a crucial role in defending the human body against diseases, including life-threatening conditions like cancer. They mediate immune responses against foreign antigens and, in some cases, self-antigens. Over time, antibody-based technologies have evolved from monoclonal antibodies (mAbs) to chimeric antigen receptor T cells (CAR-T cells), significantly impacting biotechnology, diagnostics, and therapeutics. Although these advancements have enhanced therapeutic interventions, the integration of artificial intelligence (AI) is revolutionizing antibody design and optimization. This review explores recent AI advancements, including large language models (LLMs), diffusion models, and generative AI-based applications, which have transformed antibody discovery by accelerating de novo generation, enhancing immune response precision, and optimizing therapeutic efficacy. Through advanced data analysis, AI enables the prediction and design of antibody sequences, 3D structures, complementarity-determining regions (CDRs), paratopes, epitopes, and antigen-antibody interactions. These AI-powered innovations address longstanding challenges in antibody development, significantly improving speed, specificity, and accuracy in therapeutic design. By integrating computational advancements with biomedical applications, AI is driving next-generation cancer therapies, transforming precision medicine, and enhancing patient outcomes.
Collapse
Affiliation(s)
- Varun Dewaker
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
| | - Vivek Kumar Morya
- Department of Orthopedic Surgery, Hallym University Dongtan Sacred Hospital, Hwaseong-Si, 18450, Republic of Korea
| | - Yoo Hee Kim
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea
| | - Sung Taek Park
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea
- Department of Obstetrics and Gynecology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea
| | - Hyeong Su Kim
- Institute of New Frontier Research Team, Hallym University, Chuncheon-Si, Gangwon-Do, 24252, Republic of Korea.
- Department of Internal Medicine, Division of Hemato-Oncology, Kangnam Sacred-Heart Hospital, Hallym University Medical Center, Hallym University College of Medicine, Seoul, 07441, Republic of Korea.
- EIONCELL Inc, Chuncheon-Si, 24252, Republic of Korea.
| | - Young Ho Koh
- Department of Biomedical Gerontology, Ilsong Institute of Life Science, Hallym University, Seoul, 07247, Republic of Korea.
| |
Collapse
|
43
|
Zhang L, Wu L, Guo Y, Wang E, Zhang J, You S, Su R, Qi W. Enhancing amplification efficiency and reducing molecular diagnostic reaction time through rational design of T4 gp32 Variants in recombinase polymerase amplification. Biochimie 2025; 234:1-9. [PMID: 40158835 DOI: 10.1016/j.biochi.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Recombinase polymerase amplification (RPA) is a prominent isothermal nucleic acid amplification method widely applied in molecular diagnostics. The stability and functionality of the single-stranded DNA-binding protein T4 gene 32 (gp32) crucial for pre-synaptic filament formation and D-loop stabilization, play a key role in determining RPA efficiency. In this study, V62C/T80C and Y186R mutants with improved performance were screened by rational disulfide bond construction and virtual saturation mutagenesis, respectively. The structural changes in V62C/T80C and the altered ssDNA-binding capacity in Y186R both contribute to RPA amplification by enhancing the formation of UvsX-ssDNA presynaptic filaments and stabilizing the D-loop structure during homologous recombination, respectively. The two mutants each demonstrated unique advantages in the RPA process. V62C/T80C significantly accelerates the amplification process, reducing the RPA reaction time by 47 %, while Y186R showed a 123 % increase in efficiency across the entire amplification cycle. Totally, this study applied a rational strategy on gp32 optimization, shortening RPA reaction times, enhancing the RPA reaction efficiency, and advancing its application in clinical and point-of-care diagnostics.
Collapse
Affiliation(s)
- Lin Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Lvping Wu
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Yiwei Guo
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Enjie Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Jiaxing Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China
| | - Shengping You
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China.
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China; State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300350, PR China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, PR China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, PR China; State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin, 300350, PR China; Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, PR China; Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, PR China
| |
Collapse
|
44
|
Cardamone F, Piva A, Löser E, Eichenberger B, Romero-Mulero MC, Zenk F, Shields EJ, Cabezas-Wallscheid N, Bonasio R, Tiana G, Zhan Y, Iovino N. Chromatin landscape at cis-regulatory elements orchestrates cell fate decisions in early embryogenesis. Nat Commun 2025; 16:3007. [PMID: 40148291 PMCID: PMC11950382 DOI: 10.1038/s41467-025-57719-4] [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: 08/02/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
The establishment of germ layers during early development is crucial for body formation. The Drosophila zygote serves as a model for investigating these transitions in relation to the chromatin landscape. However, the cellular heterogeneity of the blastoderm embryo poses a challenge for gaining mechanistic insights. Using 10× Multiome, we simultaneously analyzed the in vivo epigenomic and transcriptomic states of wild-type, E(z)-, and CBP-depleted embryos during zygotic genome activation at single-cell resolution. We found that pre-zygotic H3K27me3 safeguards tissue-specific gene expression by modulating cis-regulatory elements. Furthermore, we demonstrate that CBP is essential for cell fate specification functioning as a transcriptional activator by stabilizing transcriptional factors binding at key developmental genes. Surprisingly, while CBP depletion leads to transcriptional arrest, chromatin accessibility continues to progress independently through the retention of stalled RNA Polymerase II. Our study reveals fundamental principles of chromatin-mediated gene regulation essential for establishing and maintaining cellular identities during early embryogenesis.
Collapse
Affiliation(s)
- Francesco Cardamone
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- International Max Planck Research School of Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), Freiburg, Germany
| | - Annamaria Piva
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Eva Löser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Bastian Eichenberger
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Mari Carmen Romero-Mulero
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Fides Zenk
- Epigenomics of Neurodevelopment, Brain Mind Institute, School of Life Sciences, EPFL - Ecole Polytechnique Federal Lusanne, Ecublens, Switzerland
| | - Emily J Shields
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Urology and Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Laboratory of Stem Cell Biology and Ageing, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
- Centre for Integrative Biological Signalling Studies (CIBSS), Freiburg, Germany
| | - Roberto Bonasio
- Epigenetics Institute, Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guido Tiana
- Università degli Studi di Milano and INFN, Milan, Italy
| | - Yinxiu Zhan
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy.
| | - Nicola Iovino
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| |
Collapse
|
45
|
Addai FP, Chen X, Zhu H, Zhen Z, Lin F, Feng C, Han J, Wang Z, Wang Y, Zhou Y. Structural Stabilization and Activity Enhancement of Glucoamylase via the Machine-Learning Technique and Immobilization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7347-7363. [PMID: 40080106 DOI: 10.1021/acs.jafc.4c11907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Glucoamylases (GLL) hydrolyze starch to glucose syrup without yielding intermediate oligosaccharides, but their lack of stability under industrial conditions poses a major limiting factor. Using consensus- and ancestral-based machine-learning tools, a functional GLL with six mutations (GLLI73l/T130V/N212V/D238G/N327M/S332P) was constructed that exhibited superior hydrolytic activity relative to the wild-type (WT-GLL). An oxidized multi-walled carbon nanotube (oMW-CNT) was used as a solid support to immobilize the WT-GLL with an immobilization capacity of 211.28 mg/g. The specific activity of mutant GLL-6M and GLL@oMW-CNTII was improved by 2.5-fold and 3.9-fold respectively, with both retaining 64.5% residual activity after incubation at 50 °C for 2 h compared to the WT-GLL with 42.6% activity. GLL and GLL-6M were however completely inactivated at 55 °C in 30 min while oMW-CNTII retained ∼43.1% activity. Our results demonstrate that employing a machine-learning approach for enzyme redesign and immobilization is a practicable alternative for improving enzyme performance and stability for industrial applications.
Collapse
Affiliation(s)
- Frank Peprah Addai
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xinglin Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hao Zhu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zongjian Zhen
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Feng Lin
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou, Zhejiang 313001, China
| | - Chengxiang Feng
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Juan Han
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhirong Wang
- Promotion Center for Rural Revitalization of Zhejiang, Hangzhou, Zhejiang 310020, China
| | - Yun Wang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yang Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| |
Collapse
|
46
|
Li D, Mailand N, Ewing E, Hoffmann S, Caswell RC, Pang L, Eason J, Dou Y, Sullivan KE, Hakonarson H, Levine MA. Quantitative hypermorphic FAM111A alleles cause autosomal recessive Kenny-Caffey syndrome type 2 and osteocraniostenosis. JCI Insight 2025; 10:e186862. [PMID: 39932783 PMCID: PMC11949059 DOI: 10.1172/jci.insight.186862] [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: 09/10/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Kenny-Caffey syndrome (KCS) is a rare genetic disorder characterized by extreme short stature, cortical thickening and medullary stenosis of tubular bones, facial dysmorphism, abnormal T cell function, and hypoparathyroidism. Biallelic loss-of-function variants in TBCE cause autosomal recessive type 1 KCS (KCS1). By contrast, heterozygous missense variants in a restricted region of the FAM111A gene have been identified in autosomal dominant type 2 KCS (KCS2) and a more severe lethal phenotype, osteocraniostenosis (OCS); these variants have recently been shown to confer a gain of function. In this study, we describe 2 unrelated children with KCS and OCS who were homozygous for different FAM111A variant alleles that result in replacement of the same residue, Tyr414 (c.1241A>G, p.Y414C and c.1240T>A, p.Y414N), in the mature FAM111A protein. Their heterozygous relatives are asymptomatic. Functional studies of recombinant FAM111AY414C demonstrated normal dimerization and a mild gain-of-function effect. This study provides evidence that both biallelic and monoallelic variants of FAM111A with varying degrees of activation can lead to dominant or recessive KCS2 and OCS.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Niels Mailand
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Emma Ewing
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Saskia Hoffmann
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Denmark
| | - Richard C. Caswell
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Lewis Pang
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Jacqueline Eason
- Department of Clinical Genetics, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Ying Dou
- Division of Allergy and Immunology and
| | - Kathleen E. Sullivan
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Allergy and Immunology and
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael A. Levine
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Division of Endocrinology and Diabetes and Center for Bone Health, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
47
|
Luo J, Ding K, Luo Y. Pareto-optimal sampling for multi-objective protein sequence design. iScience 2025; 28:112119. [PMID: 40160427 PMCID: PMC11952807 DOI: 10.1016/j.isci.2025.112119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/03/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Supervised machine learning (ML) has significantly advanced sequence-based protein property prediction. However, its inverse application, designing protein sequences with desired properties, remains under-explored. The challenges in sequence design stem from the vast search space and the rugged protein fitness landscape. In this work, we present MosPro, an efficient ML algorithm for property-guided protein sequence design. We frame sequence design as a discrete sampling problem. Utilizing a pre-trained differentiable ML model that predicts properties of sequences, MosPro shapes a distribution that assigns high probability mass to regions for high-property sequences. To generate designs, MosPro efficiently samples sequences from this constructed distribution. We further develop a Pareto optimization algorithm to propose sequences that are simultaneously optimized for multiple properties. Evaluations on experimental fitness landscapes demonstrated that MosPro generates sequences that optimally trade off multiple desiderata. Our results suggested an unparalleled potential of generative ML for efficient and controllable design for functional proteins.
Collapse
Affiliation(s)
- Jiaqi Luo
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30308, USA
| | - Kerr Ding
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30308, USA
| | - Yunan Luo
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30308, USA
| |
Collapse
|
48
|
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
|
49
|
Ozleyen A, Duran GN, Donmez S, Ozbil M, Doveston RG, Tumer TB. Identification and inhibition of PIN1-NRF2 protein-protein interactions through computational and biophysical approaches. Sci Rep 2025; 15:8907. [PMID: 40087364 PMCID: PMC11909128 DOI: 10.1038/s41598-025-89342-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 02/04/2025] [Indexed: 03/17/2025] Open
Abstract
NRF2 is a transcription factor responsible for coordinating the expression of over a thousand cytoprotective genes. Although NRF2 is constitutively expressed, its stability is modulated by the redox-sensitive protein KEAP1 and other conditional binding partner regulators. The new era of NRF2 research has highlighted the cooperation between NRF2 and PIN1 in modifying its cytoprotective effect. Despite numerous studies, the understanding of the PIN1-NRF2 interaction remains limited. Herein, we described the binding interaction of PIN1 and three different 14-mer long phospho-peptides mimicking NRF2 protein using computer-based, biophysical, and biochemical approaches. According to our computational analyses, the residues positioned in the WW domain of PIN1 (Ser16, Arg17, Ser18, Tyr23, Ser32, Gln33, and Trp34) were found to be crucial for PIN1-NRF2 interactions. Biophysical FP assays were used to verify the computational prediction. The data demonstrated that Pintide, a peptide predominantly interacting with the PIN1 WW-domain, led to a significant reduction in the binding affinity of the NRF2 mimicking peptides. Moreover, we evaluated the impact of known PIN1 inhibitors (juglone, KPT-6566, and EGCG) on the PIN1-NRF2 interaction. Among the inhibitors, KPT-6566 showed the most potent inhibitory effect on PIN1-NRF2 interaction within an IC50 range of 0.3-1.4 µM. Furthermore, our mass spectrometry analyses showed that KPT-6566 appeared to covalently modify PIN1 via conjugate addition, rather than disulfide exchange of the sulfonyl-acetate moiety. Altogether, such inhibitors would also be highly valuable molecular probes for further investigation of PIN1 regulation of NRF2 in the cellular context and potentially pave the way for drug molecules that specifically inhibit the cytoprotective effects of NRF2 in cancer.
Collapse
Affiliation(s)
- Adem Ozleyen
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK
- Health Institutes of Türkiye, Türkiye Biotechnology Institute, 06270, Ankara, Turkey
| | - Gizem Nur Duran
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Serhat Donmez
- Graduate Program of Molecular Biology and Genetics, School of Graduate Studies, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey
- Institute of Science and Technology Austria (ISTA), 3400, Klosterneuburg, Austria
| | - Mehmet Ozbil
- Institute of Biotechnology, Gebze Technical University, 41400, Gebze, Kocaeli, Turkey
| | - Richard G Doveston
- Leicester Institute for Structural and Chemical Biology, University of Leicester, Leicester, LE1 7RH, UK.
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey.
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
50
|
Groeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, et alGroeneweg S, van Geest FS, Martín M, Dias M, Frazer J, Medina-Gomez C, Sterenborg RBTM, Wang H, Dolcetta-Capuzzo A, de Rooij LJ, Teumer A, Abaci A, van den Akker ELT, Ambegaonkar GP, Armour CM, Bacos I, Bakhtiani P, Barca D, Bauer AJ, van den Berg SAA, van den Berge A, Bertini E, van Beynum IM, Brunetti-Pierri N, Brunner D, Cappa M, Cappuccio G, Castellotti B, Castiglioni C, Chatterjee K, Chesover A, Christian P, Coenen-van der Spek J, de Coo IFM, Coutant R, Craiu D, Crock P, DeGoede C, Demir K, Dewey C, Dica A, Dimitri P, Dremmen MHG, Dubey R, Enderli A, Fairchild J, Gallichan J, Garibaldi L, George B, Gevers EF, Greenup E, Hackenberg A, Halász Z, Heinrich B, Hurst AC, Huynh T, Isaza AR, Klosowska A, van der Knoop MM, Konrad D, Koolen DA, Krude H, Kulkarni A, Laemmle A, LaFranchi SH, Lawson-Yuen A, Lebl J, Leeuwenburgh S, Linder-Lucht M, López Martí A, Lorea CF, Lourenço CM, Lunsing RJ, Lyons G, Malikova JK, Mancilla EE, McCormick KL, McGowan A, Mericq V, Lora FM, Moran C, Muller KE, Nicol LE, Oliver-Petit I, Paone L, Paul PG, Polak M, Porta F, Poswar FO, Reinauer C, Rozenkova K, Seckold R, Seven Menevse T, Simm P, Simon A, Singh Y, Spada M, Stals MAM, Stegenga MT, Stoupa A, Subramanian GM, Szeifert L, Tonduti D, Turan S, Vanderniet J, van der Walt A, Wémeau JL, van Wermeskerken AM, Wierzba J, de Wit MCY, Wolf NI, Wurm M, Zibordi F, Zung A, Zwaveling-Soonawala N, Rivadeneira F, Meima ME, Marks DS, Nicola JP, Chen CH, Medici M, Visser WE. Mapping variants in thyroid hormone transporter MCT8 to disease severity by genomic, phenotypic, functional, structural and deep learning integration. Nat Commun 2025; 16:2479. [PMID: 40075072 PMCID: PMC11904026 DOI: 10.1038/s41467-025-56628-w] [Show More Authors] [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: 02/29/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025] Open
Abstract
Predicting and quantifying phenotypic consequences of genetic variants in rare disorders is a major challenge, particularly pertinent for 'actionable' genes such as thyroid hormone transporter MCT8 (encoded by the X-linked SLC16A2 gene), where loss-of-function (LoF) variants cause a rare neurodevelopmental and (treatable) metabolic disorder in males. The combination of deep phenotyping data with functional and computational tests and with outcomes in population cohorts, enabled us to: (i) identify the genetic aetiology of divergent clinical phenotypes of MCT8 deficiency with genotype-phenotype relationships present across survival and 24 out of 32 disease features; (ii) demonstrate a mild phenocopy in ~400,000 individuals with common genetic variants in MCT8; (iii) assess therapeutic effectiveness, which did not differ among LoF-categories; (iv) advance structural insights in normal and mutated MCT8 by delineating seven critical functional domains; (v) create a pathogenicity-severity MCT8 variant classifier that accurately predicted pathogenicity (AUC:0.91) and severity (AUC:0.86) for 8151 variants. Our information-dense mapping provides a generalizable approach to advance multiple dimensions of rare genetic disorders.
Collapse
Affiliation(s)
- Stefan Groeneweg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ferdy S van Geest
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Mariano Martín
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mafalda Dias
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Jonathan Frazer
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rosalie B T M Sterenborg
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hao Wang
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Anna Dolcetta-Capuzzo
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linda J de Rooij
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Alexander Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Ayhan Abaci
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Erica L T van den Akker
- Department of Paediatrics, Division of Endocrinology, Erasmus Medical Centre -Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Gautam P Ambegaonkar
- Department of Paediatric Neurology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Christine M Armour
- Regional Genetics Program, Children's Hospital of Eastern Ontario and Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Iiuliu Bacos
- Centrul Medical Dr. Bacos Cosma, Timisoara, Romania
| | - Priyanka Bakhtiani
- University of Louisville, Louisville, KY, USA
- Childrens Hospital Los Angeles, Los Angeles, CA, USA
| | - Diana Barca
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Andrew J Bauer
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sjoerd A A van den Berg
- Diagnostic Laboratory for Endocrinology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Amanda van den Berge
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesu' Children's Research Hospital IRCCS, Rome, Italy
| | - Ingrid M van Beynum
- Department of Pediatrics, Division of Pediatric Cardiology, Erasmus Medical Centre - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Doris Brunner
- Gottfried Preyer's Children Hospital, Vienna, Austria
| | - Marco Cappa
- Research Area for Innovative Therapies in Endocrinopathies, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Gerarda Cappuccio
- Department of Translational Medicine, Federico II University, 80131, Naples, Italy
- Neurological Research Institute and Baylor College of Medicine, Houston, TX, USA
| | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Castiglioni
- Department of Neurology, Clinica Meds, School of Medicine, Universidad Finis Terrae, Santiago, Chile
| | - Krishna Chatterjee
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander Chesover
- Division of Endocrinology, The Hospital for Sick Children and Department of Paediatrics, University of Toronto, Toronto, M5G 1×8, Canada
- Department of Endocrinology, Great Ormond Street Hospital for Children, London, UK
| | - Peter Christian
- East Kent Hospitals University NHS Foundation Trust, Ashford, UK
| | - Jet Coenen-van der Spek
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Irenaeus F M de Coo
- Department of Toxicogenomics, Unit Clinical Genomics, Maastricht University, MHeNs School for Mental Health and Neuroscience, Maastricht, The Netherlands
| | - Regis Coutant
- Department of Pediatric Endocrinology and Diabetology, University Hospital, Angers, France
| | - Dana Craiu
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Patricia Crock
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Christian DeGoede
- Department of Paediatric Neurology, Clinical Research Facility, Lancashire Teaching Hospitals NHS Trust, Lancashire, UK
| | - Korcan Demir
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Cheyenne Dewey
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
| | - Alice Dica
- Carol Davila University of Medicine, Department of Clinical Neurosciences, Paediatric Neurology Discipline II, Bucharest, Romania
| | - Paul Dimitri
- The Department of Oncology and Metabolism, The University of Sheffield, Western Bank, Sheffield, S10, 2TH, UK
| | - Marjolein H G Dremmen
- Division of Paediatric Radiology, Erasmus Medical Centre - Sophia's Children Hospital, Rotterdam, The Netherlands
| | | | - Anina Enderli
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Fairchild
- Department of Diabetes and Endocrinology, Women's and Children's Hospital, North Adelaide, 5066, South Australia, Australia
| | | | | | - Belinda George
- Department of Endocrinology, St. John's Medical College Hospital, Bengaluru, India
| | - Evelien F Gevers
- Centre for Endocrinology, William Harvey Research institute, Queen Mary University of London, London, UK
| | - Erin Greenup
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Pediatric Endocrinology, Department of Pediatrics, Orlando Health Arnold Palmer Hospital for Children, Orlando, FL, USA
| | - Annette Hackenberg
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Zita Halász
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Bianka Heinrich
- Department of Neuropediatrics, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Anna C Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tony Huynh
- Department of Endocrinology & Diabetes, Queensland Children's Hospital, South Brisbane, Queensland, Australia
| | - Amber R Isaza
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anna Klosowska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology and Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - David A Koolen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Heiko Krude
- Institute of Experimental Paediatric Endocrinology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Abhishek Kulkarni
- Department of Paediatric Endocrinology, SRCC Children's Hospital, Mumbai, India
| | - Alexander Laemmle
- Institute of Clinical Chemistry and Department of Pediatrics, Inselspital, University Hospital Bern, Bern, Switzerland
| | - Stephen H LaFranchi
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Amy Lawson-Yuen
- Genomics Institute Mary Bridge Children's Hospital, MultiCare Health System, Tacoma, WA, USA
- Department of Genetics, Kaiser Permanente Washington, Seattle, WA, USA
| | - Jan Lebl
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Selmar Leeuwenburgh
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Michaela Linder-Lucht
- Division of Neuropediatrics and Muscular Disorders, Department of Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Anna López Martí
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Cláudia F Lorea
- Teaching Hospital of Universidade Federal de Pelotas, Pelotas, Brazil
- Federal University of Rio Grande do Sul, Porto Alegre-RS, Brazil
| | - Charles M Lourenço
- National Reference Center for Rare Diseases, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
- Personalized Medicine area -Special Education Sector at DLE/Grupo Pardini, Rio de Janeiro, Brazil
| | - Roelineke J Lunsing
- Department of Child Neurology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Greta Lyons
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jana Krenek Malikova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Edna E Mancilla
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kenneth L McCormick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anne McGowan
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Veronica Mericq
- Institute of Maternal and Child Research, University of Chile, Santiago, Chile, Department of Pediatrics, Clinica Las Condes, Santiago, Chile
| | - Felipe Monti Lora
- Pediatric Endocrinology Group, Sabara Children's Hospital, São Paulo, Brazil
| | - Carla Moran
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | | | - Lindsey E Nicol
- Department of Pediatrics, Doernbecher Children's Hospital, Oregon Health & Sciences University, Portland, OR, USA
| | - Isabelle Oliver-Petit
- Department of Paediatric Endocrinology and Genetics, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Laura Paone
- Endocrinology and Diabetology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Praveen G Paul
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Michel Polak
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Francesco Porta
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Fabiano O Poswar
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Christina Reinauer
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Medical Faculty, Dusseldorf, Germany
| | - Klara Rozenkova
- Department of Paediatrics, Second Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Rowen Seckold
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Tuba Seven Menevse
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Peter Simm
- Royal Children's Hospital/University of Melbourne, Parkville, Australia
| | - Anna Simon
- Department of Paediatrics, Christian Medical College, Vellore, India
| | - Yogen Singh
- Department of Paediatric Cardiology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Pediatrics, University of California - UC Davis Children's Hospital, Sacramento, CA, USA
| | - Marco Spada
- Department of Paediatrics, AOU Città della Salute e della Scienza di Torino, University of Torino, Turin, Italy
| | - Milou A M Stals
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Merel T Stegenga
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Athanasia Stoupa
- Paediatric Endocrinology, Diabetology and Gynaecology, Department, Necker Children's University Hospital, Imagine Institute Affiliate, Université de Paris Cité, Paris, France
| | - Gopinath M Subramanian
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Lilla Szeifert
- Pediatric Center, Semmelweis University Budapest, Budapest, Hungary
| | - Davide Tonduti
- Child Neurology Unit - C.O.A.L.A. (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Milano, Italy
- Department of Clinical and Biomedical Science, Università degli Studi di Milano, Milano, Italy
| | - Serap Turan
- Marmara University School of Medicine Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Joel Vanderniet
- John Hunter Children's Hospital, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Adri van der Walt
- Private paediatric Neurology practice Dr A van der Walt, Durbanville, South Africa
| | | | | | - Jolanta Wierzba
- Department of Internal and Pediatric Nursing, Institute of Nursing and Midwifery, Medical University of Gdańsk, Gdańsk, Poland
| | - Marie-Claire Y de Wit
- Department of Paediatric Neurology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Michael Wurm
- University Children's Hospital Regensburg (KUNO), University of Regensburg, Campus St. Hedwig, Regensburg, Germany
| | - Federica Zibordi
- Child Neurology Unit, Fondazione IRCCS, Istituto Neurologico Carlo Besta, Milan, Italy
| | - Amnon Zung
- Pediatric Endocrinology Unit, Kaplan Medical center, Rehovot and the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitash Zwaveling-Soonawala
- Emma Children's Hospital, Department of Paediatric Endocrinology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marcel E Meima
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Debora S Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Juan P Nicola
- Department of Clinical Biochemistry (CIBICI-CONICET), Faculty of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Chi-Hua Chen
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, USA
| | - Marco Medici
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - W Edward Visser
- Academic Center for Thyroid Diseases, Department of Internal Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|