1
|
Akçeşme B, Hekimoğlu H, Chirasani VR, İş Ş, Atmaca HN, Waldern JM, Ramos SBV. Identification of deleterious non-synonymous single nucleotide polymorphisms in the mRNA decay activator ZFP36L2. RNA Biol 2025; 22:1-15. [PMID: 39668715 DOI: 10.1080/15476286.2024.2437590] [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] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
More than 4,000 single nucleotide polymorphisms (SNP) variants have been identified in the human ZFP36L2 gene, however only a few have been studied in the context of protein function. The tandem zinc finger domain of ZFP36L2, an RNA binding protein, is the functional domain that binds to its target mRNAs. This protein/RNA interaction triggers mRNA degradation, controlling gene expression. We identified 32 non-synonymous SNPs (nsSNPs) in the tandem zinc finger domain of ZFP36L2 that could have possible deleterious impacts in humans. Using different bioinformatic strategies, we prioritized five among these 32 nsSNPs, namely rs375096815, rs1183688047, rs1214015428, rs1215671792 and rs920398592 to be validated. When we experimentally tested the functionality of these protein variants using gel shift assays, all five (Y154H, R160W, R184C, G204D, and C206F) resulted in a dramatic reduction in RNA binding compared to the WT protein. To understand the mechanistic effect of these variants on the protein/RNA interaction, we employed DUET, DynaMut and PyMOL to investigate structural changes in the protein. Additionally, we conducted Molecular Docking and Molecular Dynamics Simulations to fine tune the active behaviour of this biomolecular system at an atomic level. Our results propose atomic explanations for the impact of each of these five genetic variants identified.
Collapse
Affiliation(s)
- Betül Akçeşme
- Program of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Ilidža/Sarajevo, Bosnia and Herzegovina
- Hamidiye School of Medicine, Department of Basic Medical Sciences, Division of Medical Biology, University of Health Sciences, Üsküdar/İstanbul, Turkey
| | - Hilal Hekimoğlu
- Institute of Health Sciences, İstanbul University, Fatih/İstanbul, Turkey
| | - Venkat R Chirasani
- Biochemistry and Biophysics Department, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Biochemistry and Biophysics Department, R. L. Juliano Structural Bioinformatics Core, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Şeyma İş
- Hamidiye School of Medicine, Department of Basic Medical Sciences, Division of Medical Biology, University of Health Sciences, Üsküdar/İstanbul, Turkey
- Department of Molecular Biotechnology, Division of Bioinformatics, Turkish-German University, Beykoz/İstanbul, Turkey
| | - Habibe Nur Atmaca
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayıs University, Atakum/Samsun, Turkey
| | - Justin M Waldern
- Biology Department, University of North Carolina, Chapel Hill, NC, USA
| | - Silvia B V Ramos
- Biochemistry and Biophysics Department, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Ak B, Akısü M, Durmaz A, Yalaz M, Terek D, Sönmezler E, Oktay Y, Akın H, Aykut A. Expanding the genetic spectrum of short rib polydactyly syndrome: Novel DYNC2H1 variants and functional insights. Bone 2025; 197:117511. [PMID: 40339774 DOI: 10.1016/j.bone.2025.117511] [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: 02/11/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
INTRODUCTION Short rib polydactyly syndrome (SRPS), with or without polydactyly, also known as Verma-Naumoff/Saldino-Noonan syndrome, is a type of skeletal ciliopathy. Initially, variants in the IFT80 gene were implicated; however, approximately half of the SRPS cases are associated with variants in the DYNC2H1 gene. Additionally, digenic variants involving DYNC2H1 and NEK1 can contribute to the syndrome. MATERIALS AND METHODS This case report describes a male patient presenting with characteristic SRPS features, including a constricted thorax and shortened limbs. Exome sequencing was performed to identify causative variants, followed by functional analyses to assess the pathogenicity of the identified variants, including a synonymous variant. RESULTS Exome sequencing identified compound heterozygous variants in the DYNC2H1 gene: a novel missense variant c.6439G>T p.(Asp2147Tyr) and a synonymous variant c.6477G>A p.(Gln2159=). Functional analyses confirmed that the synonymous variant triggers nonsense-mediated decay of the affected allele. CONCLUSION This study expands the spectrum of DYNC2H1 variants associated with SRPS and emphasizes the importance of functional analyses in genetic diagnostics. Demonstrating pathogenicity for a synonymous variant highlights the necessity for comprehensive variant assessments to improve diagnostic accuracy and enable early intervention. These findings have significant implications for molecular diagnostics and personalized therapy strategies in skeletal ciliopathies.
Collapse
Affiliation(s)
- Bilgesu Ak
- Department of Medical Genetics, Ege University Hospital, Izmir, Turkey
| | - Mete Akısü
- Department of Neonatology, Ege University Hospital, Izmir, Turkey.
| | - Asude Durmaz
- Department of Medical Genetics, Ege University Hospital, Izmir, Turkey.
| | - Mehmet Yalaz
- Department of Neonatology, Ege University Hospital, Izmir, Turkey.
| | - Demet Terek
- Department of Neonatology, Ege University Hospital, Izmir, Turkey.
| | | | - Yavuz Oktay
- Izmir Biomedicine and Genome Center, Izmir, Turkey.
| | - Haluk Akın
- Department of Medical Genetics, Ege University Hospital, Izmir, Turkey.
| | - Ayça Aykut
- Department of Medical Genetics, Ege University Hospital, Izmir, Turkey.
| |
Collapse
|
3
|
Yadav AJ, Padhi AK. Synergizing multiresolution simulations, interface redesign, and hotspot mapping to decipher pathogenic mutation-driven structural modulation in VCP. Comput Biol Med 2025; 194:110560. [PMID: 40516449 DOI: 10.1016/j.compbiomed.2025.110560] [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: 05/16/2025] [Revised: 06/01/2025] [Accepted: 06/08/2025] [Indexed: 06/16/2025]
Abstract
Valosin-containing protein (VCP/p97), a pivotal AAA+ ATPase, orchestrates proteostasis via ER-associated degradation (ERAD), ubiquitin-mediated proteolysis, and organelle surveillance. Pathogenic missense mutations, notably Arg95Gly (R95G) within the evolutionarily conserved double-ψ β-barrel (DPBB) of its N-terminal domain, are implicated in proteinopathies including IBMPFD and ALS. To decode the structural-dynamics perturbations underpinning R95G-driven dysfunction, we integrated AlphaFold3-based modeling, protein-peptide docking, and multiscale enhanced-sampling molecular dynamics (MD) simulations-spanning 1.2 μs all-atom, 12 μs coarse-grained, and umbrella sampling regimes. Our findings reveal that R95G disrupts the β-barrel integrity, destabilizes long-range domain coupling, and engenders conformational heterogeneity deleterious to gp78 cofactor recruitment. Free-energy landscapes of the mutant highlight enthalpically disfavored, low-occupancy binding conformers, corroborated by MM/PBSA-based end-state binding free energy and potential of mean force (PMF) analyses, which indicate impaired binding thermodynamics. Interface hotspot mapping pinpoints dynamic perturbations at critical residues that propagate allosteric decoupling and morphological distortion of the binding interface. Collectively, our results delineate a mechanistic cascade-from local β-barrel destabilization to global interaction network disruption-underlying VCP's functional impairment in disease states. This work provides a computationally derived structural framework to inform targeted biophysical validation and the rational design of therapeutic strategies aimed at rescuing VCP function in IBMPFD and ALS.
Collapse
Affiliation(s)
- Amar Jeet Yadav
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India
| | - Aditya K Padhi
- Laboratory for Computational Biology & Biomolecular Design, School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
4
|
Akter S, Fuad M, Mahmud Z, Tamanna S, Sayem M, Raj KH, Howlader MZH. Comprehensive in silico characterization of nonsynonymous SNPs in the human ezrin (EZR) gene and their role in disease pathogenesis. Biochem Biophys Rep 2025; 42:101972. [PMID: 40129965 PMCID: PMC11930600 DOI: 10.1016/j.bbrep.2025.101972] [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: 10/17/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
Ezrin (EZR) is a crucial linker between the actin cytoskeleton and the plasma membrane. It interacts with proteins involved in cancer-related signaling pathways. To assess the impact of nonsynonymous single nucleotide polymorphisms (nsSNPs) on EZR structure and function, we employed bioinformatics tools (SIFT, PolyPhen-2, PROVEAN, PhD-SNP, SNPs&GO, SuSPect, and FATHMM) and identified deleterious variants. Stability analyses using MUpro, mCSM, I-Mutant 2.0, and DynaMut2 revealed six destabilizing nsSNPs (F240S, H288D, I248T, L59Q, L125S, and L225P). Structural modeling using HOPE, MutPred2, AlphaFold, Swiss-Model, and protein-protein docking using HADDOCK 2.4 assessed the impact on the EZR-EBP50 complex. Binding free energy calculations, salt bridge analysis, and interface residue mapping further confirmed that the L225P, F240S, and I248T mutations significantly impaired EZR-EBP50 interaction, potentially disrupting key signaling pathways. Molecular dynamics simulations indicated that mutant EZR proteins exhibited reduced stability, flexibility, and hydrogen bonding. This first comprehensive in silico analysis of EZR highlights pathogenic nsSNPs that may contribute to disease progression. These findings provide a foundation for experimental validation and may inform targeted therapies for EZR-related pathologies.
Collapse
Affiliation(s)
| | | | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Sonia Tamanna
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Mohammad Sayem
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Khalid Hasan Raj
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | |
Collapse
|
5
|
Rajendran S, Surabhi RP, Kumar AS, Gopinath P, Kanakaveti V, Shanmugasundaram G, Michael Gromiha M, Rayala SK, Venkatraman G. P21-Activated Kinase 1 (PAK1) Modulates Therapeutic Response to Ionizing Radiation in Head and Neck Squamous Cell Carcinoma Cells. Mol Carcinog 2025; 64:970-984. [PMID: 40099538 DOI: 10.1002/mc.23902] [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: 08/26/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) continues to be a formidable epithelial malignancy characterized by late-stage detection and recurrence impacting survival. P21-activated kinase-1 (PAK1) was reported to be overexpressed in head and neck cancers and activated by ionizing radiation (IR), affecting treatment outcomes. Present investigations revealed that PAK1 silencing on HNSCC cells reverted the aggressive phenotype and showed impaired DNA damage repair upon IR exposure. Further HNSCC cells were resistant to IR up to 30 Gy with elevated pPAK1 levels. Radiation-resistant (RR) HNSCC cells expressed radiation-resistant markers, namely MRE-11 and NME-1; stemness markers-OCT4 and SOX2; and EMT & metastasis markers-vimentin, snail, and α-smooth muscle actin (α-SMA). In addition, HNSCC RR cells showed increased levels of DNA damage response protein H2AX, indicative of an aggressive phenotype with an augmented DNA repair machinery and a potential target for inhibition. Since H2AX appears to be a mechanistic hub for PAK1-induced radiation resistance, using in silico methods, peptides were designed, and the PL-8 peptide was chosen to target the phosphorylation of H2AX, which could enhance the sensitivity to IR and push the cells to radiation-induced cell death. PL-8 peptide inhibited H2AX phosphorylation on HNSCC cells and triggered radiation-induced cell death as determined by functional assays. The present study reveals PAK1 induced in HNSCC cells by IR and causes resistance by enhancing DNA damage response mediated through γH2AX. To counteract this complex molecular interplay, we propose inhibiting γH2AX formation & silencing PAK1 appears to be a probable way forward in HNSCC.
Collapse
Affiliation(s)
- Swetha Rajendran
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - Rohan Prasad Surabhi
- Department of Human Genetics, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu, India
| | - A Satheesh Kumar
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Prarthana Gopinath
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Vishnupriya Kanakaveti
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, California, USA
| | | | - M Michael Gromiha
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Guindy, Chennai, Tamil Nadu, India
| | - Ganesh Venkatraman
- Department of Bio-Medical Sciences, School of Bio Sciences & Technology, Vellore Institute of Technology Vellore, Vellore, Tamil Nadu, India
| |
Collapse
|
6
|
Rasmussen DK, Sun YJ, Franco JA, Kumar A, Vu JT, Bassuk AG, Mahajan VB. Structure-function analysis of CNGA3-associated achromatopsia patient variants complements clinical genomics in pathogenicity determination. Orphanet J Rare Dis 2025; 20:261. [PMID: 40448196 DOI: 10.1186/s13023-025-03792-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Achromatopsia is an autosomal recessive genetic disease, and 95% of achromatopsia patients carry pathogenic mutations in the CNGA3 and CNGB3 genes. Once translated, these genes function together by forming a cone photoreceptor CNG channel protein complex. RESULTS There are 150 CNGA3 missense variants reported in achromatopsia patients, but the pathogenicity of 103 variants remains unknown due to inconclusive genetic information. Here, we present clinical features of a novel CNGA3 variant in an achromatopsia patient and demonstrate its pathogenicity by a three-dimensional (3D) proteoform-based structure-function analysis. We first identified six proteotypic groups using 47 pathogenic missense variants with distinctive functional consequences by mapping their spatial proximity in a 3D protein structure. This meta-analysis was further applied to 103 missense variants of unknown significance (VUS) found in patients with achromatopsia. Strikingly, 86.4% of VUS had similar/identical functional consequence to nearby pathogenic variants, which suggested their likely pathogenicity and potential molecular pathology. The distinct proteotypic consequence of CNGA3 mutants shown in our analysis strongly supported the notion that gene supplementation may be the most widely applicable therapeutic option for CNGA3-associated achromatopsia patients. CONCLUSION Thus, proteoform-based analysis can be a valuable approach for assessing novel variants and complement clinical genomics in its utilization.
Collapse
Affiliation(s)
- Ditte K Rasmussen
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Young Joo Sun
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Joel A Franco
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Aarushi Kumar
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Jennifer T Vu
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Alexander G Bassuk
- Departments of Pediatrics and Neurology and The Iowa Neuroscience Institute (INI), University of Iowa, Iowa City, IA, USA
| | - Vinit B Mahajan
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, USA.
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
7
|
Jiang Y, Huang S, Chen HF. ActiMut-XGB: Predicting thermodynamic stability of point mutations for CALB with protein language model. Int J Biol Macromol 2025:144609. [PMID: 40414395 DOI: 10.1016/j.ijbiomac.2025.144609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/13/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Predicting the functional impact of single-point mutations on protein residual activity, especially after high-temperature incubation, is critical in protein engineering. We present an innovative machine learning model based on eXtreme Gradient Boosting that leverages protein sequence data to predict thermostability, circumventing the need for three-dimensional structural information. Our model integrates features from the ESM2 language model, physicochemical properties, evolutionary features, and positional features. A key advancement is the use of transfer learning with thermal stability data from various proteins, which enhances prediction accuracy and generalizability. To fine-tune and validate the model, we used experimental data from Candida antarctica lipase B single-point mutants, a widely studied enzyme in biocatalysis and industrial applications. Despite potential limitations of Gibbs free energy values in capturing all factors influencing thermostability, our model represents a significant improvement over traditional approaches, providing valuable insights for protein engineering, enzyme optimization, and therapeutic protein development.
Collapse
Affiliation(s)
- Yuxin Jiang
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuai Huang
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hai-Feng Chen
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
8
|
Ambrosini G, Floriani F, Manni V, Dando I, Montioli R. Biochemical investigation of pathogenic missense mutations of human 4-amino butyrate aminotransferase towards the understanding of the molecular pathogenesis of GABA transaminase deficiency. Mol Genet Metab 2025; 145:109149. [PMID: 40414180 DOI: 10.1016/j.ymgme.2025.109149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Gamma-amino butyrate aminotransferase (GABA-AT or ABAT) is a pyridoxal 5'-phosphate (PLP)-dependent enzyme that catalyzes the conversion of GABA and α-ketoglutarate into succinic semialdehyde and L-glutamate. In humans, the primary physiological role of GABA-AT is to control the level of GABA in neuronal tissues. Mutations on ABAT gene are associated to GABA-AT deficiency, an ultra-rare autosomal recessive disorder characterized by accelerated linear growth, severe psychomotor retardation, seizures, hypotonia, and hyperreflexia. So far, several missense pathogenic mutations of GABA-AT have been identified; however, their molecular effects at protein level have been poorly investigated. In this work a biochemical characterization of 10 pathogenic variants of human GABA-AT was carried out by expressing the protein in HEK-293 cells. Moreover, in-silico analyses of the variants were performed to corroborate the experimental findings. Altogether, the data obtained on protein expression level, GABA transaminase activity, and the predicted structural impact allowed us to classify the variants into three distinct groups, such as: (i) variants with strong structural and catalytic defects (p.P152S, p.L211F, and p.L478P); (ii) variants characterized mainly by a strong catalytic defect (p.R220K, p.Q296H, and p.R377W); (iii) variants exhibiting moderate structural and catalytic defects maintaining substantial transaminase activity (p.R92Q, p.F213C, p.G465D, and p.G465R). Based on these results, we provide a picture of the molecular defects of different GABA-AT pathogenic variants with the aim of gaining insights into the enzymatic phenotypes in GABA-AT deficiency.
Collapse
Affiliation(s)
- Giulia Ambrosini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Fulvio Floriani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Vittoria Manni
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Riccardo Montioli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy.
| |
Collapse
|
9
|
Chentoufi FE, Redouane S, Barakat A, Benrahma H, Charoute H. Computational study of the potential impact of WHRN protein missense SNPs on WHRN-MYO15A protein complex interaction and their association with Usher syndrome. J Biomol Struct Dyn 2025:1-26. [PMID: 40389825 DOI: 10.1080/07391102.2025.2507152] [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: 09/23/2024] [Accepted: 04/11/2025] [Indexed: 05/21/2025]
Abstract
Usher syndrome is a rare genetic condition characterized by both hearing and vision impairment that occurs through mutations of multiple genes, including WHRN and MYO15A. In this computational work, we intend to explore how missense SNPs within the WHRN protein affect its interaction with the MYO15A protein, a crucial component of the Usher interactome. Therefore, the identification of missense SNPs that has a potential effect on the function of the WHRN protein was realized using various computational prediction tools, including VEP, SIFT, PolyPhen-2, CADD, REVEL, and Mutation Assessor. Further evaluation of the stability of mutated proteins was conducted through SDM2, MCSM, DeepDDG and CUP-SAT. We used ConSurf web server to identify conserved regions in the WHRN protein. Yasara and Haddock analysis tools were used to minimize the energy of protein 3D structures and to dock protein-protein complexes, respectively. and then the binding energy of the complexes was calculated through PRODIGY. Mutation pathogenicity prediction tools showed that in total, 18 missense SNPs, predicted as deleterious. However, a comprehensive analysis revealed that only SIX single nucleotide polymorphisms were predicted to be the most deleterious with high conservation and less stability. Furthermore, we conducted molecular dynamics analysis to fully comprehend the impact of these variations on the dynamic behavior of the WHRN-MYO15A protein complex, which revealed significant insights into the destabilizing effects of the deleterious SNPs impacting the protein's binding affinity and stability that occurs during the binding process of the WHRN-MYO15A protein complex.
Collapse
Affiliation(s)
- Fatima Ezzahra Chentoufi
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
- Interdisciplinary Laboratory of Biotechnology and Health, Mohammed VI Higher Institute of Biosciences and Biotechnology, Mohammed VI University of sciences and Health (UM6SS), Casablanca, Morocco
| | - Salaheddine Redouane
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Houda Benrahma
- Interdisciplinary Laboratory of Biotechnology and Health, Mohammed VI Higher Institute of Biosciences and Biotechnology, Mohammed VI University of sciences and Health (UM6SS), Casablanca, Morocco
| | - Hicham Charoute
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
10
|
Shapovalov I, Rimal P, Poudel P, Lewtas V, Bell M, Panday SK, Laight BJ, Harper D, Grieve S, Baillie GS, Nouri K, Davies PL, Alexov E, Greer PA. Quantification and structure-function analysis of calpain-1 and calpain-2 protease subunit interactions. J Biol Chem 2025:110243. [PMID: 40383147 DOI: 10.1016/j.jbc.2025.110243] [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: 02/10/2025] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025] Open
Abstract
Calpain-1 and calpain-2 are heterodimeric proteases consisting of a common small regulatory subunit CAPNS1 and a large catalytic subunit, CAPN1 or CAPN2, respectively. These calpains have emerged as potential therapeutic targets in cancer and other diseases through their roles in cell signaling pathways affecting sensitivity to chemotherapeutic and targeted drugs, and in promoting metastasis. While inhibition of calpains has the potential to provide therapeutic benefit to cancer patients, there are currently no clinically approved active site directed drugs that specifically and effectively inhibit them. However, the structures of calpain-1 and calpain-2 make them susceptible to allosteric inhibition aimed at interfering with heterodimerization of the catalytic and regulatory subunits, which is necessary for stability and proteolytic activity. Split-Nanoluciferase biosensors were generated to quantify the protein-protein interactions (PPIs) between the calcium-binding penta-EF hand (PEF) domains of CAPN1 or CAPN2 and CAPNS1. These biosensors were used to quantify the heterodimer dissociation constants (KD) of calpain-1 and calpain-2, estimated at 185 nM and 509 nM, respectively, in the presence of 5 mM Ca2+; and 362 nM and 1651 nM, respectively, in the presence of Mg2+. The half-maximal Ca2+ concentrations supporting these PPIs for calpain-1 and calpain-2 were 59.9 μM and 940.8 μM, respectively. Molecular modeling, based on the crystal structure of calpain-2, was used to predict 20 residues of the PEF domains that contribute to heterodimerization. Individual point mutation of CAPNS1 at Q263 reduced the catalytic activity of calpain-2 to 51.0 ± 6.4 % in live cells.
Collapse
Affiliation(s)
- Ivan Shapovalov
- Dept Pathology and Molecular Medicine, School of Medicine, Queen's University, Kingston, ON, Canada; Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Prawin Rimal
- Department of Physics, College of Science, Clemson University, Clemson, SC, USA
| | - Pitambar Poudel
- Department of Physics, College of Science, Clemson University, Clemson, SC, USA
| | - Victoria Lewtas
- Dept Pathology and Molecular Medicine, School of Medicine, Queen's University, Kingston, ON, Canada; Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Mathias Bell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | | | - Brian J Laight
- Dept Pathology and Molecular Medicine, School of Medicine, Queen's University, Kingston, ON, Canada; Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Danielle Harper
- Dept Pathology and Molecular Medicine, School of Medicine, Queen's University, Kingston, ON, Canada; Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - Stacy Grieve
- Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, UK
| | - Kazem Nouri
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Peter L Davies
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Emil Alexov
- Department of Physics, College of Science, Clemson University, Clemson, SC, USA
| | - Peter A Greer
- Dept Pathology and Molecular Medicine, School of Medicine, Queen's University, Kingston, ON, Canada; Division of Cancer Biology and Genetics, Sinclair Cancer Research Institute, Queen's University, Kingston, ON, Canada
| |
Collapse
|
11
|
Sangeet S, Sinha A, Nair MB, Mahata A, Sarkar R, Roy S. EVOLVE: A Web Platform for AI-Based Protein Mutation Prediction and Evolutionary Phase Exploration. J Chem Inf Model 2025; 65:4293-4310. [PMID: 40309917 DOI: 10.1021/acs.jcim.5c00026] [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: 05/02/2025]
Abstract
While predicting structure-function relationships from sequence data is fundamental in biophysical chemistry, identifying prospective single-point and collective mutation sites in proteins can help us stay ahead in understanding their potential effects on protein structure and function. Addressing the challenges of large sequence-space analysis, we present EVOLVE, a web tool enabling researchers to explore prospective mutation sites and their collective behavior. EVOLVE integrates a statistical mechanics-guided machine learning algorithms to predict probable mutational sites, with statistical mechanics calculating mutational entropy to accurately identify mutational hotspots. Validation against a number of viral protein sequences confirms its ability to predict mutations and their functional consequences. By leveraging statistical mechanics of phase transition concept, EVOLVE also quantifies mutational entropy fluctuations, offering a quantitative foundation for identifying Variants of Concern (VOC) or Variants under Monitoring (VUM) as per World Health Organization (WHO) guidelines. EVOLVE streamlines data upload and analysis with a user-friendly interface and comprehensive tutorials. Access EVOLVE free at https://evolve-iiserkol.com.
Collapse
Affiliation(s)
- Satyam Sangeet
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
- School of Physics, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anushree Sinha
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Madhav B Nair
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Arpita Mahata
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Raju Sarkar
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| | - Susmita Roy
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, West Bengal 741246, India
| |
Collapse
|
12
|
Mondal S, Shrivastava P, Mehra R. Computing pathogenicity of mutations in human cytochrome P450 superfamily. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141078. [PMID: 40349948 DOI: 10.1016/j.bbapap.2025.141078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Cytochrome P450 (CYPs) are crucial heme-containing enzymes that metabolize drugs and endogenous compounds. In humans, 57 CYP isoforms have been identified, with over 200 mutations linked to severe disorders. Our comprehensive computational study assessed the reason for the pathogenicity of mutations by comparing pathogenic and non-pathogenic variants. We analyzed 25,94,151 mutations across 26 CYP structures using structure- and sequence-based methods, revealing a meaningful stability pattern: non-pathogenic > all > pathogenic mutation datasets. Notably, pathogenic mutations were predominantly buried within CYP structures, indicating a higher potential for pathogenesis. We identified three key amino acid properties affected by mutations: Gibbs free energy, isoelectric point, and volume. Furthermore, diseased mutations significantly reduced positive residue content, particularly due to arginine mutations, which directly influenced the isoelectric point. Our findings indicate a greater likelihood of pathogenic mutations occurring at conserved sites, disrupting CYP function. A higher frequency of pathogenic mutations was observed in heme sites, primarily involving arginine, which may interfere with arginine-heme interactions. Molecular docking revealed a differential binding of heme in wild-type and pathogenic CYPs. This study provides a foundational analysis of mutation effects across multiple CYPs. It models the chemical basis of CYP-related pathogenicity, facilitating the development of a semi-quantitative disease prediction model.
Collapse
Affiliation(s)
- Somnath Mondal
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Pranchal Shrivastava
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India
| | - Rukmankesh Mehra
- Department of Chemistry, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India; Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Durg 491002, Chhattisgarh, India.
| |
Collapse
|
13
|
Reddy HP, Keren-Raifman T, Tabak G, Dascal N, Yakubovich D. Loss of expression and function of Gβγ by GNB1 encephalopathy-associated L95P mutation of the Gβ 1 subunit. Front Pharmacol 2025; 16:1592012. [PMID: 40417225 PMCID: PMC12098346 DOI: 10.3389/fphar.2025.1592012] [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: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 05/27/2025] Open
Abstract
Background G-proteins areindispensable regulators of cellular signaling, with G-protein-gated inwardly rectifying potassium channels (GIRK) as key effectors. GNB1 encephalopathy (GNB1E) is a congenital neurological syndrome resulting from mutations in the GNB1 gene, encoding the Gβ1 subunit of G-proteins trimer (Gαβγ). GNB1E manifests as a global developmental delay, accompanied by tonus disturbances, ataxia, and epilepsy. Methods We utilized the Xenopus laevis oocyte heterologous expression system to investigate the impact of the L95P mutation in Gβ1 (Gβ1-L95P) on the activation of neuronal GIRK channels GIRK2 and GIRK1/2. Mutant and wild-type (WT) Gβ1 RNAs were co-injected with RNAs encoding the Gγ2 and GIRK channel subunits. The expression levels of both Gβ1 and the channel proteins, as well as the channel activity, were systematically monitored. Additionally, rigid-body docking was used to model the GIRK1/2-Gβγ complex, evaluating L95P's effect on channel-Gβγ interaction, Gβγ stability, and Gβγ-effector affinity. Results . Gβ1-L95P exhibited reduced protein expression compared to WT. Even after RNA adjustments to restore comparable membrane localization, the mutant failed to effectively activate GIRK2 and GIRK1/2. Structural analysis revealed that L95 was not consistent in the Gβγ-effector interface. Thermodynamic calculations suggested that the mutation primarily destabilized Gβ1 and Gβ1-effector complex. Conclusion Gβ1-L95P leads to both reduced protein expression and impaired function in the GIRK-Gβγ interaction system. The later effect can be attributed to the changes associated with protein misfolding.
Collapse
Affiliation(s)
| | | | - Galit Tabak
- School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Nathan Dascal
- School of Medicine, Tel-Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, Israel
| | - Daniel Yakubovich
- The Adelson School of Medicine, Ariel University, Ariel, Israel
- Neonatology Department, Laniado Hospital, Netanya, Israel
| |
Collapse
|
14
|
Navapour L, Mogharrab N, Parvin A, Rezaei Arablouydareh S, Movahedpour A, Jebraeily M, Taheri-Anganeh M, Ghasemnejad-Berenji H. Identification of high-risk non-synonymous SNPs (nsSNPs) in DNAH1 and DNAH17 genes associated with male infertility: a bioinformatics analysis. J Appl Genet 2025; 66:333-346. [PMID: 38874855 DOI: 10.1007/s13353-024-00884-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
Male infertility is a significant reproductive issue affecting a considerable number of couples worldwide. While there are various causes of male infertility, genetic factors play a crucial role in its development. We focused on identifying and analyzing the high-risk nsSNPs in DNAH1 and DNAH17 genes, which encode proteins involved in sperm motility. A total of 20 nsSNPs for DNAH1 and 10 nsSNPs for DNAH17 were analyzed using various bioinformatics tools including SIFT, PolyPhen-2, CADD, PhD-SNPg, VEST-4, and MutPred2. As a result, V1287G, L2071R, R2356W, R3169C, R3229C, E3284K, R4096L, R4133C, and A4174T in DNAH1 gene and C1803Y, C1829Y, R1903C, and L3595P in DNAH17 gene were identified as high-risk nsSNPs. These nsSNPs were predicted to decrease protein stability, and almost all were found in highly conserved amino acid positions. Additionally, 4 nsSNPs were observed to alter post-translational modification status. Furthermore, the interaction network analysis revealed that DNAH1 and DNAH17 interact with DNAH2, DNAH3, DNAH5, DNAH7, DNAH8, DNAI2, DNAL1, CFAP70, DNAI3, DNAI4, ODAD1, and DNAI7, demonstrating the importance of DNAH1 and DNAH17 proteins in the overall functioning of the sperm motility machinery. Taken together, these findings revealed the detrimental effects of identified high-risk nsSNPs on protein structure and function and highlighted their potential relevance to male infertility. Further studies are warranted to validate these findings and to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Leila Navapour
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Navid Mogharrab
- Biophysics and Computational Biology Laboratory (BCBL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Ali Parvin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sahar Rezaei Arablouydareh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mohamad Jebraeily
- Department of Health Information Technology, School of Allied Medical Sciences, Urmia University of Medical Sciences, Urmia, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
15
|
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
|
16
|
Ahammed KS, Fasken MB, Corbett AH, van Hoof A. Humanized Saccharomyces cerevisiae provides a facile and effective tool to identify damaging human variants that cause exosomopathies. G3 (BETHESDA, MD.) 2025; 15:jkaf036. [PMID: 39982806 PMCID: PMC12005145 DOI: 10.1093/g3journal/jkaf036] [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: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/23/2025]
Abstract
The RNA exosome is an evolutionarily conserved, multiprotein complex that is the major RNase in 3' processing and degradation of a wide range of RNAs in eukaryotes. Single amino acid changes in RNA exosome subunits cause rare genetic diseases collectively called exosomopathies. However, distinguishing disease-causing variants from nonpathogenic ones remains challenging, and the mechanism by which these variants cause disease is largely unknown. Previous studies have employed a budding yeast model of RNA exosome-linked diseases that relies on mutating the orthologous yeast genes. Here, we develop a humanized yeast model of exosomopathies that allows us to unambiguously assess damaging effects of the exact patient variant in budding yeast. Individual replacement of the yeast subunits with corresponding mammalian orthologs identified 6 out of 9 noncatalytic core subunits of the budding yeast RNA exosome that can be replaced by a mammalian subunit, with 3 of the replacements supporting close to normal growth. Further analysis of the disease-associated variants utilizing the hybrid yeast/mammalian RNA exosome revealed functional defects caused by both previously characterized and uncharacterized variants of EXOSC2, EXOSC4, EXOSC7, and EXOSC9. Analysis of the protein levels of these variants indicates that a subset of the patient-derived variants causes reduced protein levels, while other variants are defective but are expressed as well as the reference allele, suggesting a more direct contribution of these residues to RNA exosome function. This humanized yeast model of exosomopathies provides a convenient and sensitive genetic tool to help distinguish damaging RNA exosome variants from benign variants. This disease model can be further exploited to uncover the underpinning mechanism of RNA exosome defects.
Collapse
Affiliation(s)
- Khondakar Sayef Ahammed
- Department of Microbiology and Molecular Genetics and MD Anderson UTHealth Houston Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Milo B Fasken
- Department of Biology, Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Anita H Corbett
- Department of Biology, Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Ambro van Hoof
- Department of Microbiology and Molecular Genetics and MD Anderson UTHealth Houston Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
17
|
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
|
18
|
Azevedo RDS, Santana H, Seus VR, Camargo AD, Werhli AV, Machado KDS, Cançado LJ, Quirino BF, Marins LF. Development of a β-glucosidase improved for glucose retroinhibition for cellulosic ethanol production: an integrated bioinformatics and genetic engineering approach. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2025; 18:44. [PMID: 40188331 PMCID: PMC11972475 DOI: 10.1186/s13068-025-02643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND The global energy crisis, driven by economic growth and the increasing demand for energy, highlights the urgency of searching for alternative energy sources to mitigate environmental pollution and climate change. β-Glucosidases act in the final step of the enzymatic hydrolysis of cellulose, cleaving the β-1,4-glycosidic bonds in cellobiose to produce second-generation ethanol. However, these enzymes are easily inhibited by glucose, their final product, which limits the production of this biofuel. Genetic engineering combined with bioinformatics tools can improve key enzymatic characteristics, such as catalytic activity and glucose tolerance, in a more precise, faster, and cost-effective manner compared to traditional methods. In this work, a variant of a β-glucosidase from the GH1 family, isolated from the microbial community of Amazonian soil (Brazil), with enhanced catalytic activity and improved for glucose retroinhibition, was developed. RESULTS Bioinformatics analyses suggested the substitution of tryptophan at position 404 with leucine. The produced variant (W404L) was expressed in Escherichia coli and showed activity 3.2 times higher in the presence of glucose than the non-mutated control. Moreover, the partially purified mutated variant of β-glucosidase exhibited a 26-fold increase in catalytic activity compared to the original form of the enzyme. The results confirmed that the mutation proposed by computational analyses had a significant impact on enzyme catalytic activity and glucose retroinhibition. CONCLUSIONS This new variant may become a promising alternative to reduce the costs of enzyme cocktails used in the hydrolysis of lignocellulosic biomass used as a raw material in the production of second-generation ethanol.
Collapse
Affiliation(s)
- Raíza Dos Santos Azevedo
- LEGENE - Research Group in Genetic Engineering and Biotechnology, Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil.
| | | | - Vinícius Rosa Seus
- Combi-Lab - Computational Biology Laboratory, Center for Computational Sciences (C3), Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Alex Dias Camargo
- Combi-Lab - Computational Biology Laboratory, Center for Computational Sciences (C3), Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Adriano Velasque Werhli
- Combi-Lab - Computational Biology Laboratory, Center for Computational Sciences (C3), Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Karina Dos Santos Machado
- Combi-Lab - Computational Biology Laboratory, Center for Computational Sciences (C3), Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | | | | | - Luis Fernando Marins
- LEGENE - Research Group in Genetic Engineering and Biotechnology, Laboratory of Molecular Biology, Institute of Biological Sciences, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| |
Collapse
|
19
|
Panda S, Rout M, Mishra S, Turuk J, Pati S, Dehury B. Molecular docking and MD simulations reveal protease inhibitors block the catalytic residues in Prp8 intein of Aspergillus fumigatus: a potential target for antimycotics. J Biomol Struct Dyn 2025; 43:3526-3541. [PMID: 38149850 DOI: 10.1080/07391102.2023.2298735] [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: 10/03/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
Resistance to azoles and amphotericin B especially in Aspergillus fumigatus is a growing concern towards the treatment of invasive fungal infection. At this critical juncture, intein splicing would be a productive, and innovative target to establish therapies against resistant strains. Intein splicing is the central event for the activation of host protein, essential for the growth and survival of various microorganisms including A. fumigatus. The splicing process is a four-step protease-like nucleophilic cascade. Thus, we hypothesise that protease inhibitors would successfully halt intein splicing and potentially restrict the growth of the aforementioned pathogen. Using Rosetta Fold and molecular dynamics simulations, we modelled Prp8 intein structure; resembling classic intein fold with horse shoe shaped splicing domain. To fully comprehend the active site of Afu Prp8 intein, C1, T62, H65, H818, N819 from intein sequences and S820, the first C-extein residue are selected. Molecular docking shows that two FDA-approved drugs, i.e. Lufotrelvir and Remdesivir triphosphate efficiently interact with Prp8 intein from the assortment of 212 protease inhibitors. MD simulation portrayed that Prp8 undergoes conformational change upon ligand binding, and inferred the molecular recognition and stability of the docked complexes. Per-residue decomposition analysis confirms the importance of F: block R802, V803, and Q807 binding pocket in intein splicing domain towards recognition of inhibitors, along with active site residues through strong hydrogen bonds and hydrophobic contacts. However, in vitro and in vivo assays are required to confirm the inhibitory action on Prp8 intein splicing; which may pave the way for the development of new antifungals for A. fumigatus.
Collapse
Affiliation(s)
- Sunita Panda
- Mycology Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Madhusmita Rout
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Sarbani Mishra
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Jyotirmayee Turuk
- Mycology Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Sanghamitra Pati
- Mycology Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Budheswar Dehury
- Bioinformatics Division, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| |
Collapse
|
20
|
Dissanayake UC, Roy A, Maghsoud Y, Polara S, Debnath T, Cisneros GA. Computational studies on the functional and structural impact of pathogenic mutations in enzymes. Protein Sci 2025; 34:e70081. [PMID: 40116283 PMCID: PMC11926659 DOI: 10.1002/pro.70081] [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: 11/08/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Enzymes are critical biological catalysts involved in maintaining the intricate balance of metabolic processes within living organisms. Mutations in enzymes can result in disruptions to their functionality that may lead to a range of diseases. This review focuses on computational studies that investigate the effects of disease-associated mutations in various enzymes. Through molecular dynamics simulations, multiscale calculations, and machine learning approaches, computational studies provide detailed insights into how mutations impact enzyme structure, dynamics, and catalytic activity. This review emphasizes the increasing impact of computational simulations in understanding molecular mechanisms behind enzyme (dis)function by highlighting the application of key computational methodologies to selected enzyme examples, aiding in the prediction of mutation effects and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Upeksha C. Dissanayake
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Arkanil Roy
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Yazdan Maghsoud
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Biochemistry and Molecular PharmacologyBaylor College of MedicineHoustonTexasUSA
| | - Sarthi Polara
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Tanay Debnath
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - G. Andrés Cisneros
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
21
|
Mobini S, Chizari M, Rismani E, Mafakher L, Sadrzadeh MJ, Vosough M. Targeting CD84 protein on myeloid-derived suppressor cells as a novel immunotherapy in solid tumors. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 261:108607. [PMID: 39847992 DOI: 10.1016/j.cmpb.2025.108607] [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: 09/23/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND AND OBJECTIVE Myeloid-derived suppressor cells (MDSCs) are a crucial and diverse group of cells found in the tumor microenvironment (TME) that facilitate progression, invasion, and metastasis within solid tumors. CD84, a homophilic adhesion molecule expressed on MDSCs, plays a critical role in their accumulation and function within the TME. This study aims to investigate the protein-protein interactions of CD84 using molecular dynamics simulations and to explore potential therapeutic strategies targeting these interactions. METHODS Through computational techniques, we generated highly potent mutated CD84 mini-proteins and peptides as antagonists with significantly higher affinity for CD84 to mimic the key features of the IgV-like domain of the protein. Additionally, we engineered an antibody capable of blocking CD84. Binding affinities were assessed using dissociation constant (Kd) calculations. RESULTS Data analysis shows that the Kd values for the designed peptides ranged from 10 to 100 times stronger than those of the natural CD84 interactions, indicating efficient inhibition of CD84 interactions. Additionally, mutagenesis of the Ig-like V domain of CD84 resulted in variants with improved binding stability, with a Gibbs free energy change (ΔΔG) indicating enhanced interaction potential. CONCLUSIONS This study provides insights into CD84 interactions and their implications for immunotherapy targeting MDSCs in solid tumors. However, experimental validation is necessary to confirm the findings of this study and evaluate peptide selectivity as potential molecular therapeutics.
Collapse
Affiliation(s)
- Saeed Mobini
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Milad Chizari
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Elham Rismani
- Molecular Medicine Department, Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Ladan Mafakher
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83 Stockholm, Sweden.
| |
Collapse
|
22
|
Srivastava A, Skopelitou D, Miao B, Giagiobbe S, Paramasivam N, Kumar A, Diquigiovanni C, Bonora E, Bandapalli OR, Försti A, Hemminki K. Prioritization of predisposition genes for familial non-medullary thyroid cancer by whole-genome sequencing. Eur J Endocrinol 2025; 192:398-407. [PMID: 40177881 DOI: 10.1093/ejendo/lvaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/05/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE Thyroid cancer (TC) is the most common endocrine malignancy, with 90%-95% of the cases representing non-medullary thyroid cancer (NMTC). Familial cases account only for a few of all cases and the underlying genetic causes are still poorly understood. METHODS We whole-genome sequenced affected and unaffected members of an Italian NMTC family and applied our in-house developed Familial Cancer Variant Prioritization Pipeline (FCVPPv2) which prioritized 12 coding variants. We refined this selection using the VarSome American College of Medical Genetics and Genomics (ACMG) implementation, SNAP2 predictions and further in silico scores. RESULTS We prioritized 4 possibly pathogenic variants in 4 genes including Ret proto-oncogene (RET), polypeptide N-acetylgalactosaminyltransferase 10 (GALNT10), ubinuclein-1 (UBN1), and prostaglandin I2 receptor (PTGIR). The role of RET point mutations in medullary thyroid carcinoma is well established. Similarly, somatic rearrangements of RET are known in papillary TC, a specific histotype of NMTC. In contrast to RET, no germline variants in PTGIR, GALNT10, or UBN1 have been linked to the development of TC to date. However, alterations in these genes have been shown to affect pathways related to cell proliferation, apoptosis, growth, and differentiation, as well as posttranslational modification and gene regulation. A thorough review of the available literature together with computational evidence supported the interpretation of the 4 shortlisted variants as possibly disease-causing in this family. CONCLUSIONS Our results implicate the first germline variant in RET in a family with NMTC as well as the first germline variants in PTGIR, GALNT10, and UBN1 in TC.
Collapse
Affiliation(s)
- Aayushi Srivastava
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Heidelberg University, Medical Faculty, Heidelberg 69120, Germany
| | - Diamanto Skopelitou
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Heidelberg University, Medical Faculty, Heidelberg 69120, Germany
| | - Beiping Miao
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Sara Giagiobbe
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg 69120, Germany
| | - Nagarajan Paramasivam
- Computational Oncology, Molecular Diagnostics Program, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Abhishek Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Clinical Cooperation Unit Molecular Hematology-Oncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Chiara Diquigiovanni
- Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40126, Italy
| | - Elena Bonora
- Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40126, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40126, Italy
| | - Obul Reddy Bandapalli
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Heidelberg University, Medical Faculty, Heidelberg 69120, Germany
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Hopp Children's Cancer Center (KiTZ), Heidelberg 69120, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg 69120, Germany
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, Pilsen 32300, Czech Republic
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| |
Collapse
|
23
|
Iqbal MW, Ahmad M, Shahab M, Sun X, Baig MM, Yu K, Dawoud TM, Bourhia M, Dabiellil F, Zheng G, Yuan Q. Exploring deleterious non-synonymous SNPs in FUT2 gene, and implications for norovirus susceptibility and gut microbiota composition. Sci Rep 2025; 15:10395. [PMID: 40140394 PMCID: PMC11947322 DOI: 10.1038/s41598-025-92220-4] [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: 11/05/2024] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Fucosyltransferase 2 (FUT2) gene has been extensively reported to play its role in potential gut microbiota changes and norovirus susceptibility. The normal activity of FUT2 has been found to be disrupted by non-synonymous single nucleotide polymorphisms (nsSNPs) in its gene. To explore the possible mutational changes and their deleterious effects, we employed state-of-the-art computational strategies. Firstly, nine widely-used bioinformatics tools were utilized for initial screening of possibly deleterious nsSNPs. Subsequently, the structural and functional effects of screened nsSNPs on FUT2 were evaluated by utilizing relevant computational tools. Following this, the two shortlisted nsSNPs, including G149S (rs200543547) and V196G (rs367923363), were further validated by their molecular docking with norovirus capsid protein, VP1. As compared to wild-type, the higher stability and lower binding energy scores of the both the mutants indicated their stable binding with VP1, which ultimately leads to norovirus implications. These docking results were further verified by a comprehensive computational approach, molecular dynamic simulation, which gave results in the form of lower RMSD, RMSF, RoG, and hydrogen bond values of both the mutants, depicted in relevant graphs. Overall, this research explores and validated the two FUT2 nsSNPs (G146S and V196G), which may possibly linked with the norovirus susceptibility and gut microbiota changes. Moreover, our findings highlights the value of computational strategies in mutational analysis and welcomes any further experimental validation.
Collapse
Affiliation(s)
- Muhammad Waleed Iqbal
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Muneer Ahmad
- College of Medicine and Bioinformation Engineering, Northeastern University, Shenyang, 110819, People's Republic of China
| | - Muhammad Shahab
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xinxiao Sun
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Mudassar Mehmood Baig
- Institute of Fundamental and Frontier Science, University of Electronic Science and Technology, Chengdu, 611731, People's Republic of China
| | - Kun Yu
- College of Medicine and Bioinformation Engineering, Northeastern University, Shenyang, 110819, People's Republic of China
| | - Turki M Dawoud
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| | - Fakhreldeen Dabiellil
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco.
- University of Bahr el Ghazal, Freedom Street, 91113, Wau, South Sudan.
| | - Guojun Zheng
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
24
|
Khalid M, Khalid Z, Abbasi SW, Gul A. Computational analysis on non-synonymous single nucleotide polymorphisms (nsSNPs) in L-type lectin receptor kinases (LECRK) protein in Arabidopsis Thaliana. J Biomol Struct Dyn 2025:1-7. [PMID: 40080787 DOI: 10.1080/07391102.2025.2477768] [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: 09/12/2024] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
L-type lectin receptor kinases (LECRK) plays a significant role in biotic and abiotic stress response against environmental stimuli in plants. In the Arabidopsis model plant, a total of 45 LECRK was identified but function elucidation is still unresolved. This study carried out a comprehensive analysis of the SNPs associated with L-type lectin protein and how these mutations affect the structure and function of the protein. The computational tools utilized covers both sequence and structure based analysis of the candidate SNPs. The evolutionary analysis identified the conserved residues that are either buried and structurally important or exposed means functionally important hence, can affect the stress response of the protein. A few of the significant mutations are identified M411I, S415C, W431C, A442S, L445F, Q389K, H458Y, and E651V are expected to damage the structure or function of the protein. Among them, the docking studies identified the mutants S415C and W431C as most crucial which can most likely disrupt the protein-protein interactions. Molecular dynamic simulation and principal component analysis further highlights the structural and functional changes in protein resulting by high risks mutations.
Collapse
Affiliation(s)
- Maria Khalid
- Department of Life Sciences, Health Services Academy (HAS), Islamabad, Pakistan
| | - Zoya Khalid
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Sumra Wajid Abbasi
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Alvina Gul
- Department of Agricultural Sciences and Technology, ASAB, NUST, Islamabad, Pakistan
| |
Collapse
|
25
|
Bezawork-Geleta A, Devereux CJ, Keenan SN, Lou J, Cho E, Nie S, De Souza DP, Narayana VK, Siddall NA, Rodrigues CHM, Portelli S, Zheng T, Nim HT, Ramialison M, Hime GR, Dodd GT, Hinde E, Ascher DB, Stroud DA, Watt MJ. Proximity proteomics reveals a mechanism of fatty acid transfer at lipid droplet-mitochondria- endoplasmic reticulum contact sites. Nat Commun 2025; 16:2135. [PMID: 40032835 PMCID: PMC11876333 DOI: 10.1038/s41467-025-57405-5] [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: 04/11/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Membrane contact sites between organelles are critical for the transfer of biomolecules. Lipid droplets store fatty acids and form contacts with mitochondria, which regulate fatty acid oxidation and adenosine triphosphate production. Protein compartmentalization at lipid droplet-mitochondria contact sites and their effects on biological processes are poorly described. Using proximity-dependent biotinylation methods, we identify 71 proteins at lipid droplet-mitochondria contact sites, including a multimeric complex containing extended synaptotagmin (ESYT) 1, ESYT2, and VAMP Associated Protein B and C (VAPB). High resolution imaging confirms localization of this complex at the interface of lipid droplet-mitochondria-endoplasmic reticulum where it likely transfers fatty acids to enable β-oxidation. Deletion of ESYT1, ESYT2 or VAPB limits lipid droplet-derived fatty acid oxidation, resulting in depletion of tricarboxylic acid cycle metabolites, remodeling of the cellular lipidome, and induction of lipotoxic stress. These findings were recapitulated in Esyt1 and Esyt2 deficient mice. Our study uncovers a fundamental mechanism that is required for lipid droplet-derived fatty acid oxidation and cellular lipid homeostasis, with implications for metabolic diseases and survival.
Collapse
Affiliation(s)
| | - Camille J Devereux
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Stacey N Keenan
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Jieqiong Lou
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform (BOMP), The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility (MMSPF), Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Vinod K Narayana
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Nicole A Siddall
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Carlos H M Rodrigues
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Stephanie Portelli
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Tenghao Zheng
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Hieu T Nim
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Gary R Hime
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Elizabeth Hinde
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - David A Stroud
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
26
|
Rodrigues CHM, Portelli S, Ascher DB. Exploring the effects of missense mutations on protein thermodynamics through structure-based approaches: findings from the CAGI6 challenges. Hum Genet 2025; 144:327-335. [PMID: 38227011 PMCID: PMC11976750 DOI: 10.1007/s00439-023-02623-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/18/2023] [Indexed: 01/17/2024]
Abstract
Missense mutations are known contributors to diverse genetic disorders, due to their subtle, single amino acid changes imparted on the resultant protein. Because of this, understanding the impact of these mutations on protein stability and function is crucial for unravelling disease mechanisms and developing targeted therapies. The Critical Assessment of Genome Interpretation (CAGI) provides a valuable platform for benchmarking state-of-the-art computational methods in predicting the impact of disease-related mutations on protein thermodynamics. Here we report the performance of our comprehensive platform of structure-based computational approaches to evaluate mutations impacting protein structure and function on 3 challenges from CAGI6: Calmodulin, MAPK1 and MAPK3. Our stability predictors have achieved correlations of up to 0.74 and AUCs of 1 when predicting changes in ΔΔG for MAPK1 and MAPK3, respectively, and AUC of up to 0.75 in the Calmodulin challenge. Overall, our study highlights the importance of structure-based approaches in understanding the effects of missense mutations on protein thermodynamics. The results obtained from the CAGI6 challenges contribute to the ongoing efforts to enhance our understanding of disease mechanisms and facilitate the development of personalised medicine approaches.
Collapse
Affiliation(s)
- Carlos H M Rodrigues
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
| | - David B Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia.
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
27
|
Pyankov IA, Gonay V, Stepanov YA, Shestun P, Kostareva AA, Uspenskaya MV, Petukhov MG, Kajava AV. A computational approach to predict the effects of missense mutations on protein amyloidogenicity: A case study in hereditary transthyretin cardiomyopathy. J Struct Biol 2025; 217:108176. [PMID: 39933599 DOI: 10.1016/j.jsb.2025.108176] [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: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
With many amyloidosis-associated missense mutations still unidentified and early diagnostic methods largely unavailable, there is an urgent need for a reliable computational approach to predict hereditary amyloidoses from gene sequencing data. Progress has been made in predicting amyloidosis-triggering sequences within intrinsically disordered regions. However, some diseases are caused by mutations in amyloidogenic regions within structured domains that must unfold for amyloid formation. Accurate prediction of amyloidogenic regions requires tools for detecting amyloidogenicity and assessing mutation effects on protein stability. We developed datasets of mutations linked to hereditary ATTR cardiomyopathy and others likely unrelated, evaluating TTR mutants with amyloidogenicity and stability predictors. Notably, the stability predictors consistently indicated that ATTR-related mutations tend to destabilize the TTR structure more than non-ATTR-associated mutations. Using these datasets and newly generated mutation features, we developed a machine learning model SDAM-TTR to predict mutations leading to ATTR cardiomyopathy.
Collapse
Affiliation(s)
- Ivan A Pyankov
- ITMO University, Chemical Engineering Center, Kronverksky Pr. 49, bldg. A, St. Petersburg 197101, Russian Federation; Department of Chemical Medicine, Institute of Chemistry, St. Petersburg State University, Russian Federation
| | - Valentin Gonay
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université de Montpellier 1919 Route de Mende, Montpellier, France; PROTERA SAS, 176 avenue Charles de Gaulle, 92522 Neuilly-sur-Seine Cedex, France
| | - Yaroslav A Stepanov
- ITMO University, Chemical Engineering Center, Kronverksky Pr. 49, bldg. A, St. Petersburg 197101, Russian Federation
| | - Pavel Shestun
- ITMO University, Chemical Engineering Center, Kronverksky Pr. 49, bldg. A, St. Petersburg 197101, Russian Federation
| | - Anna A Kostareva
- Almazov National Medical Research Centre, 2 Akkuratova street, St. Petersburg 197341, Russian Federation
| | - Mayya V Uspenskaya
- Department of Chemical Medicine, Institute of Chemistry, St. Petersburg State University, Russian Federation; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russian Federation
| | - Michael G Petukhov
- Petersburg Institute of Nuclear Physics, NRC Kurchatov Institute, Gatchina, Russian Federation; Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russian Federation
| | - Andrey V Kajava
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Université de Montpellier 1919 Route de Mende, Montpellier, France.
| |
Collapse
|
28
|
Panchal NK, Samdani P, Sengupta T, Prince SE. Computational Analysis of Non-synonymous SNPs in ATM Kinase: Structural Insights, Functional Implications, and Inhibitor Discovery. Mol Biotechnol 2025; 67:1201-1221. [PMID: 38489015 DOI: 10.1007/s12033-024-01120-x] [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: 10/11/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024]
Abstract
Ataxia telangiectasia-mutated (ATM) protein kinase, a key player in cellular integrity regulation, is known for its role in DNA damage response. This study investigates the broader impact of ATM on cellular processes and potential clinical manifestations arising from mutations, aiming to expand our understanding of ATM's diverse functions beyond conventional roles. The research employs a comprehensive set of computational techniques for a thorough analysis of ATM mutations. The mutation data are curated from dbSNP and HuVarBase databases. A meticulous assessment is conducted, considering factors such as deleterious effects, protein stability, oncogenic potential, and biophysical characteristics of the identified mutations. Conservation analysis, utilizing diverse computational tools, provides insights into the evolutionary significance of these mutations. Molecular docking and dynamic simulation analyses are carried out for selected mutations, investigating their interactions with Y2080D, AZD0156, and quercetin inhibitors to gauge potential therapeutic implications. Among the 419 mutations scrutinized, five (V1913C, Y2080D, L2656P, C2770G, and C2930G) are identified as both disease causing and protein destabilizing. The study reveals the oncogenic potential of these mutations, supported by findings from the COSMIC database. Notably, Y2080D is associated with haematopoietic and lymphoid cancers, while C2770G shows a correlation with squamous cell carcinomas. Molecular docking and dynamic simulation analyses highlight strong binding affinities of quercetin for Y2080D and AZD0156 for C2770G, suggesting potential therapeutic options. In summary, this computational analysis provides a comprehensive understanding of ATM mutations, revealing their potential implications in cellular integrity and cancer development. The study underscores the significance of Y2080D and C2770G mutations, offering valuable insights for future precision medicine targeting-specific ATM. Despite informative computational analyses, a significant research gap exists, necessitating essential in vitro and in vivo studies to validate the predicted effects of ATM mutations on protein structure and function.
Collapse
Affiliation(s)
- Nagesh Kishan Panchal
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| | - Poorva Samdani
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Tiasa Sengupta
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Sabina Evan Prince
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India.
| |
Collapse
|
29
|
Turina P, Petrosino M, Enriquez Sandoval CA, Novak L, Pasquo A, Alexov E, Alladin MA, Ascher DB, Babbi G, Bakolitsa C, Casadio R, Cheng J, Fariselli P, Folkman L, Kamandula A, Katsonis P, Li M, Li D, Lichtarge O, Mahmud S, Martelli PL, Pal D, Panday SK, Pires DEV, Portelli S, Pucci F, Rodrigues CHM, Rooman M, Savojardo C, Schwersensky M, Shen Y, Strokach AV, Sun Y, Woo J, Radivojac P, Brenner SE, Chiaraluce R, Consalvi V, Capriotti E. Assessing the predicted impact of single amino acid substitutions in MAPK proteins for CAGI6 challenges. Hum Genet 2025; 144:265-280. [PMID: 39976676 PMCID: PMC11975483 DOI: 10.1007/s00439-024-02724-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/27/2024] [Indexed: 03/05/2025]
Abstract
New thermodynamic and functional studies have been recently conducted to evaluate the impact of amino acid substitutions on the Mitogen Activated Protein Kinases 1 and 3 (MAPK1/3). The Critical Assessment of Genome Interpretation (CAGI) data provider, at Sapienza University of Rome, measured the unfolding free energy and the enzymatic activity of a set of variants (MAPK challenge dataset). Thermodynamic measurements for the denaturant-induced equilibrium unfolding of the phosphorylated and unphosphorylated forms of the MAPKs were obtained by monitoring the far-UV circular dichroism and intrinsic fluorescence changes as a function of denaturant concentration. These values have been used to calculate the change in unfolding free energy between the variant and wild-type proteins at zero concentration of denaturant ( Δ Δ G H 2 O ). The enzymatic activity of the phosphorylated MAPKs variants was also measured using Chelation-Enhanced Fluorescence to monitor the phosphorylation of a peptide substrate. The MAPK challenge dataset, composed of a total of 23 single amino acid substitutions (11 and 12 for MAPK1 and MAPK3, respectively), was used to assess the effectiveness of the computational methods in predicting the Δ Δ G H 2 O values, associated with the variants, and categorize them as destabilizing and not destabilizing. The data on the enzymatic activity of the MAPKs mutants were used to assess the performance of the methods for predicting the functional impact of the variants. For the sixth edition of CAGI, thirteen independent research groups from four continents (Asia, Australia, Europe and North America) submitted > 80 sets of predictions, obtained from different approaches. In this manuscript, we summarized the results of our assessment to highlight the possible limitations of the available algorithms.
Collapse
Affiliation(s)
- Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Maria Petrosino
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Roma, 00185, Rome, Italy
| | | | - Leonore Novak
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Roma, 00185, Rome, Italy
| | - Alessandra Pasquo
- Diagnostics and Metrology Laboratory FSN-TECFIS-DIM, ENEA CR Frascati, 00044, Frascati, Italy
| | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - Muttaqi Ahmad Alladin
- Department of Computational and Data Sciences, Indian Institute of Science, Bangaluru, 560012, India
| | - David B Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Constantina Bakolitsa
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, NextGen Precision Health Institute, University of Missouri, Columbia, MO, 65211, USA
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
| | - Lukas Folkman
- Institute for Integrated and Intelligent Systems, Griffith University, Southport, QLD, 4222, Australia
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Minghui Li
- School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Dong Li
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050, Brussels, Belgium
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sajid Mahmud
- Department of Electrical Engineering and Computer Science, NextGen Precision Health Institute, University of Missouri, Columbia, MO, 65211, USA
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bangaluru, 560012, India
| | | | - Douglas E V Pires
- School of Computing and Information Systems, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050, Brussels, Belgium
| | - Carlos H M Rodrigues
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050, Brussels, Belgium
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 1050, Brussels, Belgium
| | - Yang Shen
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, 77843, USA
| | - Alexey V Strokach
- Department of Computer Science, University of Toronto, Toronto, ON, M5S 2E4, Canada
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, 77843, USA
| | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Steven E Brenner
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Roberta Chiaraluce
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Roma, 00185, Rome, Italy.
| | - Valerio Consalvi
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Roma, 00185, Rome, Italy.
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
- Computational Genomics Platform, IRCCS University Hospital of Bologna, 40138, Bologna, Italy.
| |
Collapse
|
30
|
Xu W, Li A, Zhao Y, Peng Y. Decoding the effects of mutation on protein interactions using machine learning. BIOPHYSICS REVIEWS 2025; 6:011307. [PMID: 40013003 PMCID: PMC11857871 DOI: 10.1063/5.0249920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/28/2025]
Abstract
Accurately predicting mutation-caused binding free energy changes (ΔΔGs) on protein interactions is crucial for understanding how genetic variations affect interactions between proteins and other biomolecules, such as proteins, DNA/RNA, and ligands, which are vital for regulating numerous biological processes. Developing computational approaches with high accuracy and efficiency is critical for elucidating the mechanisms underlying various diseases, identifying potential biomarkers for early diagnosis, and developing targeted therapies. This review provides a comprehensive overview of recent advancements in predicting the impact of mutations on protein interactions across different interaction types, which are central to understanding biological processes and disease mechanisms, including cancer. We summarize recent progress in predictive approaches, including physicochemical-based, machine learning, and deep learning methods, evaluating the strengths and limitations of each. Additionally, we discuss the challenges related to the limitations of mutational data, including biases, data quality, and dataset size, and explore the difficulties in developing accurate prediction tools for mutation-induced effects on protein interactions. Finally, we discuss future directions for advancing these computational tools, highlighting the capabilities of advancing technologies, such as artificial intelligence to drive significant improvements in mutational effects prediction.
Collapse
Affiliation(s)
- Wang Xu
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Anbang Li
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Yunjie Zhao
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| | - Yunhui Peng
- Institute of Biophysics and Department of Physics, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
31
|
Turina P, Dal Cortivo G, Enriquez Sandoval CA, Alexov E, Ascher DB, Babbi G, Bakolitsa C, Casadio R, Fariselli P, Folkman L, Kamandula A, Katsonis P, Li D, Lichtarge O, Martelli PL, Panday SK, Pires DEV, Portelli S, Pucci F, Rodrigues CHM, Rooman M, Savojardo C, Schwersensky M, Shen Y, Strokach AV, Sun Y, Woo J, Radivojac P, Brenner SE, Dell'Orco D, Capriotti E. Assessing the predicted impact of single amino acid substitutions in calmodulin for CAGI6 challenges. Hum Genet 2025; 144:113-125. [PMID: 39714488 PMCID: PMC11975486 DOI: 10.1007/s00439-024-02720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Recent thermodynamic and functional studies have been conducted to evaluate the impact of amino acid substitutions on Calmodulin (CaM). The Critical Assessment of Genome Interpretation (CAGI) data provider at University of Verona (Italy) measured the melting temperature (Tm) and the percentage of unfolding (%unfold) of a set of CaM variants (CaM challenge dataset). Thermodynamic measurements for the equilibrium unfolding of CaM were obtained by monitoring far-UV Circular Dichroism as a function of temperature. These measurements were used to determine the Tm and the percentage of protein remaining unfolded at the highest temperature. The CaM challenge dataset, comprising a total of 15 single amino acid substitutions, was used to evaluate the effectiveness of computational methods in predicting the Tm and unfolding percentages associated with the variants, and categorizing them as destabilizing or not. For the sixth edition of CAGI, nine independent research groups from four continents (Asia, Australia, Europe, and North America) submitted over 52 sets of predictions, derived from various approaches. In this manuscript, we summarize the results of our assessment to highlight the potential limitations of current algorithms and provide insights into the future development of more accurate prediction tools. By evaluating the thermodynamic stability of CaM variants, this study aims to enhance our understanding of the relationship between amino acid substitutions and protein stability, ultimately contributing to more accurate predictions of the effects of genetic variants.
Collapse
Affiliation(s)
- Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | | | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - David B Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Constantina Bakolitsa
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Lukas Folkman
- Institute for Integrated and Intelligent Systems, Griffith University, Southport, QLD, Australia
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Dong Li
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | | | - Douglas E V Pires
- School of Computing and Information Systems, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Carlos H M Rodrigues
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Yang Shen
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | - Alexey V Strokach
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Steven E Brenner
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
- Biophysics Graduate Group, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy.
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
- Computational Genomics Platform, IRCCS University Hospital of Bologna, 40138, Bologna, Italy.
| |
Collapse
|
32
|
Smith CJ, Eavis H, Briggs C, Henrici R, Karpiyevich M, Ansbro MR, Hoshizaki J, van der Heden van Noort GJ, Ascher DB, Sutherland CJ, Lee MCS, Artavanis-Tsakonas K. Drug resistance-associated mutations in Plasmodium UBP-1 disrupt its essential deubiquitinating activity. J Biol Chem 2025; 301:108266. [PMID: 39909372 PMCID: PMC11927682 DOI: 10.1016/j.jbc.2025.108266] [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: 09/15/2022] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Deubiquitinating enzymes function to cleave ubiquitin (Ub) moieties from modified proteins, serving to maintain the pool of free Ub in the cell while simultaneously impacting the fate and function of a target protein. Like all eukaryotes, Plasmodium parasites rely on the dynamic addition and removal of Ub for their own growth and survival. While humans possess around 100 deubiquitinases, Plasmodium contains ∼20 putative Ub hydrolases, many of which bear little to no resemblance to those of other organisms. In this study, we characterize Plasmodium falciparum UBP-1, a large Ub hydrolase unique to Plasmodium spp., which has been linked to endocytosis and drug resistance. We demonstrate its Ub activity, linkage specificity, and assess the repercussions of point mutations associated with drug resistance on catalytic activity and parasite fitness. We confirm that the deubiquitinating activity of UBP-1 is essential for parasite survival, implicating an important role for Ub signaling in endocytosis.
Collapse
Affiliation(s)
- Cameron J Smith
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Heledd Eavis
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Carla Briggs
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ryan Henrici
- Department of Immunology, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Megan R Ansbro
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | - David B Ascher
- University of Queensland, School of Chemistry and Molecular Biosciences, Queensland, Australia
| | - Colin J Sutherland
- Department of Immunology, London School of Hygiene & Tropical Medicine, London, UK
| | - Marcus C S Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK; Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, United Kingdom
| | | |
Collapse
|
33
|
Hosseini Faradonbeh SM, Seyedalipour B, Keivan Behjou N, Rezaei K, Baziyar P, Hosseinkhani S. Structural insights into SOD1: from in silico and molecular dynamics to experimental analyses of ALS-associated E49K and R115G mutants. Front Mol Biosci 2025; 12:1532375. [PMID: 40070688 PMCID: PMC11893412 DOI: 10.3389/fmolb.2025.1532375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025] Open
Abstract
Protein stability is a crucial characteristic that influences both protein activity and structure and plays a significant role in several diseases. Cu/Zn superoxide dismutase 1 (SOD1) mutations serve as a model for elucidating the destabilizing effects on protein folding and misfolding linked to the lethal neurological disease, amyotrophic lateral sclerosis (ALS). In the present study, we have examined the structure and dynamics of the SOD1 protein upon two ALS-associated point mutations at the surface (namely, E49K and R115G), which are located in metal-binding loop IV and Greek key loop VI, respectively. Our analysis was performed through multiple algorithms on the structural characterization of the hSOD1 protein using computational predictions, molecular dynamics (MD) simulations, and experimental studies to understand the effects of amino acid substitutions. Predictive results of computational analysis predicted the deleterious and destabilizing effect of mutants on hSOD1 function and stability. MD outcomes also indicate that the mutations result in structural destabilization by affecting the increased content of β-sheet structures and loss of hydrogen bonds. Moreover, comparative intrinsic and extrinsic fluorescence results of WT-hSOD1 and mutants indicated structural alterations and increased hydrophobic surface pockets, respectively. As well, the existence of β-sheet-dominated structures was observed under amyloidogenic conditions using FTIR spectroscopy. Overall, our findings suggest that mutations in the metal-binding loop IV and Greek key loop VI lead to significant structural and conformational changes that could affect the structure and stability of the hSOD1 molecule, resulting in the formation of toxic intermediate species that cause ALS.
Collapse
Affiliation(s)
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Nasrin Keivan Behjou
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Kimiya Rezaei
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Payam Baziyar
- Department of Molecular and Cell Biology, Faculty of Basic Science, University of Mazandaran, Babolsar, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
34
|
Moltrasio C, Moura RR, Brandão L, Tricarico PM, Suleman M, Maronese CA, Crovella S, Marzano AV. Keratin Variants in Pyoderma Gangrenosum: Pathogenetic Insights from a Whole-Exome Sequencing-Based Bioinformatic Analysis. J Invest Dermatol 2025:S0022-202X(25)00115-0. [PMID: 39983981 DOI: 10.1016/j.jid.2025.01.029] [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: 09/10/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 02/23/2025]
Abstract
Pyoderma gangrenosum (PG) is an inflammatory skin disorder that belongs to the group of neutrophilic dermatoses. Clinically, it is typified by cutaneous ulcers with distinctive erythematoviolaceous borders and may occur alone or in association with other inflammatory, autoinflammatory, or neoplastic conditions. Although its pathophysiology remains incompletely understood, mounting evidence points toward a predisposing genetic background and dysregulation of both the innate and adaptive immune responses, with follicular or epidermal structures as putative initial targets. To investigate the genetic factors associated with PG susceptibility and severity (arbitrarily defined as unilesional or multilesional), whole-exome sequencing was performed on 11 unrelated patients with PG. Eight carried at least 1 variant of the keratin-encoding genes, including keratin (K)18 gene K18, K20, K23, K32, and K33B. Strikingly, a recurrent variant (rs77999286) of the K18 gene was identified in 5 of 6 patients with multilesional PG and 1 of 5 of those with unilesional PG. AlphaFold modeling and mutation analysis revealed the destabilizing effect of the K18 rs77999286 variant on protein structure. Furthermore, immunohistochemistry revealed undetectable K18 staining in lesional skin compared with that in healthy control skin. Overall, these findings suggest that keratin variants may play a role in PG pathogenesis and indicate that the K18 rs77999286 variant is a potential genetic factor linked to multilesional disease.
Collapse
Affiliation(s)
- Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ronald Rodrigues Moura
- Department of Advanced Diagnostics, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo," Trieste, Italy
| | - Lucas Brandão
- Department of Pathology, Federal University of Pernambuco, Recife, Brazil
| | - Paola Maura Tricarico
- Department of Pediatrics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo," Trieste, Italy
| | - Muhammad Suleman
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Carlo Alberto Maronese
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar.
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
35
|
Nazir A, Sajjad M. Recent trends in biotechnological production, engineering, and applications of lysophospholipases. Biotechnol Prog 2025:e70014. [PMID: 39968651 DOI: 10.1002/btpr.70014] [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: 09/07/2024] [Revised: 10/11/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
Oil degumming process involves the removal of gums, which is required to improve the physicochemical and storage properties of the vegetable oils. Degumming of oils can be carried out by using chemicals, membranes (polymeric, inorganic, and ceramic), or enzymes, for example, phospholipases. Phospholipases are enzymes of tremendous significance in the degumming process as they convert gums to fatty acids and lipophilic substances. They provide a cost-effective and safe alternative to other degumming processes without affecting the oil yield. Lysophospholipases (LPLs) are highly valuable tools for degumming vegetable oils. LPLs can hydrolyze fatty acyl ester bonds of phosphatidylcholine at the sn-1 and sn-2 positions of glycerol moiety. In addition, they have the ability to catalyze hydrolysis lysophospholipids' ester bond either at sn-1 or sn-2 position. In this review, biotechnological production and biochemical characteristics of LPLs from three domains of life are highlighted. In comparison to bacterial and eukaryotic LPLs, archaeal LPLs were found to be active at high temperatures. Broad substrate specificity and thermostability of archaeal LPLs make them ideal candidates for the industrial degumming of oils. However, improvement of activity and substrate specificity of archaeal LPLs is required for enhancing their industrial utility. In the current review, various protein-engineering approaches (directed evolution, rational design, site-saturation mutagenesis, and fusion technology) as well as in silico tools have been discussed to increase the commercial significance of LPLs.
Collapse
Affiliation(s)
- Arshia Nazir
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Muhammad Sajjad
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
36
|
Yang M, Tong Z, Yuan Z, Jiang B, Zhao Y, Xu D, Yuan Y. A Novel Missense Variant of BMPR1A in Juvenile Polyposis Syndrome: Assessment of Structural and Functional Alternations. Hum Mutat 2025; 2025:7317429. [PMID: 40226309 PMCID: PMC11919155 DOI: 10.1155/humu/7317429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/27/2025] [Indexed: 04/15/2025]
Abstract
Juvenile polyposis syndrome (JPS) is a rare precancerous condition associated with a high susceptibility to colorectal cancer. The genetic basis of JPS has been reported to lie in germline mutations in BMPR1A or SMAD4, resulting in diverse clinical manifestations and an elusive underlying mechanism. We firstly utilized a 139-gene next-generation sequencing (NGS) panel to detect the germline variants and further employed various prediction tools to assess the pathogenicity and functional alternations. Consequently, we identified a novel pathogenic BMPR1A missense variant (c.355C>T; p.R119C). More importantly, we proposed for the first time that the missense variant would lead to a decrease in molecular weight, potentially associated with reduced protein stability, diminished posttranslational modifications, and aberrant alternative splicing. These findings may provide novel perspectives for further exploration into the role of BMPR1A in JPS development. Also, we hope to encourage clinicians to underscore the importance of genetic testing and analysis in facilitating the diagnosis and treatment of diseases.
Collapse
Affiliation(s)
- Mengyuan Yang
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ziyan Tong
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhijun Yuan
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bingjing Jiang
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yingxin Zhao
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dong Xu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Kurt-Şükür ED, Timucin E, Baştuğ T, Ozaltin F. A Novel NUP85 Variant Expanding the Phenotypic Spectrum of NUP85-Associated Steroid-Resistant Nephrotic Syndrome. Clin Genet 2025. [PMID: 39949197 DOI: 10.1111/cge.14703] [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: 10/14/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 03/21/2025]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a severe kidney disorder linked to over 60 genes, including NUP85, which plays a key role in nuclear pore function and glomerulogenesis. We identified a novel homozygous NUP85 variant (NM_024844.5: c.1379G > A, p.Arg460Gln) in a pediatric SRNS patient who also presented with cleft lip-palate and mild intellectual disability, marking the first reported association of these phenotypes with a NUP85 variant. Molecular dynamics simulations revealed that the variant destabilizes the protein's helix bundle, providing mechanistic insights into its potential pathogenic effects. This study broadens the known phenotypic spectrum of NUP85-related conditions and highlights the value of computational tools, such as molecular dynamics, in unraveling the impact of novel variants.
Collapse
Affiliation(s)
- Eda Didem Kurt-Şükür
- Department of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
- Center for Genomics and Rare Diseases, Hacettepe University, Ankara, Türkiye
| | - Emel Timucin
- Department of Biostatistics and Medical Informatics, School of Medicine, Acibadem University, Istanbul, Türkiye
| | - Turgut Baştuğ
- Department of Biophysics, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Fatih Ozaltin
- Department of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
- Center for Genomics and Rare Diseases, Hacettepe University, Ankara, Türkiye
- Department of Bioinformatics, Hacettepe University Institute of Health Sciences, Ankara, Türkiye
| |
Collapse
|
38
|
Cif L, Demailly D, Lin JP, Barwick KE, Sa M, Abela L, Malhotra S, Chong WK, Steel D, Sanchis-Juan A, Ngoh A, Trump N, Meyer E, Vasques X, Rankin J, Allain MW, Applegate CD, Isfahani SA, Baleine J, Balint B, Bassetti JA, Baple EL, Bhatia KP, Blanchet C, Burglen L, Cambonie G, Seng EC, Bastaraud SC, Cyprien F, Coubes C, d’Hardemare V, Deciphering Developmental Disorders Study, Doja A, Dorison N, Doummar D, Dy-Hollins ME, Farrelly E, Fitzpatrick DR, Fearon C, Fieg EL, Fogel BL, Forman EB, Fox RG, Genomics England Research Consortium, Gahl WA, Galosi S, Gonzalez V, Graves TD, Gregory A, Hallett M, Hasegawa H, Hayflick SJ, Hamosh A, Hully M, Jansen S, Jeong SY, Krier JB, Krystal S, Kumar KR, Laurencin C, Lee H, Lesca G, François LL, Lynch T, Mahant N, Martinez-Agosto JA, Milesi C, Mills KA, Mondain M, Morales-Briceno H, NIHR BioResource, Ostergaard JR, Pal S, Pallais JC, Pavillard F, Perrigault PF, Petersen AK, Polo G, Poulen G, Rinne T, Roujeau T, Rogers C, Roubertie A, Sahagian M, Schaefer E, Selim L, Selway R, Sharma N, Signer R, Soldatos AG, Stevenson DA, Stewart F, Tchan M, Undiagnosed Diseases Network, Verma IC, de Vries BBA, Wilson JL, Wong DA, Zaitoun R, Zhen D, et alCif L, Demailly D, Lin JP, Barwick KE, Sa M, Abela L, Malhotra S, Chong WK, Steel D, Sanchis-Juan A, Ngoh A, Trump N, Meyer E, Vasques X, Rankin J, Allain MW, Applegate CD, Isfahani SA, Baleine J, Balint B, Bassetti JA, Baple EL, Bhatia KP, Blanchet C, Burglen L, Cambonie G, Seng EC, Bastaraud SC, Cyprien F, Coubes C, d’Hardemare V, Deciphering Developmental Disorders Study, Doja A, Dorison N, Doummar D, Dy-Hollins ME, Farrelly E, Fitzpatrick DR, Fearon C, Fieg EL, Fogel BL, Forman EB, Fox RG, Genomics England Research Consortium, Gahl WA, Galosi S, Gonzalez V, Graves TD, Gregory A, Hallett M, Hasegawa H, Hayflick SJ, Hamosh A, Hully M, Jansen S, Jeong SY, Krier JB, Krystal S, Kumar KR, Laurencin C, Lee H, Lesca G, François LL, Lynch T, Mahant N, Martinez-Agosto JA, Milesi C, Mills KA, Mondain M, Morales-Briceno H, NIHR BioResource, Ostergaard JR, Pal S, Pallais JC, Pavillard F, Perrigault PF, Petersen AK, Polo G, Poulen G, Rinne T, Roujeau T, Rogers C, Roubertie A, Sahagian M, Schaefer E, Selim L, Selway R, Sharma N, Signer R, Soldatos AG, Stevenson DA, Stewart F, Tchan M, Undiagnosed Diseases Network, Verma IC, de Vries BBA, Wilson JL, Wong DA, Zaitoun R, Zhen D, Znaczko A, Dale RC, de Gusmão CM, Friedman J, Fung VSC, King MD, Mohammad SS, Rohena L, Waugh JL, Toro C, Raymond FL, Topf M, Coubes P, Gorman KM, Kurian MA. KMT2B-related disorders: expansion of the phenotypic spectrum and long-term efficacy of deep brain stimulation. ARXIV 2025:arXiv:2502.06320v1. [PMID: 39990802 PMCID: PMC11844621] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Heterozygous mutations in KMT2B are associated with an early-onset, progressive and often complex dystonia (DYT28). Key characteristics of typical disease include focal motor features at disease presentation, evolving through a caudocranial pattern into generalized dystonia, with prominent oromandibular, laryngeal and cervical involvement. Although KMT2B-related disease is emerging as one of the most common causes of early-onset genetic dystonia, much remains to be understood about the full spectrum of the disease. We describe a cohort of 53 patients with KMT2B mutations, with detailed delineation of their clinical phenotype and molecular genetic features. We report new disease presentations, including atypical patterns of dystonia evolution and a subgroup of patients with a non-dystonic neurodevelopmental phenotype. In addition to the previously reported systemic features, our study has identified co-morbidities, including the risk of status dystonicus, intrauterine growth retardation, and endocrinopathies. Analysis of this study cohort (n = 53) in tandem with published cases (n = 80) revealed that patients with chromosomal deletions and protein truncating variants had a significantly higher burden of systemic disease (with earlier onset of dystonia) than those with missense variants. Eighteen individuals had detailed longitudinal data available after insertion of deep brain stimulation for medically refractory dystonia. Median age at deep brain stimulation was 11.5 years (range: 4.5-37.0 years). Follow-up after deep brain stimulation ranged from 0.25 to 22 years. Significant improvement of motor function and disability (as assessed by the Burke Fahn Marsden's Dystonia Rating Scales, BFMDRS-M and BFMDRS-D) was evident at ł months, 1 year and last follow-up (motor, P = 0.001, P = 0.004, and P = 0.012; disability, P = 0.009, P = 0.002 and P = 0.012). At 1 year post-deep brain stimulation, >50% of subjects showed BFMDRS-M and BFMDRS-D improvements of >30%. In the long-term deep brain stimulation cohort (deep brain stimulation inserted for >5 years, n = 8), improvement of >30% was maintained in 5/8 and 3/8 subjects for the BFMDRS-M and BFMDRS-D, respectively. The greatest BFMDRS-M improvements were observed for trunk (53.2%) and cervical (50.5%) dystonia, with less clinical impact on laryngeal dystonia. Improvements in gait dystonia decreased from 20.9% at 1 year to 1ł.2% at last assessment; no patient maintained a fully independent gait. Reduction of BFMDRS-D was maintained for swallowing (52.9%). Five patients developed mild parkinsonism following deep brain stimulation. KMT2B-related disease comprises an expanding continuum from infancy to adulthood, with early evidence of genotype-phenotype correlations. Except for laryngeal dysphonia, deep brain stimulation provides a significant improvement in quality of life and function with sustained clinical benefit depending on symptoms distribution.
Collapse
Affiliation(s)
- Laura Cif
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Diane Demailly
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Jean-Pierre Lin
- Complex Motor Disorder Service, Children’s Neurosciences Department, Evelina London Children’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Children’s Neuromodulation Group, Women and Children’s Health Institute, Faculty of life Sciences and Medicine (FOLSM), King’s Health Partners, London, UK
| | - Katy E. Barwick
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Mario Sa
- Complex Motor Disorder Service, Children’s Neurosciences Department, Evelina London Children’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Lucia Abela
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sony Malhotra
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, UK
| | - Wui K. Chong
- Developmental Imaging and Biophysics, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dora Steel
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Alba Sanchis-Juan
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Haematology, NHS Blood and Transplant Centre, University of Cambridge, Cambridge, UK
| | - Adeline Ngoh
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Natalie Trump
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Esther Meyer
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Julia Rankin
- Clinical Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Meredith W. Allain
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | - Carolyn D. Applegate
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanaz Attaripour Isfahani
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Julien Baleine
- Unité de Soins Intensifs et Réanimation Pédiatrique et Néonatale, Hôpital Universitaire de Montpellier, Montpellier, France
| | - Bettina Balint
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jennifer A. Bassetti
- Division of Medical Genetics, Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Emma L. Baple
- Clinical Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
- Institute of Biomedical and Clinical Science RILD Wellcome Wolfson Centre, University of Exeter Medical School, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Kailash P. Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine Blanchet
- Département d’Oto-Rhino-Laryngologie et Chirurgie Cervico-Faciale, Hôpital Universitaire de Montpellier, Montpellier, France
| | - Lydie Burglen
- Département de génétique médicale, APHP Hôpital Armand Trousseau, Paris, France
| | - Gilles Cambonie
- Unité de Soins Intensifs et Réanimation Pédiatrique et Néonatale, Hôpital Universitaire de Montpellier, Montpellier, France
| | - Emilie Chan Seng
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | | | - Fabienne Cyprien
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Christine Coubes
- Département de Génétique médicale, Maladies rares et médecine personnalisée, CHU Montpellier, Montpellier, France
| | - Vincent d’Hardemare
- Unité Dyspa, Neurochirurgie Pédiatrique, Hôpital Fondation Rothschild, Paris, France
| | | | - Asif Doja
- Division of Neurology, Children’s Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Nathalie Dorison
- Unité Dyspa, Neurochirurgie Pédiatrique, Hôpital Fondation Rothschild, Paris, France
| | - Diane Doummar
- Neuropédiatrie, Centre de référence neurogénétique mouvement anormaux de l’enfant, Hôpital Armand Trousseau, AP-HP, Sorbonne Université, France
| | - Marisela E. Dy-Hollins
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Ellyn Farrelly
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Palo Alto, CA, USA
- Department of Pediatrics, Lucile Packard Children’s Hospital at Stanford, CA, USA
| | - David R. Fitzpatrick
- Human Genetics Unit, Medical and Developmental Genetics, University of Edinburgh Western General Hospital, Edinburgh, Scotland, UK
| | - Conor Fearon
- Department of Neurology, The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland
| | - Elizabeth L. Fieg
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Brent L. Fogel
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Eva B. Forman
- Department of Paediatric Neurology and Clinical Neurophysiology, Children’s Health Ireland at Temple Street, Dublin, Ireland
| | - Rachel G. Fox
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | | | - William A. Gahl
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Serena Galosi
- Department of Human Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Victoria Gonzalez
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Tracey D. Graves
- Department of Neurology, Hinchingbrooke Hospital, North West Anglia NHS Foundation Trust, Huntingdon, UK
| | - Allison Gregory
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Mark Hallett
- Human Motor Control Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Harutomo Hasegawa
- Complex Motor Disorder Service, Children’s Neurosciences Department, Evelina London Children’s Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
- Children’s Neuromodulation Group, Women and Children’s Health Institute, Faculty of life Sciences and Medicine (FOLSM), King’s Health Partners, London, UK
| | - Susan J. Hayflick
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
- Department of Paediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Ada Hamosh
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Marie Hully
- Département de Neurologie, APHP-Necker-Enfants Malades, Paris, France
| | - Sandra Jansen
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Suh Young Jeong
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Joel B. Krier
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sidney Krystal
- Département de Neuroradiologie, Hôpital Fondation Rothschild, Paris
| | - Kishore R. Kumar
- Translational Genomics Group, Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- Department of Neurogenetics, Kolling Institute, University of Sydney and Royal North Shore Hospital, St Leonards, NSW, Australia
- Molecular Medicine Laboratory, Concord Hospital, Sydney, NSW, Australia
| | - Chloé Laurencin
- Département de Neurologie, Hôpital Neurologique Pierre Wertheimer, Lyon, France
| | - Hane Lee
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Gaetan Lesca
- Département de Génétique, Hôpital Universitaire de Lyon, Lyon, France
| | | | - Timothy Lynch
- Department of Neurology, The Dublin Neurological Institute at the Mater Misericordiae University Hospital, Dublin, Ireland
- UCD School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Neil Mahant
- Movement Disorders Unit, Department of Neurology, Westmead Hospital, Westmead, NSW, Australia
| | - Julian A. Martinez-Agosto
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Christophe Milesi
- Unité de Soins Intensifs et Réanimation Pédiatrique et Néonatale, Hôpital Universitaire de Montpellier, Montpellier, France
| | - Kelly A. Mills
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michel Mondain
- Département d’Oto-Rhino-Laryngologie et Chirurgie Cervico-Faciale, Hôpital Universitaire de Montpellier, Montpellier, France
| | - Hugo Morales-Briceno
- Movement Disorders Unit, Department of Neurology, Westmead Hospital, Westmead, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - NIHR BioResource
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Swasti Pal
- Institute of Genetics and Genomics, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, India
| | - Juan C. Pallais
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Frédérique Pavillard
- Département d’Anesthésie-Réanimation Gui de Chauliac, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Pierre-Francois Perrigault
- Département d’Anesthésie-Réanimation Gui de Chauliac, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | | | - Gustavo Polo
- Département de Neurochirurgie Fonctionnelle, Hôpital Neurologique et Neurochirurgical, Pierre Wertheimer, Lyon, France
| | - Gaetan Poulen
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Tuula Rinne
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Roujeau
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
| | - Caleb Rogers
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Agathe Roubertie
- Département de Neuropédiatrie, Hôpital Universitaire de Montpellier, Montpellier, France
- INSERM U1051, Institut des Neurosciences de Montpellier, Montpellier, France
| | - Michelle Sahagian
- Division of Neurology, Rady Children’s Hospital San Diego, CA, USA
- Department of Neuroscience, University of California San Diego, CA, USA
| | - Elise Schaefer
- Medical Genetics, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laila Selim
- Cairo University Children Hospital, Pediatric Neurology and Metabolic division, Cairo, Egypt
| | - Richard Selway
- Department of Neurosurgery, King’s College Hospital, London, UK
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Rebecca Signer
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ariane G. Soldatos
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A. Stevenson
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | - Fiona Stewart
- Department of Genetic Medicine, Belfast Health and Social Care Trust, Belfast, UK
| | - Michel Tchan
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Department of Genetics, Westmead Hospital, Westmead, NSW, Australia
| | - Undiagnosed Diseases Network
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ishwar C. Verma
- Institute of Genetics and Genomics, Sir Ganga Ram Hospital, Rajender Nagar, New Delhi, India
| | - Bert B. A. de Vries
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jenny L. Wilson
- Division of Pediatric Neurology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | - Derek A. Wong
- Division of Medical Genetics, Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Raghda Zaitoun
- Department of Paediatrics, Neurology Division, Ain Shams University Hospital, Cairo, Egypt
| | - Dolly Zhen
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Anna Znaczko
- Department of Genetic Medicine, Belfast Health and Social Care Trust, Belfast, UK
| | - Russell C. Dale
- Department of Paediatric Neurology, The Children’s Hospital at Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney NSW, Australia
| | - Claudio M. de Gusmão
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
| | - Jennifer Friedman
- Division of Neurology, Rady Children’s Hospital San Diego, CA, USA
- Department of Neuroscience, University of California San Diego, CA, USA
- Departments of Paediatrics, University of California, San Diego, CA, USA
- Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Victor S. C. Fung
- Movement Disorders Unit, Department of Neurology, Westmead Hospital, Westmead, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mary D. King
- Department of Paediatric Neurology and Clinical Neurophysiology, Children’s Health Ireland at Temple Street, Dublin, Ireland
- UCD School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Shekeeb S. Mohammad
- Department of Paediatric Neurology, The Children’s Hospital at Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney NSW, Australia
| | - Luis Rohena
- Division of Medical Genetics, Department of Pediatrics, San Antonio Military Medical Center, San Antonio, TX, USA
- Department of Pediatrics, Long School of Medicine, UT Health, San Antonio, TX, USA
| | - Jeff L. Waugh
- Division of Pediatric Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Camilo Toro
- Undiagnosed Diseases Program, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - F. Lucy Raymond
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Maya Topf
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, University of London, London, UK
| | - Philippe Coubes
- Département de Neurochirurgie, Unité des Pathologies Cérébrales Résistantes, Unité de Recherche sur les Comportements et Mouvements Anormaux, Hôpital Gui de Chauliac, Centre Hospitalier Régional Montpellier, Montpellier, France
- Faculté demédecine, Université de Montpellier, France
| | - Kathleen M. Gorman
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Manju A. Kurian
- Molecular Neurosciences, Developmental Neurosciences, UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
39
|
Chen Z, He S, Chi X, Bo X. Predicting Antibody Affinity Changes upon Mutation Based on Unbound Protein Structures. Int J Mol Sci 2025; 26:1343. [PMID: 39941111 PMCID: PMC11818220 DOI: 10.3390/ijms26031343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Antibodies are key proteins in the immune system that can reversibly and non-covalently bind specifically to their corresponding antigens, forming antigen-antibody complexes. They play a crucial role in recognizing foreign or self-antigens during the adaptive immune response. Monoclonal antibodies have emerged as a promising class of biological macromolecule therapeutics with broad market prospects. In the process of antibody drug development, a key engineering challenge is to improve the affinity of candidate antibodies, without experimentally resolved structures of the antigen-antibody complexes as input for computer-aided predictive methods. In this work, we present an approach for predicting the effect of residue mutations on antibody affinity without the structures of the antigen-antibody complexes. The method involves the graph representation of proteins and utilizes a pre-trained encoder. The encoder captures the residue-level microenvironment of the target residue on the antibody along with the antigen context pre- and post-mutation. The encoder inherently possesses the potential to identify paratope residues. In addition, we curated a benchmark dataset specifically for mutations of the antibody. Compared to baseline methods based on complex structures and sequences, our approach achieves superior or comparable average accuracy on benchmark datasets. Additionally, we validate its advantage of not requiring antigen-antibody complex structures as input for predicting the effects of mutations in antibodies against SARS-CoV-2, influenza, and human cytomegalovirus. Our method shows its potential for identifying mutations that improve antibody affinity in practical antibody engineering applications.
Collapse
Affiliation(s)
| | | | - Xiangyang Chi
- Academy of Military Medical Sciences, Beijing 100850, China; (Z.C.); or (S.H.)
| | - Xiaochen Bo
- Academy of Military Medical Sciences, Beijing 100850, China; (Z.C.); or (S.H.)
| |
Collapse
|
40
|
Elasbali AM, Anjum F, AlKhamees OA, Abu Al-Soud W, Adnan M, Shamsi A, Hassan MI. A structural genomics approach to investigate Dystrophin mutations and their impact on the molecular pathways of Duchenne muscular dystrophy. Front Genet 2025; 16:1517707. [PMID: 39981262 PMCID: PMC11841421 DOI: 10.3389/fgene.2025.1517707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/06/2025] [Indexed: 02/22/2025] Open
Abstract
Background Dystrophin is a key protein encoded by the DMD gene, serves as a scaffold linking the cytoskeleton to the extracellular matrix that plays a critical role in muscle contraction, relaxation, and structural integrity. Mutations, particularly single-point amino acid substitutions, can lead to dysfunctional Dystrophin, causing muscular dystrophies, with Duchenne muscular dystrophy (DMD) being the most severe form. Objective This study aimed to evaluate the effects of 184 single-point amino acid substitutions on the structure and function of Dystrophin using computational approaches. Methods Many computational tools were used to predict the impact of amino acid substitutions on protein stability, solubility, and function. Pathogenic potential was assessed using disease phenotype predictors and CADD scores, while allele frequency data from gnomAD contextualized mutation prevalence. Additionally, aggregation propensity, frustration analysis, and post-translational modification sites were analyzed for functional disruptions. Results Of the 184 substitutions analyzed, 50 were identified as deleterious, with 41 predicted to be pathogenic. Seventeen mutations were localized in the Calponin-homology (CH) 1 domain, a critical functional region of Dystrophin. Six substitutions (N26H, N26K, G47W, D98G, G109A, and G109R) were predicted to decrease protein solubility and were located in minimally frustrated regions, potentially compromising Dystrophin functionality and contributing to DMD pathogenesis. Conclusion This study provides novel insights into the molecular mechanisms of DMD, highlighting specific mutations that disrupt Dystrophin's solubility and function. These findings could inform future therapeutic strategies targeting Dystrophin mutations to address DMD pathogenesis.
Collapse
Affiliation(s)
- Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Sakakah, Saudi Arabia
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Osama A. AlKhamees
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | | | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
41
|
Gallo E. Revolutionizing Synthetic Antibody Design: Harnessing Artificial Intelligence and Deep Sequencing Big Data for Unprecedented Advances. Mol Biotechnol 2025; 67:410-424. [PMID: 38308755 DOI: 10.1007/s12033-024-01064-2] [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: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/05/2024]
Abstract
Synthetic antibodies (Abs) represent a category of engineered proteins meticulously crafted to replicate the functions of their natural counterparts. Such Abs are generated in vitro, enabling advanced molecular alterations associated with antigen recognition, paratope site engineering, and biochemical refinements. In a parallel realm, deep sequencing has brought about a paradigm shift in molecular biology. It facilitates the prompt and cost-effective high-throughput sequencing of DNA and RNA molecules, enabling the comprehensive big data analysis of Ab transcriptomes, including specific regions of interest. Significantly, the integration of artificial intelligence (AI), based on machine- and deep- learning approaches, has fundamentally transformed our capacity to discern patterns hidden within deep sequencing big data, including distinctive Ab features and protein folding free energy landscapes. Ultimately, current AI advances can generate approximations of the most stable Ab structural configurations, enabling the prediction of de novo synthetic Abs. As a result, this manuscript comprehensively examines the latest and relevant literature concerning the intersection of deep sequencing big data and AI methodologies for the design and development of synthetic Abs. Together, these advancements have accelerated the exploration of antibody repertoires, contributing to the refinement of synthetic Ab engineering and optimizations, and facilitating advancements in the lead identification process.
Collapse
Affiliation(s)
- Eugenio Gallo
- Avance Biologicals, Department of Medicinal Chemistry, 950 Dupont Street, Toronto, ON, M6H 1Z2, Canada.
- RevivAb, Department of Protein Engineering, Av. Ipiranga, 6681, Partenon, Porto Alegre, RS, 90619-900, Brazil.
| |
Collapse
|
42
|
Abuzahra M, Al-Shuhaib MBS, Wijayanti D, Effendi MH, Mustofa I, Moses IB. A novel p.127Val>Ile single nucleotide polymorphism in the MTNR1A gene and its relation to litter size in Thin-tailed Indonesian ewes. Anim Biosci 2025; 38:209-222. [PMID: 38938032 PMCID: PMC11725752 DOI: 10.5713/ab.24.0187] [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: 03/29/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
OBJECTIVE The primary objective was to identify and characterize the single nucleotide polymorphisms (SNPs) within the MTNR1A gene sequence in Thin-tailed Indonesian ewes to assess the possible association of MTNR1A gene polymorphism with litter size trait. METHODS Forty-seven Thin-tailed Indonesian sheep were selected for the study. Genotyping involved collecting blood samples, and sequencing exon 2 of the MTNR1A gene. RESULTS The study identified 19 novel SNPs, with 10 being non-synonymous variations, in the MTNR1A gene of Thin-tailed Indonesian ewes. One non-synonymous SNP (rs1087815963) showed a significant association with litter size, with the GC genotype exhibiting a higher average litter size than the GG genotype. The deleterious impact of p.Val127Ile SNP was predicted by various in silico tools that predicted a highly damaging effect of p.Val127Ile SNP on the structure, function, and stability of MTNR1A. Docking reactions showed a critical involvement of this locus with the binding with melatonin. CONCLUSION In conclusion, the results of our study suggest that rs1087815963 has a remarkable negative impact on the MTNR1A with a putative alteration in the binding with melatonin. Therefore, the implementation of the novel p.Val127Ile could be a useful marker in marker-assisted selection.
Collapse
Affiliation(s)
- Mutasem Abuzahra
- Doctoral Program in Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115,
Indonesia
| | - Mohammed Baqur S. Al-Shuhaib
- Department of Animal Production, College of Agriculture, Al-Qasim Green University, Al-Qasim, 51013, Babil,
Iraq
| | - Dwi Wijayanti
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100,
China
| | - Mustofa Helmi Effendi
- Department of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya 60115,
Indonesia
| | - Imam Mustofa
- Department of Veterinary Reproduction, Faculty of Veterinary Medicine, Airlangga University, Surabaya 60115,
Indonesia
| | - Ikechukwu Benjamin Moses
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Abakaliki 481101,
Nigeria
| |
Collapse
|
43
|
Begum R, Das A, Alam MJ, Sultana GNN. Insights Into Genetic Variations of the OCT1 Gene in Metformin Poor Responders Among Bangladeshi Type 2 Diabetic Patients. Adv Pharmacol Pharm Sci 2025; 2025:8568658. [PMID: 39949862 PMCID: PMC11824854 DOI: 10.1155/adpp/8568658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 12/24/2024] [Indexed: 02/16/2025] Open
Abstract
Metformin is the most widely prescribed drug for the treatment of Type 2 diabetes mellitus (T2DM), but its response varies from person to person. This study aims to analyze the complete mutation spectrum of the OCT1 gene in metformin poor responders and to explore the potential pathogenic effects of the identified mutations. Clinical features of 56 Bangladeshi T2DM patients (who showed altered response to metformin) were analyzed, and genomic DNA was extracted from their blood samples. Subsequently, the entire exons (1-11), along with flanking introns of the OCT1 gene were amplified and sequenced. Molecular consequences of the identified mutations on OCT1 protein activity were determined through in silico analyses. In this study, 29 mutations of the OCT1 gene were identified; among which 5 mutations (c.412-86G>T, c.970G>C, c.1386-3088_1386-3083delGAATCA, c.1498+66G>T, and c.1653C>A) were novel. It was found that nsSNPs c.181C>T, c.1022C>T, c.493G>T, c.1207A>G, and c.970G>C (novel) as well as frameshift deletions have potential deleterious effects on OCT1 protein stability and function. Some of these mutations also cause alternative splicing, as per the HSF tool. In addition, alteration of interatomic bonding in the OCT1 protein due to two high-risk mutations (c.181C>T and c.1022C>T) was found from web-based analysis. The mutations, as mentioned earlier, are the most probable causative factor of decreased metformin effectiveness and adverse side effects in T2DM patients who are poor responders. Understanding the OCT1 gene variations of patients can help tailor treatment strategies for optimal metformin response or identify alternative medications.
Collapse
Affiliation(s)
- Rokeya Begum
- Genetic Engineering and Biotechnology Research Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka 1000, Bangladesh
| | - Arindita Das
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Md. Jahangir Alam
- Department of Biochemistry, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Gazi Nurun Nahar Sultana
- Genetic Engineering and Biotechnology Research Laboratory, Centre for Advanced Research in Sciences (CARS), University of Dhaka, Dhaka 1000, Bangladesh
| |
Collapse
|
44
|
Kamal MM, Shantanu KFH, Teeya ST, Rahman MM, Hasan AKMM, Chivers DP, Wani TA, Alshammari AH, Rachamalla M, da Silva Junior FC, Hossen MM. Investigating the functional and structural effect of non-synonymous single nucleotide polymorphisms in the cytotoxic T-lymphocyte antigen-4 gene: An in-silico study. PLoS One 2025; 20:e0316465. [PMID: 39854591 PMCID: PMC11759363 DOI: 10.1371/journal.pone.0316465] [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: 08/16/2024] [Accepted: 12/11/2024] [Indexed: 01/26/2025] Open
Abstract
The cytotoxic T-lymphocyte antigen-4 (CTLA4) is essential in controlling T cell activity within the immune system. Thus, uncovering the molecular dynamics of single nucleotide polymorphisms (SNPs) within the CTLA4 gene is critical. We identified the non-synonymous SNPs (nsSNPs), examined their impact on protein stability, and identified the protein sequences associated with them in the human CTLA4 gene. There were 3134 SNPs (rsIDs) in our study. Out of these, 186 missense variants (5.93%), 1491 intron variants (47.57%), and 91 synonymous variants (2.90%), while the remaining SNPs were unspecified. We utilized SIFT, PolyPhen-2, PROVEAN, and SNAP for identifying deleterious nsSNPs, and SNPs&GO, PhD SNP, and PANTHER for verifying risk nsSNPs in the CTLA4 gene. Following SIFT analysis, six nsSNPs were identified as deleterious and reporting second and third nsSNPs as probably damaging and one as benign, respectively. From upstream analysis, rs138279736, rs201778935, rs369567630, and rs376038796 were found to be deleterious, probably damaging, and disease associated. ConSurf predicted conservation scores for four nsSNPs, and Project Hope suggested that all mutations could disrupt protein interactions. Furthermore, mCSM and DynaMut2 analyses indicated a decrease in ΔΔG stability for the mutants. GeneMANIA and STRING networks highlighted correlations with CD86 and CD80 genes. Finally, MD simulation revealed consistent fluctuation in RMSD and RMSF, consequently Rg, hydrogen bonds, and PCA in the mutant proteins compared with wild-type, which might alter the functional and structural stability of CTLA4 protein. The current comprehensive study shows how various nsSNPs in the CTLA4 gene can modify the structural and functional characteristics of the protein, potentially influencing the pathogenesis of diseases in humans. Further, experimental studies are needed to analyze the effect of these nsSNPs on the susceptibility of pathological phenotype populations.
Collapse
Affiliation(s)
- Md. Mostafa Kamal
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | | | - Shamiha Tabassum Teeya
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Md. Motiar Rahman
- Department of Chemistry, The State University of New York, Binghamton, New York, United States of America
| | | | - Douglas P. Chivers
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Tanveer A. Wani
- Department of Pharmaceutical Chemistry, King Saud University, Riyadh, Saudi Arabia
| | | | - Mahesh Rachamalla
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | | | - Md. Munnaf Hossen
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| |
Collapse
|
45
|
Goutam RK, Huang G, Medina E, Ding F, Edenfield WJ, Sanabria H. Impact of frequent ARID1A mutations on protein stability provides insights into cancer pathogenesis. Sci Rep 2025; 15:3072. [PMID: 39856215 PMCID: PMC11760938 DOI: 10.1038/s41598-025-87103-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The ARID1A gene, frequently mutated in cancer, encodes the AT-rich interactive domain-containing protein 1 A, a key component of the chromatin remodeling SWI/SNF complex. The ARID1A protein features a conserved DNA-binding domain (ARID domain) of approximately 100 residues crucial for its function. Despite the frequency of mutations, the impact on ARID1A's stability and contribution to cancer progression remains unclear. We analyzed five frequent missense mutations R1020S, M1022K, K1047Q, G1063V, and A1089T identified in The Cancer Genome Atlas (TCGA) to assess their effects on the stability of the ARID domain using a hybrid experimental and computational approach. By combining computational stability from web server tools, the structural dynamics from replica exchange discrete molecular simulation (rexDMD), and thermal and chemical denaturation experiments, we found that the R1020S mutation severely decreases structural stability, making it the most impactful, while M1022K has minimal effect, and others lie in between. These findings enhance our understanding of the structural-functional relationship of ARID1A missense mutations at the molecular levels and their role in cancer pathogenesis. This research paves the way for identifying and categorizing which ARID1A mutations are most pathogenic, potentially guiding the development of targeted therapies tailored to specific mutation profiles in cancer treatment.
Collapse
Affiliation(s)
- Rajen K Goutam
- Department of Physics and Astronomy, Clemson University, Clemson, SC, USA
| | - Gangtong Huang
- Department of Physics and Astronomy, Clemson University, Clemson, SC, USA
| | - Exequiel Medina
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC, USA
| | - William J Edenfield
- Institute for Translational Oncology Research, Prisma Health, Greenville, SC, USA
| | - Hugo Sanabria
- Department of Physics and Astronomy, Clemson University, Clemson, SC, USA.
| |
Collapse
|
46
|
Escalera-Balsera A, Robles-Bolivar P, Parra-Perez AM, Murillo-Cuesta S, Chua HC, Rodríguez-de la Rosa L, Contreras J, Domarecka E, Amor-Dorado JC, Soto-Varela A, Varela-Nieto I, Szczepek AJ, Gallego-Martinez A, Lopez-Escamez JA. A rare haplotype of the GJD3 gene segregating in familial Meniere's disease interferes with connexin assembly. Genome Med 2025; 17:4. [PMID: 39815343 PMCID: PMC11737067 DOI: 10.1186/s13073-024-01425-1] [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: 01/09/2024] [Accepted: 12/11/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Familial Meniere's disease (FMD) is a rare polygenic disorder of the inner ear. Mutations in the connexin gene family, which encodes gap junction proteins, can also cause hearing loss, but their role in FMD is largely unknown. METHODS We retrieved exome sequencing data from 94 individuals in 70 Meniere's disease (MD) families. Through gene burden analysis, we calculated the enrichment of rare variants (allele frequency < 0.05) in connexins genes in FMD individuals compared with the reference population. The connexin monomer and the homomeric connexon structural models were predicted using AlphaFold2 and HDOCK. RT-qPCR and immunofluorescence were done in mice cochleae to identify expression of the mouse ortholog candidate gene Gjd3. RESULTS We found an enrichment of rare missense variants in the GJD3 gene when comparing allelic frequencies in FMD (N = 94) with the Spanish reference population (OR = 3.9[1.92-7.91], FDR = 2.36E-03). In the GJD3 sequence, we identified a rare haplotype (TGAGT) composed of two missense, two synonymous, and one downstream variant. This haplotype was found in five individuals with FMD, segregating in three unrelated families with a total of ten individuals; and in another eight MD individuals. GJD3 encodes the gap junction protein delta 3, also known as human connexin 31.9 (Cx31.9). The protein model predicted that the NP_689343.3:p.(His175Tyr) missense variant could modify the interaction between connexins and the connexon assembly, affecting the homotypic GJD3 gap junction between cells. Our studies in mice revealed that Gjd3-encoding Gjd3 or mouse connexin 30.2 (Cx30.2)-was expressed in the organ of Corti and vestibular organs, particularly in the tectorial membrane, the base of inner and outer hair cells and the nerve fibers. CONCLUSIONS The present results describe a novel association between GJD3 and FMD, providing evidence that FMD is related to changes in the inner ear channels, and supporting a new role of tectorial membrane proteins in MD.
Collapse
Affiliation(s)
- Alba Escalera-Balsera
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitario, Ibs.GRANADA, Universidad de Granada, 18071, Granada, Spain
- Division of Otolaryngology, Department of Surgery, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - Paula Robles-Bolivar
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitario, Ibs.GRANADA, Universidad de Granada, 18071, Granada, Spain
- Division of Otolaryngology, Department of Surgery, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - Alberto M Parra-Perez
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitario, Ibs.GRANADA, Universidad de Granada, 18071, Granada, Spain
- Division of Otolaryngology, Department of Surgery, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - Silvia Murillo-Cuesta
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
- Institute for Biomedical Research Sols-Morreale (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Han Chow Chua
- Sydney Pharmacy School, Faculty of Medicine and Health and Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Lourdes Rodríguez-de la Rosa
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
- Institute for Biomedical Research Sols-Morreale (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Julio Contreras
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
- Anatomy and Embryology Department, Faculty of Veterinary, Universidad Complutense de Madrid, Madrid, Spain
| | - Ewa Domarecka
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Andrés Soto-Varela
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Division of Otoneurology, Department of Otorhinolaryngology, Complexo Hospitalario Universitario, Santiago de Compostela, Spain
- Department of Surgery and Medical-Surgical Specialities, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Isabel Varela-Nieto
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
- Institute for Biomedical Research Sols-Morreale (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Agnieszka J Szczepek
- Department of Otorhinolaryngology, Head and Neck Surgery, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Alvaro Gallego-Martinez
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitario, Ibs.GRANADA, Universidad de Granada, 18071, Granada, Spain
- Division of Otolaryngology, Department of Surgery, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - Jose A Lopez-Escamez
- Otology & Neurotology Group CTS495, Instituto de Investigación Biosanitario, Ibs.GRANADA, Universidad de Granada, 18071, Granada, Spain.
- Division of Otolaryngology, Department of Surgery, Universidad de Granada, Granada, Spain.
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain.
- Faculty of Medicine & Health, School of Medical Sciences, Meniere's Disease Neuroscience Research Program, The Kolling Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
47
|
Elamin G, Zhang Z, Dwarka D, Kasumbwe K, Mellem J, Mkhwanazi NP, Madlala P, Soliman MES. Integrative genomic analyses combined with molecular dynamics simulations reveal the impact of deleterious mutations of Bcl-2 gene on the apoptotic machinery and implications in carcinogenesis. Front Genet 2025; 15:1502152. [PMID: 39840282 PMCID: PMC11747654 DOI: 10.3389/fgene.2024.1502152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Objectives Unlike other diseases, cancer is not just a genome disease but should broadly be viewed as a disease of the cellular machinery. Therefore, integrative multifaceted approaches are crucial to understanding the complex nature of cancer biology. Bcl-2 (B-cell lymphoma 2), encoded by the human BCL-2 gene, is an anti-apoptotic molecule that plays a key role in apoptosis and genetic variation of Bcl-2 proteins and is vital in disrupting the apoptotic machinery. Single nucleotide polymorphisms (SNPs) are considered viable diagnostic and therapeutic biomarkers for various cancers. Therefore, this study explores the association between SNPs in Bcl-2 and the structural, functional, protein-protein interactions (PPIs), drug binding and dynamic characteristics. Methods Comprehensive cross-validated bioinformatics tools and molecular dynamics (MD) simulations. Multiple sequence, genetic, structural and disease phenotype analyses were applied in this study. Results Analysis revealed that out of 130 mutations, approximately 8.5% of these mutations were classified as pathogenic. Furthermore, two particular variants, namely, Bcl-2G101V and Bcl-2F104L, were found to be the most deleterious across all analyses. Following 500 ns, MD simulations showed that these mutations caused a significant distortion in the protein conformational, protein-protein interactions (PPIs), and drug binding landscape compared to Bcl-2WT. Conclusion Despite being a predictive study, the findings presented in this report would offer a perspective insight for further experimental investigation, rational drug design, and cancer gene therapy.
Collapse
Affiliation(s)
- Ghazi Elamin
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
- Department of Pharmaceutical Chemistry, College of Pharmacy, Karary University, Khartoum, Sudan
| | - Zhichao Zhang
- School of Chemistry, Dalian University of Technology, Dalian, Liaoning, China
| | - Depika Dwarka
- Ezintsha, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Kabange Kasumbwe
- Biotechnology and Food Science, Durban University of Technology, Durban, South Africa
| | - John Mellem
- Biotechnology and Food Science, Durban University of Technology, Durban, South Africa
| | - Nompumelelo P. Mkhwanazi
- HIV Pathogenesis Programme, School of Laboratory Medicine and Medical Science, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Paradise Madlala
- HIV Pathogenesis Programme, School of Laboratory Medicine and Medical Science, The Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Mahmoud E. S. Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, South Africa
| |
Collapse
|
48
|
Hermans P, Tsishyn M, Schwersensky M, Rooman M, Pucci F. Exploring Evolution to Uncover Insights Into Protein Mutational Stability. Mol Biol Evol 2025; 42:msae267. [PMID: 39786559 PMCID: PMC11721782 DOI: 10.1093/molbev/msae267] [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: 05/28/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/12/2025] Open
Abstract
Determining the impact of mutations on the thermodynamic stability of proteins is essential for a wide range of applications such as rational protein design and genetic variant interpretation. Since protein stability is a major driver of evolution, evolutionary data are often used to guide stability predictions. Many state-of-the-art stability predictors extract evolutionary information from multiple sequence alignments of proteins homologous to a query protein, and leverage it to predict the effects of mutations on protein stability. To evaluate the power and the limitations of such methods, we used the massive amount of stability data recently obtained by deep mutational scanning to study how best to construct multiple sequence alignments and optimally extract evolutionary information from them. We tested different evolutionary models and found that, unexpectedly, independent-site models achieve similar accuracy to more complex epistatic models. A detailed analysis of the latter models suggests that their inference often results in noisy couplings, which do not appear to add predictive power over the independent-site contribution, at least in the context of stability prediction. Interestingly, by combining any of the evolutionary features with a simple structural feature, the relative solvent accessibility of the mutated residue, we achieved similar prediction accuracy to supervised, machine learning-based, protein stability change predictors. Our results provide new insights into the relationship between protein evolution and stability, and show how evolutionary information can be exploited to improve the performance of mutational stability prediction.
Collapse
Affiliation(s)
- Pauline Hermans
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Matsvei Tsishyn
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, Brussels 1050, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, Brussels 1050, Belgium
| |
Collapse
|
49
|
Sun J, Zhu T, Cui Y, Wu B. Structure-based self-supervised learning enables ultrafast protein stability prediction upon mutation. Innovation (N Y) 2025; 6:100750. [PMID: 39872490 PMCID: PMC11763918 DOI: 10.1016/j.xinn.2024.100750] [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: 02/04/2024] [Accepted: 12/02/2024] [Indexed: 01/30/2025] Open
Abstract
Predicting free energy changes (ΔΔG) is essential for enhancing our understanding of protein evolution and plays a pivotal role in protein engineering and pharmaceutical development. While traditional methods offer valuable insights, they are often constrained by computational speed and reliance on biased training datasets. These constraints become particularly evident when aiming for accurate ΔΔG predictions across a diverse array of protein sequences. Herein, we introduce Pythia, a self-supervised graph neural network specifically designed for zero-shot ΔΔG predictions. Our comparative benchmarks demonstrate that Pythia outperforms other self-supervised pretraining models and force field-based approaches while also exhibiting competitive performance with fully supervised models. Notably, Pythia shows strong correlations and achieves a remarkable increase in computational speed of up to 105-fold. We further validated Pythia's performance in predicting the thermostabilizing mutations of limonene epoxide hydrolase, leading to higher experimental success rates. This exceptional efficiency has enabled us to explore 26 million high-quality protein structures, marking a significant advancement in our ability to navigate the protein sequence space and enhance our understanding of the relationships between protein genotype and phenotype. In addition, we established a web server at https://pythia.wulab.xyz to allow users to easily perform such predictions.
Collapse
Affiliation(s)
- Jinyuan Sun
- AIM Center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tong Zhu
- AIM Center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yinglu Cui
- AIM Center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Bian Wu
- AIM Center, College of Life Sciences and Technology, Beijing University of Chemical Technology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Yuan M, Chatterjee S, Leys M, Odom JV, Salido EM. Prevalence of IMPG1 and IMPG2 Mutations Leading to Retinitis Pigmentosa or Vitelliform Macular Dystrophy in a Cohort of Patients with Inherited Retinal Dystrophies. Genes (Basel) 2025; 16:43. [PMID: 39858590 PMCID: PMC11764596 DOI: 10.3390/genes16010043] [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: 11/30/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The interphotoreceptor matrix proteoglycans 1 and 2 (IMPG1 and IMPG2) are two interdependent proteoglycans of the interphotoreceptor matrix (IPM). Mutations in IMPG1 or IMPG2 are linked to retinal diseases such as retinitis pigmentosa (RP) and vitelliform macular dystrophy (VMD), yet the specific mutations responsible for each condition remain undefined. This study identifies mutations in IMPG1 and IMPG2 linked to either RP or VMD. It also provides an in-depth in silico analysis of these mutations' structural and functional impact on protein domains, alongside a detailed examination of the corresponding disease phenotypes. METHODS From a cohort of 480 patients with inherited retinal diseases (IRDs), we identified seven patients with mutations in IMPG1 or IMPG2. Multimodal imaging was performed to assess the clinical phenotypes, including fundus photography, fundus autofluorescence, fluorescein angiography, and spectral domain optical coherence tomography (SD-OCT). We provide structure modeling and analysis of each variant. RESULTS Our findings indicate a prevalence of 1.45% of IRD patients being affected by IMPG mutations; two were diagnosed with RP and five with VMD. One VMD patient carried a novel IMPG1 p.Asp423Glu mutation. Most patients exhibited heterozygous mutations, and one RP patient presented a compound heterozygous mutation in IMPG2. CONCLUSIONS This work describes a novel mutation and expands our understanding of the specific IMPG protein domains implicated in RP and VMD. Furthermore, it establishes, for the first time, the prevalence of IMPG mutations in an IRD population.
Collapse
Affiliation(s)
- Ming Yuan
- West Virginia School of Osteopathic Medicine, Lewisburg, WV 24901, USA;
| | - Souradip Chatterjee
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA;
| | - Monique Leys
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA; (M.L.); (J.V.O.)
| | - J. Vernon Odom
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA; (M.L.); (J.V.O.)
| | - Ezequiel M. Salido
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, WV 26506, USA;
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506, USA; (M.L.); (J.V.O.)
| |
Collapse
|