1
|
Luppino F, Lenz S, Chow CFW, Toth-Petroczy A. Deep learning tools predict variants in disordered regions with lower sensitivity. BMC Genomics 2025; 26:367. [PMID: 40221640 PMCID: PMC11992697 DOI: 10.1186/s12864-025-11534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The recent AI breakthrough of AlphaFold2 has revolutionized 3D protein structural modeling, proving crucial for protein design and variant effects prediction. However, intrinsically disordered regions-known for their lack of well-defined structure and lower sequence conservation-often yield low-confidence models. The latest Variant Effect Predictor (VEP), AlphaMissense, leverages AlphaFold2 models, achieving over 90% sensitivity and specificity in predicting variant effects. However, the effectiveness of tools for variants in disordered regions, which account for 30% of the human proteome, remains unclear. RESULTS In this study, we found that predicting pathogenicity for variants in disordered regions is less accurate than in ordered regions, particularly for mutations at the first N-Methionine site. Investigations into the efficacy of variant effect predictors on intrinsically disordered regions (IDRs) indicated that mutations in IDRs are predicted with lower sensitivity and the gap between sensitivity and specificity is largest in disordered regions, especially for AlphaMissense and VARITY. CONCLUSIONS The prevalence of IDRs within the human proteome, coupled with the increasing repertoire of biological functions they are known to perform, necessitated an investigation into the efficacy of state-of-the-art VEPs on such regions. This analysis revealed their consistently reduced sensitivity and differing prediction performance profile to ordered regions, indicating that new IDR-specific features and paradigms are needed to accurately classify disease mutations within those regions.
Collapse
Affiliation(s)
- Federica Luppino
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Swantje Lenz
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
| | - Chi Fung Willis Chow
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany
| | - Agnes Toth-Petroczy
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
- Center for Systems Biology Dresden, Pfotenhauerstrasse 108, 01307, Dresden, Germany.
- Cluster of Excellence Physics of Life, TU Dresden, 01062, Dresden, Germany.
| |
Collapse
|
2
|
Anteghini M, Gualdi F, Oliva B. How did we get there? AI applications to biological networks and sequences. Comput Biol Med 2025; 190:110064. [PMID: 40184941 DOI: 10.1016/j.compbiomed.2025.110064] [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: 10/28/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
The rapidly advancing field of artificial intelligence (AI) has transformed numerous scientific domains, including biology, where a vast and complex volume of data is available for analysis. This paper provides a comprehensive overview of the current state of AI-driven methodologies in genomics, proteomics, and systems biology. We discuss how machine learning algorithms, particularly deep learning models, have enhanced the accuracy and efficiency of embedding sequences, motif discovery, and the prediction of gene expression and protein structure. Additionally, we explore the integration of AI in the embedding and analysis of biological networks, including protein-protein interaction networks and multi-layered networks. By leveraging large-scale biological data, AI techniques have enabled unprecedented insights into complex biological processes and disease mechanisms. This work underlines the potential of applying AI to complex biological data, highlighting current applications and suggesting directions for future research to further explore AI in this rapidly evolving field.
Collapse
Affiliation(s)
- Marco Anteghini
- BioFolD Unit, Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy; Visual and Data-Centric Computing, Zuse Institut Berlin, Berlin, Germany.
| | - Francesco Gualdi
- Structural Bioinformatics Lab, Universitat Pompeu Fabra, Barcelona, Spain; Istituto dalle Molle di Studi sull'Intelligenza Artificiale, USI/SUPSI (Università Svizzera Italiana/Scuola Universitaria Professionale Svizzera Italiana) Lugano, Switzerland.
| | - Baldo Oliva
- Structural Bioinformatics Lab, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
3
|
Masum MHU, Mahdeen AA, Barua A. Revolutionizing Chikungunya Vaccines: mRNA Breakthroughs With Molecular and Immune Simulations. Bioinform Biol Insights 2025; 19:11779322251324859. [PMID: 40182080 PMCID: PMC11967231 DOI: 10.1177/11779322251324859] [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: 10/10/2024] [Accepted: 02/14/2025] [Indexed: 04/05/2025] Open
Abstract
With the ability to cause massive epidemics that have consequences on millions of individuals globally, the Chikungunya virus (CHIKV) emerges as a severe menace. Developing an effective vaccine is urgent as no effective therapeutics are available for such viral infections. Therefore, we designed a novel mRNA vaccine against CHIKV with a combination of highly antigenic and potential MHC-I, MHC-II, and B-cell epitopes from the structural polyprotein. The vaccine demonstrated well-characterized physicochemical properties, indicating its solubility and potential functional stability within the body (GRAVY score of -0.639). Structural analyses of the vaccine revealed a well-stabilized secondary and tertiary structure (Ramachandran score of 82.8% and a Z-score of -4.17). Docking studies of the vaccine with TLR-2 (-1027.7 KJ/mol) and TLR-4 (-1212.4 KJ/mol) exhibited significant affinity with detailed hydrogen bond interactions. Molecular dynamics simulations highlighted distinct conformational dynamics among the vaccine, "vaccine-TLR-2" and "vaccine-TLR-4" complexes. The vaccine's ability to elicit both innate and adaptive immune responses, including the presence of memory B-cells and T-cells, persistent B-cell immunity for a year, and the activation of TH cells leading to the release of IFN-γ and IL-2, has significant implications for its potential effectiveness. The CHIKV vaccine developed in this study shows promise as a potential candidate for future vaccine production against CHIKV, suggesting its suitability for further clinical advancement, including in vitro and in vivo experiments.
Collapse
Affiliation(s)
- Md. Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University, Khulshi, Chattogram, Bangladesh
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Abanti Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
4
|
Jing Y, Zhang D, Li L. H2GnnDTI: hierarchical heterogeneous graph neural networks for drug-target interaction prediction. Bioinformatics 2025; 41:btaf117. [PMID: 40097269 PMCID: PMC11954568 DOI: 10.1093/bioinformatics/btaf117] [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: 06/24/2024] [Revised: 02/10/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025] Open
Abstract
MOTIVATION Identifying drug-target interactions (DTIs) is a crucial step in drug repurposing and drug discovery. The significant increase in demand and the expensive nature for experimentally identifying DTIs necessitate computational tools for automated prediction and comprehension of DTIs. Despite recent advancements, current methods fail to fully leverage the hierarchical information in DTIs. RESULTS Here, we introduce H2GnnDTI, a novel two-level hierarchical heterogeneous graph learning model to predict DTIs, by integrating the structures of drugs and proteins via a low-level view GNN and a high-level view GNN. The hierarchical graph consists of high-level heterogeneous nodes representing drugs and proteins, connected by edges representing known DTIs. Each drug or protein node is further detailed in a low-level graph, where nodes represent molecules within each drug or amino acids within each protein, accompanied by their respective chemical descriptors. Two distinct low-level graph neural networks are first deployed to capture structural and chemical features specific to drugs and proteins from these low-level graphs. Subsequently, a high-level graph encoder (GE) is used to comprehensively capture and merge interactive features pertaining to drugs and proteins from the high-level graph. The high-level encoder incorporates a structure and attribute information fusion module designed to explicitly integrate representations acquired from both a feature encoder and a GE, facilitating consensus representation learning. Extensive experiments conducted on three benchmark datasets have shown that our proposed H2GnnDTI model consistently outperforms state-of-the-art deep learning methods. AVAILABILITY AND IMPLEMENTATION The codes are freely available at https://github.com/LiminLi-xjtu/H2GnnDTI.
Collapse
Affiliation(s)
- Yueying Jing
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Dongxue Zhang
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Limin Li
- School of Mathematics and Statistics, Xi’an Jiaotong University, Xi'an, Shaanxi 710049, China
| |
Collapse
|
5
|
Razzak A, Saha O, Sultana KF, Amin MR, Zahid AB, Sultana A, Bristi UP, Rajia S, Sarker N, Rahaman MM, Bahadur NM, Hossen F. Development of a Novel mRNA Vaccine Against Shigella Pathotypes Causing Widespread Shigellosis Endemic: An In-Silico Immunoinformatic Approach. Bioinform Biol Insights 2025; 19:11779322251328302. [PMID: 40160890 PMCID: PMC11951904 DOI: 10.1177/11779322251328302] [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: 06/29/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Shigellosis remains a major global health concern, particularly in regions with poor sanitation and limited access to clean water. This study used immunoinformatics and reverse vaccinology to design a potential mRNA vaccine targeting Shigella pathotypes out of 4071 proteins from Shigella sonnei str. Ss046, 4 key antigenic candidates were identified: putative outer membrane protein (Q3YZL0), PapC-like porin protein (Q3YZM5), putative fimbrial-like protein (Q3Z3I2), and lipopolysaccharide (LPS)-assembly protein LptD (Q3Z5V5), ensuring broad pathotype coverage. A multitope vaccine was designed incorporating cytotoxic T lymphocyte, helper T lymphocyte, and B-cell epitopes, linked with suitable linkers and adjuvants to enhance immunogenicity. Computational analyses predicted vaccine's favorable antigenicity, solubility, and stability, while molecular docking and dynamic simulations demonstrated strong binding affinity and stability with Toll-like receptor 4 (TLR-4), indicating potential for robust immune activation. Immune simulations predicted strong humoral and cellular immune responses, characterized by significant cytokine production and long-term immune memory. Structural evaluations of the complex, including radius of gyration, root mean square deviation, root mean square fluctuation, and solvent accessibility, confirmed the vaccine's structural integrity, and stability under physiological conditions. This research contributes to the ongoing effort to alleviate the global burden of Shigella infections, providing a foundation for future wet laboratory investigations aimed at vaccine development.
Collapse
Affiliation(s)
- Abdur Razzak
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Otun Saha
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Mohammad Ruhul Amin
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Abdullah bin Zahid
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Afroza Sultana
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Uditi Paul Bristi
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sultana Rajia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nikkon Sarker
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Newaz Mohammed Bahadur
- Department of Chemistry, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Foysal Hossen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
6
|
Tiwari H, Ilyas A, Rai PK, Upadhyay S, Borkotoky S. Computational investigation of antiviral peptide interactions with Mpox DNA polymerase. In Silico Pharmacol 2025; 13:49. [PMID: 40162132 PMCID: PMC11953516 DOI: 10.1007/s40203-025-00342-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
The Mpox DNA polymerase (DNA pol) plays a crucial role in the viral replication process, making it an ideal target for antiviral therapies. It facilitates the synthetic process of viral DNA, which is an integral stage in the life of a virus. The inhibition of the operation of Mpox DNA pol would interfere with the multiplication of the virus and help manage the disease. Peptides have emerged as a possible therapeutic alternative against viruses due to their distinct characteristics. Peptides have broad-spectrum antiviral activity, being effective against a variety of viruses. Using computational techniques, we attempted to explore the molecular details of the interaction between antiviral peptides and Mpox DNA pol. Two databases of antiviral peptides were screened in this study. This study used molecular docking, followed by molecular dynamics (MD) simulation and post-simulation binding energy predictions. From the 19 selected peptides with activity against DNA polymerases, two peptides-DRAVPe01393 and DRAVPe01399-were identified as particularly promising candidates. These peptides exhibited stable interactions with Mpox DNA pol and demonstrated good cell penetration potential as evident from the MD simulation studies. Notably, the peptides DRAVPe01399 and DRAVPe01393 have a better binding affinity of - 60.86 kcal/mol and - 47.92 kcal/mol respectively than the control ligand Cidofovir diphosphate (- 10.79 kcal/mol). These findings could lead to the development of innovative antiviral treatments to prevent monkeypox, helping global efforts to battle this emerging infectious disease.
Collapse
Affiliation(s)
- Harshit Tiwari
- Department of Biotechnology, Invertis University, Bareilly, India
| | - Ashal Ilyas
- Department of Biotechnology, Invertis University, Bareilly, India
| | - Pankaj Kumar Rai
- Department of Biotechnology, Invertis University, Bareilly, India
| | | | | |
Collapse
|
7
|
Morel CA, Asencio C, Moreira D, Blancard C, Salin B, Gontier E, Duvezin-Caubet S, Rojo M, Bringaud F, Tetaud E. A new member of the dynamin superfamily modulates mitochondrial membrane branching in Trypanosoma brucei. Curr Biol 2025; 35:1337-1352.e5. [PMID: 40081380 DOI: 10.1016/j.cub.2025.02.033] [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: 07/25/2024] [Revised: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025]
Abstract
Unlike most other eukaryotes, where mitochondria continuously fuse and divide, the mitochondrion of trypanosome cells forms a single and continuously interconnected network that divides only during cytokinesis. However, the machinery governing mitochondrial remodeling and interconnection of trypanosome mitochondrion remain largely unknown. We functionally characterize a new member of the dynamin superfamily protein (DSP) from T. brucei (TbMfnL), which shares similarity with a family of homologs present in various eukaryotic and prokaryotic phyla but not in opisthokonts like mammals and budding yeast. The sequence and domain organization of TbMfnL is distinct, and it is phylogenetically very distant from the yeast and mammalian dynamin-related proteins involved in mitochondrial fusion/fission dynamics, such as optic atrophy 1 (Opa1) and mitofusin (Mfn). TbMfnL localizes to the inner mitochondrial membrane facing the matrix and, upon overexpression, induces a strong increase in the interconnection and branching of mitochondrial filaments in a GTPase-dependent manner. TbMfnL is a component of a novel membrane remodeling machinery with an unprecedented matrix-side localization that is able to modulate the degree of inter-mitochondrial connections.
Collapse
Affiliation(s)
| | - Corinne Asencio
- Univ. Bordeaux, CNRS, MFP, UMR 5234, F-33000 Bordeaux, France
| | - David Moreira
- Ecologie Systématique Evolution, CNRS, Université Paris-Saclay, AgroParisTech, 91190 Gif-sur-Yvette, France
| | | | - Bénédicte Salin
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France
| | - Etienne Gontier
- Univ. Bordeaux, CNRS, INSERM, BIC, US4, UAR 3420, F-33000 Bordeaux, France
| | | | - Manuel Rojo
- Univ. Bordeaux, CNRS, IBGC, UMR 5095, F-33000 Bordeaux, France
| | | | - Emmanuel Tetaud
- Univ. Bordeaux, CNRS, MFP, UMR 5234, F-33000 Bordeaux, France.
| |
Collapse
|
8
|
Göktepe YE. Protein-protein interaction prediction using enhanced features with spaced conjoint triad and amino acid pairwise distance. PeerJ Comput Sci 2025; 11:e2748. [PMID: 40134873 PMCID: PMC11935777 DOI: 10.7717/peerj-cs.2748] [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: 10/30/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Protein-protein interactions (PPIs) are pivotal in cellular processes, influencing a wide range of functions, from metabolism to immune responses. Despite the advancements in experimental techniques for PPI detection, their inherent limitations, such as high false-positive rates and significant resource demands, necessitate the development of computational approaches. This study presents a novel computational model named MFPIC (Multi-Feature Protein Interaction Classifier) for predicting PPIs, integrating enhanced sequence-based features, including a novel spaced conjoint triad (SCT) and amino acid pairwise distance (AAPD), with existing methods such as position-specific scoring matrices (PSSM) and AAindex-based features. The SCT captures complex sequence motifs by considering non-adjacent amino acid interactions, while AAPD provides critical spatial information about amino acid residues within protein sequences. The proposed model was evaluated across three benchmark datasets-Saccharomyces cerevisiae, Helicobacter pylori, and human proteins-demonstrating superior performance in comparison to state-of-the-art models. The results underscore the efficacy of integrating diverse and complementary features, achieving significant improvements in predictive accuracy, with the model achieving 95.90%, 99.33%, and 90.95% accuracy on the Saccharomyces cerevisiae, Helicobacter pylori, and human dataset, respectively. This approach not only enhances our understanding of PPI mechanisms but also offers valuable insights for the development of targeted therapeutic strategies.
Collapse
|
9
|
Liu R, Hu C, Gao D, Li M, Yuan X, Chen L, Shu Q, Wang Z, Yang X, Dai Z, Yu H, Yang F, Zheng A, Lv M, Garg V, Jiao C, Zhang H, Hou W, Teng C, Zhou X, Du C, Xiang C, Xu D, Tang Y, Chitikineni A, Duan Y, Maalouf F, Agrawal SK, Wei L, Zhao N, Barmukh R, Li X, Wang D, Ding H, Liu Y, Chen X, Varshney RK, He Y, Zong X, Yang T. A special short-wing petal faba genome and genetic dissection of floral and yield-related traits accelerate breeding and improvement of faba bean. Genome Biol 2025; 26:62. [PMID: 40098156 PMCID: PMC11916958 DOI: 10.1186/s13059-025-03532-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND A comprehensive study of the genome and genetics of superior germplasms is fundamental for crop improvement. As a widely adapted protein crop with high yield potential, the improvement in breeding and development of the seeds industry of faba bean have been greatly hindered by its giant genome size and high outcrossing rate. RESULTS To fully explore the genomic diversity and genetic basis of important agronomic traits, we first generate a de novo genome assembly and perform annotation of a special short-wing petal faba bean germplasm (VF8137) exhibiting a low outcrossing rate. Comparative genome and pan-genome analyses reveal the genome evolution characteristics and unique pan-genes among the three different faba bean genomes. In addition, the genome diversity of 558 accessions of faba bean germplasm reveals three distinct genetic groups and remarkable genetic differences between the southern and northern germplasms. Genome-wide association analysis identifies several candidate genes associated with adaptation- and yield-related traits. We also identify one candidate gene related to short-wing petals by combining quantitative trait locus mapping and bulked segregant analysis. We further elucidate its function through multiple lines of evidence from functional annotation, sequence variation, expression differences, and protein structure variation. CONCLUSIONS Our study provides new insights into the genome evolution of Leguminosae and the genomic diversity of faba bean. It offers valuable genomic and genetic resources for breeding and improvement of faba bean.
Collapse
Affiliation(s)
- Rong Liu
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China
| | - Chaoqin Hu
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Dan Gao
- Smartgenomics Technology Institute, Tianjin, 301700, China
| | - Mengwei Li
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China
| | - Xingxing Yuan
- Institute of Industrial Crops, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, 210014, China
| | - Liyang Chen
- Smartgenomics Technology Institute, Tianjin, 301700, China
| | - Qin Shu
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China
| | - Zonghe Wang
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China
| | - Xin Yang
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Zhengming Dai
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Haitian Yu
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Feng Yang
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Aiqing Zheng
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Meiyuan Lv
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China
| | - Vanika Garg
- State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch, WA, 6150, Australia
| | - Chengzhi Jiao
- Smartgenomics Technology Institute, Tianjin, 301700, China
| | - Hongyan Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, 810016, China
- Qinghai Academy of Agricultural and Forestry Sciences, Qinghai University, Xining, Qinghai, 810016, China
| | - Wanwei Hou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, 810016, China
- Qinghai Academy of Agricultural and Forestry Sciences, Qinghai University, Xining, Qinghai, 810016, China
| | - Changcai Teng
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, 810016, China
- Qinghai Academy of Agricultural and Forestry Sciences, Qinghai University, Xining, Qinghai, 810016, China
| | - Xianli Zhou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, 810016, China
- Qinghai Academy of Agricultural and Forestry Sciences, Qinghai University, Xining, Qinghai, 810016, China
| | - Chengzhang Du
- Chongqing Academy of Agricultural Sciences, Chongqing, 401329, China
| | - Chao Xiang
- Crop Research Institute, Sichuan Academy of Agricultural Sciences, Chengdu, Sichuan, 610066, China
| | - Dongxu Xu
- Zhangjiakou Academy of Agricultural Sciences, Zhangjiakou, Hebei, 075032, China
| | - Yongsheng Tang
- Qujing Academy of Agricultural Sciences, Qujingaq, Yunnan, 655000, China
| | - Annapurna Chitikineni
- State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch, WA, 6150, Australia
| | - Yinmei Duan
- Dali Academy of Agricultural Sciences, Dali, Yunnan, 671005, China
| | - Fouad Maalouf
- International Center for Agricultural Researchin the, Dry Areas (ICARDA), Beirut, 1108-2010, Lebanon
| | - Shiv Kumar Agrawal
- International Center for Agricultural Researchin the, Dry Areas (ICARDA), Beirut, 1108-2010, Lebanon
| | - Libin Wei
- Jiangsu Yanjiang Institute of Agricultural Sciences, Nantong, Jiangsu, 226541, China
| | - Na Zhao
- Jiangsu Yanjiang Institute of Agricultural Sciences, Nantong, Jiangsu, 226541, China
| | - Rutwik Barmukh
- State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch, WA, 6150, Australia
| | - Xiang Li
- Yuxi Academy of Agricultural Sciences, Yuxi, Yunnan, 653100, China
| | - Dong Wang
- Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences/Shandong Provincial Key Laboratory of Crop Genetic Improvement, Ecology and Physiology, Jinan, Shandong, 250100, China
| | - Hanfeng Ding
- Institute of Crop Germplasm Resources, Shandong Academy of Agricultural Sciences/Shandong Provincial Key Laboratory of Crop Genetic Improvement, Ecology and Physiology, Jinan, Shandong, 250100, China
| | - Yujiao Liu
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, 810016, China.
| | - Xin Chen
- Institute of Industrial Crops, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, 210014, China.
| | - Rajeev K Varshney
- State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch, WA, 6150, Australia.
| | - Yuhua He
- Food Crops Research Institute, Yunnan Academy of Agricultural Sciences, Kunming, Yunnan, 650205, China.
| | - Xuxiao Zong
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China.
| | - Tao Yang
- State Key Laboratory of Crop Gene Resources and Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Haidian District, Beijing, 100081, China.
| |
Collapse
|
10
|
Gupta S, Sinha S, Bhakta K, Bhowmick A, Ghosh A. Unravelling the role of the A domain and N-terminal alpha-helices of FtsY in archaeal signal recognition particle. Int J Biol Macromol 2025; 306:141645. [PMID: 40032113 DOI: 10.1016/j.ijbiomac.2025.141645] [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: 10/16/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Signal recognition particle (SRP) system is critical for protein translocation across membranes in all domains of life. In archaea, this pathway relies on two GTPase proteins, SRP54 and FtsY, which interact with SRP RNA to facilitate the targeting of nascent proteins to the membrane. Although the SRP components in eukaryotes and bacteria are well characterized, the mechanisms underlying SRP-dependent membrane targeting in archaea remain poorly understood, particularly concerning the role of the FtsY N-terminal domains. This study provides an in-depth exploration of the archaeal SRP system, focusing on the N-terminal domains of the FtsY protein and their role in the formation and functionality of the targeting complex (TC). We characterized the minimal structural domains of FtsY required for SRP54 binding and membrane association, demonstrating the critical involvement of the A domain and N-terminal alpha helices in facilitating these processes. The deletion of these domains led to a progressive reduction in the affinity between SRP54 and FtsY, disrupting TC formation and compromising its catalytic efficiency. Molecular dynamics simulations and thermodynamic analyses corroborated these experimental findings, revealing that the A domain is integral to stabilizing TC and enhancing reciprocal GTP hydrolysis. Furthermore, the study showed that membrane association, mediated by the orientation of the A domain and the αN1 helix, is essential for stabilizing the interaction between SRP and the membrane. These results shed light on the molecular basis of SRP assembly and membrane targeting in archaea, marking an important advancement in our understanding of the archaeal SRP machinery.
Collapse
Affiliation(s)
- Sayandeep Gupta
- Department of Bioengineering, University of Oregon, 1505 Franklin Blvd., Eugene, OR 97403, USA
| | - Souvik Sinha
- Department of Bioengineering, University of California, 900 University Avenue, Riverside, CA 92521, USA
| | - Koustav Bhakta
- Department of Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, WB, India
| | - Arghya Bhowmick
- Department of Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, WB, India
| | - Abhrajyoti Ghosh
- Department of Biological Sciences, Bose Institute, EN 80, Sector V, Bidhan Nagar, Kolkata 700091, WB, India.
| |
Collapse
|
11
|
Xu J, Wen X, Sun L, Xing K, Xue L, Zhou S, Hu J, Ai Z, Kong Q, Wen Z, Guo L, Hao M, Xing D. Large Model Era: Deep Learning in Osteoporosis Drug Discovery. J Chem Inf Model 2025. [PMID: 40008920 DOI: 10.1021/acs.jcim.4c02264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Osteoporosis is a systemic microstructural degradation of bone tissue, often accompanied by fractures, pain, and other complications, resulting in a decline in patients' life quality. In response to the increased incidence of osteoporosis, related drug discovery has attracted more and more attention, but it is often faced with challenges due to long development cycle and high cost. Deep learning with powerful data processing capabilities has shown significant advantages in the field of drug discovery. With the development of technology, it is more and more applied to all stages of drug discovery. In particular, large models, which have been developed rapidly recently, provide new methods for understanding disease mechanisms and promoting drug discovery because of their large parameters and ability to deal with complex tasks. This review introduces the traditional models and large models in the deep learning domain, systematically summarizes their applications in each stage of drug discovery, and analyzes their application prospect in osteoporosis drug discovery. Finally, the advantages and limitations of large models are discussed in depth, in order to help future drug discovery.
Collapse
Affiliation(s)
- Junlin Xu
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Pharmacy, Qingdao University, Qingdao 266071, China
| | - Sha Zhou
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Jiayi Hu
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Zhijuan Ai
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Qian Kong
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Zishu Wen
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Li Guo
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
- School of Basic Medicine, Qingdao University, Qingdao 266071, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Alliance Manchester Business School, The University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
12
|
Gaza J, Brini E, MacCallum JL, Dill KA, Perez A. MELD in Action: Harnessing Data to Accelerate Molecular Dynamics. J Chem Inf Model 2025; 65:1685-1693. [PMID: 39893583 DOI: 10.1021/acs.jcim.4c02108] [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: 02/04/2025]
Abstract
We review MELD, an accelerator of Molecular Dynamics simulations of biomolecules. MELD (Modeling Employing Limited Data) integrates molecular dynamics (MD) with a variety of types of structural information through Bayesian inference, generating ensembles of protein and DNA structures having proper Boltzmann populations. MELD minimizes the computational sampling of irrelevant regions of phase space by applying energetic penalties to areas that conflict with the available data. MELD is effective in refining protein structures using NMR or cryo-EM data or predicting protein-ligand binding poses. As a plugin for OpenMM, MELD is interoperable with other enhanced sampling methods, offering a versatile tool for structural determination in computational chemistry and biophysics.
Collapse
Affiliation(s)
- Jokent Gaza
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| | - Emiliano Brini
- School of Chemistry and Materials Science, 85 Lomb Memorial Drive, Rochester, New York 14623, United States
| | - Justin L MacCallum
- Department of Chemistry, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Ken A Dill
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
- Department of Physics and Astronomy, Stony Brook University, Stony Brook, New York 11794, United States
| | - Alberto Perez
- Department of Chemistry, University of Florida, Gainesville, Florida 32611, United States
- Quantum Theory Project, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
13
|
Schirripa A, Schöppe H, Nebenfuehr S, Zojer M, Klampfl T, Kugler V, Maw BS, Ceylan H, Uras IZ, Scheiblecker L, Gamper E, Stelzl U, Stefan E, Kaserer T, Sexl V, Kollmann K. Cdk6's functions are critically regulated by its unique C-terminus. iScience 2025; 28:111697. [PMID: 39898030 PMCID: PMC11787673 DOI: 10.1016/j.isci.2024.111697] [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: 05/21/2024] [Revised: 08/09/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
The vital cell cycle machinery is tightly regulated and alterations of its central signaling hubs are a hallmark of cancer. The activity of CDK6 is controlled by interaction with several partners including cyclins and INK4 proteins, which have been shown to mainly bind to the amino-terminal lobe. We analyzed the impact of CDK6's C-terminus on its functions in a leukemia model, revealing a central role in promoting proliferation. C-terminally truncated Cdk6 (Cdk6 ΔC) shows reduced nuclear translocation and therefore chromatin interaction and fails to enhance proliferation and disease progression. The combination of proteomic analysis and protein modeling highlights that Cdk6's C-terminus is essential for protein flexibility and for its binding potential to cyclin D, p27Kip1 and INK4 proteins but not cyclin B. We demonstrate that the C-terminus is a unique and essential part of the CDK6 protein, regulating interaction partner binding and therefore CDK6's functionality.
Collapse
Affiliation(s)
- Alessia Schirripa
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Helge Schöppe
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Thorsten Klampfl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Valentina Kugler
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Belinda S. Maw
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Huriye Ceylan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Iris Z. Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Lisa Scheiblecker
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Elisabeth Gamper
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, Pharmaceutical Chemistry, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020 Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Teresa Kaserer
- Institute of Pharmacy/Pharmaceutical Chemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- University of Innsbruck, Innsbruck, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| |
Collapse
|
14
|
Zhang T, Yang Z, Zhang Y, Yi L, Duan F, Zhao Q, Gu Y, Wang S. Proteomics-guided isolation of a novel serine protease with milk-clotting activity from tamarillo (Solanum betaceum Cav.). Food Chem 2025; 465:141956. [PMID: 39541676 DOI: 10.1016/j.foodchem.2024.141956] [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: 07/10/2024] [Revised: 10/17/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Tamarillo is widely grown in Yunnan Province, China, and has been found that it can be used in cheese-making with a distinctive fruity flavour. However, this primary component responsible for curdling milk remains unclear. This study aimed to identify the main component in tamarillo responsible for curdling milk using proteomics and ammonium sulfate (AS) precipitation. Herein, 3199 proteins were identified in tamarillo, of which 546 exhibited hydrolase activity. In particular, a novel serine protease with milk-clotting activity (MCA) and a molecular weight of 79.1 kDa, named "MCP746", was isolated from tamarillo. The milk-clotting proteases (MCPs) from tamarillo exhibited the highest MCA at 80 °C and stability under incubation temperatures below 70 °C, pH range of 5-8, and NaCl concentrations below 1 mol/L. This study revealed that serine protease is the primary MCPs of tamarillo along with a characterization of its milk-clotting characteristics, providing valuable insights into its potential application in cheese-making.
Collapse
Affiliation(s)
- Tong Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Zhihong Yang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Yingcui Zhang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Lunzhao Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Fengmin Duan
- Yunnan Institute of Measuring and Testing Technology, Kunming 650228, China
| | - Qiong Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Ying Gu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| | - Shuo Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China; Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
15
|
Parvin R, Masum MHU, Heema HP, Akter A, Hossain MA, Siddiki AMAMZ. Designing of a multiepitope-based vaccine against echinococcosis utilizing the potent Ag5 antigen: Immunoinformatics and simulation approaches. PLoS One 2025; 20:e0310510. [PMID: 39937717 DOI: 10.1371/journal.pone.0310510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 11/13/2024] [Indexed: 02/14/2025] Open
Abstract
Echinococcosis is a significant parasitic zoonotic disease with severe implications for human and animal health. To date, there has been no effective vaccine candidate available for echinococcosis. Therefore, we employed computational approaches to develop a multiepitope-based vaccine using the most potent epitopes of MHC-I, MHC-II, and B-cell derived from the Ag5 protein of Echinococcus spp. The final vaccine construct containing the epitopes, linkers, and adjuvant exhibited potent antigenicity (score > 0.1) with no evidence of allergenicity (score < 0) and toxicity (score < 0) in several computational platforms. The vaccine also exhibited favorable physicochemical characteristics such as being highly soluble (SOLpro score of 0.781243) and hydrophilic (Grand average of hydropathy of -0.433). Moreover, the tertiary structure of the vaccine was also found to be structurally stable, with a Z score of -5.71. Further, the molecular docking analysis confirmed the vaccine's significant binding affinity to the RP-105 (docking score of -1252.7) and TLR-9 (docking score of -970.9). The molecular dynamic simulations confirmed the structural stability of the docked complexes under a virtual physiological system. The negative ΔTOTAL values derived from the MM-PBSA and MM-GBSA analyses confirmed a spontaneous and thermodynamically favorable binding process between the vaccine and receptors. Moreover, the vaccine demonstrated high potentiality to elicit both innate (natural killer cell, dendritic and macrophage) and adaptive (B-cell, helper T cell and cytotoxic T cell) immune responses with sustained humoral immune responses evidenced by increased IFN-γ and IL-2 levels. Following codon optimization and in silico cloning, the vaccine was successfully expressed (CAI value of 0.9607 and average GC content of 52.34%) after being inserted into the pET-28a (+) plasmid of E. coli. These findings highlight the potential of the designed vaccine candidate to combat echinococcosis and lay the groundwork for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Rehana Parvin
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
| | - Md Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
| | - Homaira Pervin Heema
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
| | - Aklima Akter
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
| | - Mohammad Alamgir Hossain
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
| | - A M A M Zonaed Siddiki
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Chattogram, Bangladesh
- Nextgen Informatics Ltd, Bangladesh
| |
Collapse
|
16
|
Heido J, Keng S, Poyrazoglu H, Priyam E, Jayasinghe S. Not All Bacterial Outer-Membrane Proteins Are β-Barrels. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001394. [PMID: 39989908 PMCID: PMC11845990 DOI: 10.17912/micropub.biology.001394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/25/2025]
Abstract
The discovery of Wza, an octomeric helical barrel integral bacterial outer-transmembrane protein, has challenged the widely held understanding that all integral outer-membrane proteins of Gram-negative bacteria are closed β-barrels composed of transmembrane β- strands. Wza is a member of the Outer-Membrane Polysaccharide Exporter family and our bioinformatics analysis suggests that other members of the family may also contain outer-membrane transmembrane segments that are helical. A review of the literature indicates that in addition to Wza, outer-membrane core complex proteins of the type IV secretion systems also contain transmembrane segments that are helical.
Collapse
Affiliation(s)
- John Heido
- Chemistry and Biochemistry, California State University San Marcos, San Marcos, California, United States
| | - Simon Keng
- Chemistry and Biochemistry, California State University San Marcos, San Marcos, California, United States
| | - Hulya Poyrazoglu
- Chemistry and Biochemistry, California State University San Marcos, San Marcos, California, United States
| | - Ekta Priyam
- Chemistry and Biochemistry, California State University San Marcos, San Marcos, California, United States
| | - Sajith Jayasinghe
- Chemistry and Biochemistry, California State University San Marcos, San Marcos, California, United States
| |
Collapse
|
17
|
Crone KK, Labonte JW, Elias MH, Freeman MF. α-N-Methyltransferase regiospecificity is mediated by proximal, redundant enzyme-substrate interactions. Protein Sci 2025; 34:e70021. [PMID: 39840790 PMCID: PMC11751858 DOI: 10.1002/pro.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/15/2024] [Accepted: 12/15/2024] [Indexed: 01/23/2025]
Abstract
N-Methylation of the peptide backbone confers pharmacologically beneficial characteristics to peptides that include greater membrane permeability and resistance to proteolytic degradation. The borosin family of ribosomally synthesized and post-translationally modified peptides offer a post-translational route to install amide backbone α-N-methylations. Previous work has elucidated the substrate scope and engineering potential of two examples of type I borosins, which feature autocatalytic precursors that encode N-methyltransferases that methylate their own C-termini in trans. We recently reported the first discrete N-methyltransferase and precursor peptide from Shewanella oneidensis MR-1, a minimally iterative, type IV borosin that allowed the first detailed kinetic analyses of borosin N-methyltransferases. Herein, we characterize the substrate scope and resilient regiospecificity of this discrete N-methyltransferase by comparison of relative rates and methylation patterns of over 40 precursor peptide variants along with structure analyses of nine enzyme-substrate complexes. Sequences critical to methylation are identified and demonstrated in assaying minimal peptide substrates and non-native peptide sequences for assessment of secondary structure requirements and engineering potential. This work grants understanding towards the mechanism of substrate recognition and iterative activity by discrete borosin N-methyltransferases.
Collapse
Affiliation(s)
- Kathryn K. Crone
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Twin CitiesSt. PaulMinnesotaUSA
| | - Jason W. Labonte
- Department of ChemistryNotre Dame of Maryland UniversityBaltimoreMarylandUSA
| | - Mikael H. Elias
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Twin CitiesSt. PaulMinnesotaUSA
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology InstituteUniversity of Minnesota Twin CitiesSt. PaulMinnesotaUSA
| | - Michael F. Freeman
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Twin CitiesSt. PaulMinnesotaUSA
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology InstituteUniversity of Minnesota Twin CitiesSt. PaulMinnesotaUSA
| |
Collapse
|
18
|
Omer AAM, Kumar S, Selegård R, Bengtsson T, Khalaf H. Characterization of Novel Plantaricin-Derived Antiviral Peptides Against Flaviviruses. Int J Mol Sci 2025; 26:1038. [PMID: 39940807 PMCID: PMC11817140 DOI: 10.3390/ijms26031038] [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: 12/20/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Flaviviruses, including West Nile virus, Zika virus, and Dengue virus, pose global health challenges due to their distribution, pathogenicity, and lack of effective treatments or vaccines. This study investigated the antiviral activity of novel truncated peptides derived from the two-peptide plantaricins PLNC8 αβ, PlnEF, PlnJK, and PlnA. The antiviral potential was predicted using machine learning tools, followed by in vitro evaluation against the Kunjin virus using plaque reduction assays in Vero cells. Molecular docking assessed peptide interactions with KUNV and ZIKV. Full-length and truncated peptides from PlnA, PlnE, PlnF, PlnJ, and PlnK demonstrated limited antiviral efficacy against KUNV in vitro, despite in silico predictions suggesting antiviral potential for PlnA, PlnE, and PlnJ. Large discrepancies were observed between the predicted and experimentally determined activities. However, complementary two-peptide plantaricins PlnEF and PlnJK exhibited significant synergistic effects. Furthermore, the truncated peptides PLNC8 α1-15 and PLNC8 β1-20 reduced KUNV viral load by over 90%, outperforming their full-length counterparts. Molecular docking revealed interactions of PLNC8 α and PLNC8 β, and their truncated variants, with KUNV and ZIKV, suggesting a mechanism involving viral envelope disruption. These findings highlight the potential of plantaricin-derived peptides as promising antiviral candidates against flaviviruses, warranting further investigation into their mechanisms and applications.
Collapse
Affiliation(s)
- Abubakr A. M. Omer
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden; (A.A.M.O.); (S.K.); (T.B.)
| | - Sanjiv Kumar
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden; (A.A.M.O.); (S.K.); (T.B.)
| | - Robert Selegård
- Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden;
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden; (A.A.M.O.); (S.K.); (T.B.)
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden; (A.A.M.O.); (S.K.); (T.B.)
| |
Collapse
|
19
|
Tu TH, Bennani FE, Masroori N, Liu C, Nemati A, Rozza N, Grunbaum AM, Kremer R, Milhalcioiu C, Roy DC, Rudd CE. The identification of a SARs-CoV2 S2 protein derived peptide with super-antigen-like stimulatory properties on T-cells. Commun Biol 2025; 8:14. [PMID: 39762551 PMCID: PMC11704208 DOI: 10.1038/s42003-024-07350-8] [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: 03/28/2023] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Severe COVID-19 can trigger a cytokine storm, leading to acute respiratory distress syndrome (ARDS) with similarities to superantigen-induced toxic shock syndrome. An outstanding question is whether SARS-CoV-2 protein sequences can directly induce inflammatory responses. In this study, we identify a region in the SARS-CoV-2 S2 spike protein with sequence homology to bacterial super-antigens (termed P3). Computational modeling predicts P3 binding to sites on MHC class I/II and the TCR that partially overlap with sites for the binding of staphylococcal enterotoxins B and H. Like SEB and SEH derived peptides, P3 stimulated 25-40% of human CD4+ and CD8 + T-cells, increasing IFN-γ and granzyme B production. viSNE and SPADE profiling identified overlapping and distinct IFN-γ+ and GZMB+ subsets. The super-antigenic properties of P3 were further evident by its selective expansion of T-cells expressing specific TCR Vα and Vβ chain repertoires. In vivo experiments in mice revealed that the administration of P3 led to a significant upregulation of proinflammatory cytokines IL-1β, IL-6, and TNF-α. While the clinical significance of P3 in COVID-19 remains unclear, its homology to other mammalian proteins suggests a potential role for this peptide family in human inflammation and autoimmunity.
Collapse
Affiliation(s)
- Thai Hien Tu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Fatima Ezzahra Bennani
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasser Masroori
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Chen Liu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Atena Nemati
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Nicholas Rozza
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Amichai Meir Grunbaum
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Richard Kremer
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Catalin Milhalcioiu
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
- Department of Medical Oncology, McGill University Health Center, Montreal, QC, Canada
| | - Denis-Claude Roy
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada.
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada.
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada.
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco.
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
20
|
Phung DK, Pilotto S, Matelska D, Blombach F, Pinotsis N, Hovan L, Gervasio FL, Werner F. Archaeal NusA2 is the ancestor of ribosomal protein eS7 in eukaryotes. Structure 2025; 33:149-159.e6. [PMID: 39504966 DOI: 10.1016/j.str.2024.10.019] [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: 07/05/2024] [Revised: 09/06/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
N-utilization substance A (NusA) is a regulatory factor with pleiotropic functions in gene expression in bacteria. Archaea encode two conserved small proteins, NusA1 and NusA2, with domains orthologous to the two RNA binding K Homology (KH) domains of NusA. Here, we report the crystal structures of NusA2 from Sulfolobus acidocaldarius and Saccharolobus solfataricus obtained at 3.1 Å and 1.68 Å, respectively. NusA2 comprises an N-terminal zinc finger followed by two KH-like domains lacking the GXXG signature. Despite the loss of the GXXG motif, NusA2 binds single-stranded RNA. Mutations in the zinc finger domain compromise the structural integrity of NusA2 at high temperatures and molecular dynamics simulations indicate that zinc binding provides an energy barrier preventing the domain from reaching unfolded states. A structure-guided phylogenetic analysis of the KH-like domains supports the notion that the NusA2 clade is ancestral to the ribosomal protein eS7 in eukaryotes, implying a potential role of NusA2 in translation.
Collapse
Affiliation(s)
- Duy Khanh Phung
- RNAP Laboratory, Institute for Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Simona Pilotto
- RNAP Laboratory, Institute for Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Dorota Matelska
- RNAP Laboratory, Institute for Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Fabian Blombach
- RNAP Laboratory, Institute for Structural and Molecular Biology, University College London, London WC1E 6BT, UK
| | - Nikos Pinotsis
- Institute for Structural and Molecular Biology, Birkbeck College, London WC1E 7HX, UK
| | - Ladislav Hovan
- Pharmaceutical Sciences, University of Geneva, 1206 Genève, Switzerland
| | - Francesco Luigi Gervasio
- Pharmaceutical Sciences, University of Geneva, 1206 Genève, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, 1206 Genève, Switzerland; Department of Chemistry, University College London, London WC1E 6BT, UK
| | - Finn Werner
- RNAP Laboratory, Institute for Structural and Molecular Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
21
|
Zhang J, Qian J, Zou Q, Zhou F, Kurgan L. Recent Advances in Computational Prediction of Secondary and Supersecondary Structures from Protein Sequences. Methods Mol Biol 2025; 2870:1-19. [PMID: 39543027 DOI: 10.1007/978-1-0716-4213-9_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] [Indexed: 11/17/2024]
Abstract
The secondary structures (SSs) and supersecondary structures (SSSs) underlie the three-dimensional structure of proteins. Prediction of the SSs and SSSs from protein sequences enjoys high levels of use and finds numerous applications in the development of a broad range of other bioinformatics tools. Numerous sequence-based predictors of SS and SSS were developed and published in recent years. We survey and analyze 45 SS predictors that were released since 2018, focusing on their inputs, predictive models, scope of their prediction, and availability. We also review 32 sequence-based SSS predictors, which primarily focus on predicting coiled coils and beta-hairpins and which include five methods that were published since 2018. Substantial majority of these predictive tools rely on machine learning models, including a variety of deep neural network architectures. They also frequently use evolutionary sequence profiles. We discuss details of several modern SS and SSS predictors that are currently available to the users and which were published in higher impact venues.
Collapse
Affiliation(s)
- Jian Zhang
- School of Computer and Information Technology, Xinyang Normal University, Xinyang, China.
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China.
| | - Jingjing Qian
- School of Computer and Information Technology, Xinyang Normal University, Xinyang, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, China
| | - Feng Zhou
- School of Computer and Information Technology, Xinyang Normal University, Xinyang, China
| | - Lukasz Kurgan
- Department of Computer Science, College of Engineering, Virginia Commonwealth University, Virginia, VA, USA.
| |
Collapse
|
22
|
Rivera-Asencios D, Espinoza-Culupú A, Carmen-Sifuentes S, Ramirez P, García-de-la-Guarda R. Design of a multi-epitope vaccine candidate against carrion disease by immunoinformatics approach. Comput Biol Med 2025; 184:109397. [PMID: 39566279 DOI: 10.1016/j.compbiomed.2024.109397] [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: 06/28/2024] [Revised: 10/09/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024]
Abstract
Carrion's disease, caused by the bacterium Bartonella bacilliformis, is a serious public health problem in Peru, Ecuador and Colombia. Currently there is no available vaccine against B. bacilliformis. While antibiotics are the standard treatment, resistant strains have been reported, and there is a potential spread of the vector that transmits the bacteria. This study aimed to design a multi-epitope vaccine candidate against the causative agent of Carrion's disease using immunoinformatics tools. Predictions of B-cell epitopes, as well as CD4+ and CD8+T cell epitopes, were performed from the entire proteome of B. bacilliformis KC583 using the most frequent alleles from Peru, Ecuador, Colombia, and worldwide. B-cell epitopes and T-cell nested epitopes from outer membrane and virulence-associated proteins were selected. Epitopes were filtered out based on promiscuity, non-allergenicity, conservation, non-homology and non-toxicity. Two vaccine constructs were assembled using linkers. The tertiary structure of the constructs was predicted, and their stability was evaluated through molecular dynamics simulations. The most stable construct was selected for molecular docking with the TLR4 receptor. This study proposes a vaccine construct evaluated in silico as a potential vaccine candidate against Bartonella bacilliformis.
Collapse
Affiliation(s)
- Damaris Rivera-Asencios
- Molecular Microbiology and Biotechnology Laboratory, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Abraham Espinoza-Culupú
- Molecular Microbiology and Biotechnology Laboratory, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | | | - Pablo Ramirez
- Molecular Microbiology and Biotechnology Laboratory, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Ruth García-de-la-Guarda
- Molecular Microbiology and Biotechnology Laboratory, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima, Peru.
| |
Collapse
|
23
|
Zhao B, Basu S, Kurgan L. DescribePROT Database of Residue-Level Protein Structure and Function Annotations. Methods Mol Biol 2025; 2867:169-184. [PMID: 39576581 DOI: 10.1007/978-1-0716-4196-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
DescribePROT is a freely available online database of structural and functional descriptors of proteins at the amino acid level. It provides access to 13 diverse descriptors that include sequence conservation, putative secondary structure, solvent accessibility, intrinsic disorder, and signal peptides, and putative annotations of residues that interact with proteins, peptides and nucleic acids. These data can be used to elucidate protein functions, to support efforts to develop therapeutics, and to develop and evaluate future predictors of protein structure and function. DescribePROT includes 7.8 billion predictions for 1.4 million proteins from 83 complete proteomes of popular model organisms. This information can be downloaded at multiple levels of scope (entire database, specific organisms, and individual proteins) and can be interacted with using a graphical interface that simultaneously displays data on multiple descriptors. We describe the contents of this resource, provide directions on how to use its interface, and offer instructions on how to obtain and interact with the underlying data. Moreover, we briefly discuss plans for a future expansion of this database. DescribePROT is available at http://biomine.cs.vcu.edu/servers/DESCRIBEPROT/ .
Collapse
Affiliation(s)
- Bi Zhao
- Genomics program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Sushmita Basu
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
24
|
Wu T, Cheng W, Cheng J. Improving Protein Secondary Structure Prediction by Deep Language Models and Transformer Networks. Methods Mol Biol 2025; 2867:43-53. [PMID: 39576574 DOI: 10.1007/978-1-0716-4196-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
Protein secondary structure prediction is useful for many applications. It can be considered a language translation problem, that is, translating a sequence of 20 different amino acids into a sequence of secondary structure symbols (e.g., alpha helix, beta strand, and coil). Here, we develop a novel protein secondary structure predictor called TransPross based on the transformer network and attention mechanism widely used in natural language processing to directly extract the evolutionary information from the protein language (i.e., raw multiple sequence alignment [MSA] of a protein) to predict the secondary structure. The method is different from traditional methods that first generate a MSA and then calculate expert-curated statistical profiles from the MSA as input. The attention mechanism used by TransPross can effectively capture long-range residue-residue interactions in protein sequences to predict secondary structures. Benchmarked on several datasets, TransPross outperforms the state-of-art methods. Moreover, our experiment shows that the prediction accuracy of TransPross positively correlates with the depth of MSAs, and it is able to achieve the average prediction accuracy (i.e., Q3 score) above 80% for hard targets with few homologous sequences in their MSAs. TransPross is freely available at https://github.com/BioinfoMachineLearning/TransPro .
Collapse
Affiliation(s)
- Tianqi Wu
- Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO, USA
| | - Weihang Cheng
- Department of Chemistry, Hubei University, Wuhan, Hubei, China
| | - Jianlin Cheng
- Electrical Engineering and Computer Science Department, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
25
|
Baghirov J, Zhu H, Wang X, Kihara D. Protein Secondary Structure and DNA/RNA Detection for Cryo-EM and Cryo-ET Using Emap2sec and Emap2sec . Methods Mol Biol 2025;2867:105-120. [PMID: 39576577 DOI: 10.1007/978-1-0716-4196-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
Cryo-electron microscopy (cryo-EM) has become a powerful tool for determining the structures of macromolecules, such as proteins and DNA/RNA complexes. While high-resolution cryo-EM maps are increasingly available, there is still a substantial number of maps determined at intermediate or low resolution. These maps present challenges when it comes to extracting structural information. In response to this, two computational methods, Emap2sec and Emap2sec+, have been developed by our group to address these challenges and benefit the analysis of cryo-EM maps. In this chapter, we describe how to use the web servers of two of our structure analysis software for cryo-EM, Emap2sec and Emapsec+. Both methods identify local structures in medium-resolution EM maps of 5-10 Å to help find and fit protein and DNA/RNA structures in EM maps. Emap2sec identifies the secondary structures of proteins, while Emap2sec+ also identifies DNA/RNA locations in cryo-EM maps. As cryo-electron tomogram (cryo-ET) has started to produce data of this resolution, these methods would be useful for cryo-ET, too. Both methods are available in the form of webservers and source code at https://kiharalab.org/emsuites/ .
Collapse
Affiliation(s)
- Javad Baghirov
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Han Zhu
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Xiao Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Daisuke Kihara
- Department of Computer Science, Purdue University, West Lafayette, IN, USA.
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
26
|
Roy Choudhury A, Murali A. Exploring the interaction between Fe 3+ and REGLE motif of the high-affinity iron permease (Ftr1): An in silico approach. J Mol Graph Model 2025; 134:108907. [PMID: 39550798 DOI: 10.1016/j.jmgm.2024.108907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Mucormycosis is an invasive fungal infection with high mortality rate in immunocompromised individuals. Due to COVID-19 pandemic, the disease has resurfaced recently and lack of appropriate antifungals resulted in a poor outcome in patients. The iron uptake mechanism in Rhizopus delemar, the predominant causal agent, is crucial for its survival and pathogenesis in human host. The current study is first of its kind to focus on structural dynamics of high affinity iron permease (Ftr1), a virulence factor for Mucormycosis. Ftr1 is a transmembrane protein which is responsible for transport of Fe3+ ion from extracellular milieu to cytoplasm under iron starving conditions in Rhizopus. In this work, the three-dimensional modelling of Ftr1 was carried out. The Ftr1 possessed seven transmembrane helices with N- & C-termini in extracellular and intracellular regions respectively. Moreover, the present study delineates interaction of glutamic acid residues, found in the REGLE motif of fourth transmembrane helix with Fe3+. The molecular dynamics simulation study revealed that the glycine present in the motif destabilizes the helix thereby bringing E157 closer to positively charged ion. Understanding the interaction between Fe3+ ion and Ftr1 would be helpful in designing effective small molecule drugs against this novel therapeutic target for treating mucormycosis.
Collapse
Affiliation(s)
- Ahana Roy Choudhury
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India.
| | - Ayaluru Murali
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India.
| |
Collapse
|
27
|
Kibler RD, Lee S, Kennedy MA, Wicky BIM, Lai SM, Kostelic MM, Carr A, Li X, Chow CM, Nguyen TK, Carter L, Wysocki VH, Stoddard BL, Baker D. Design of pseudosymmetric protein hetero-oligomers. Nat Commun 2024; 15:10684. [PMID: 39695145 DOI: 10.1038/s41467-024-54913-8] [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: 07/20/2023] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Pseudosymmetric hetero-oligomers with three or more unique subunits with overall structural (but not sequence) symmetry play key roles in biology, and systematic approaches for generating such proteins de novo would provide new routes to controlling cell signaling and designing complex protein materials. However, the de novo design of protein hetero-oligomers with three or more distinct chains with nearly identical structures is a challenging unsolved problem because it requires the accurate design of multiple protein-protein interfaces simultaneously. Here, we describe a divide-and-conquer approach that breaks the multiple-interface design challenge into a set of more tractable symmetric single-interface redesign tasks, followed by structural recombination of the validated homo-oligomers into pseudosymmetric hetero-oligomers. Starting from de novo designed circular homo-oligomers composed of 9 or 24 tandemly repeated units, we redesigned the inter-subunit interfaces to generate 19 new homo-oligomers and structurally recombined them to make 24 new hetero-oligomers, including ABC heterotrimers, A2B2 heterotetramers, and A3B3 and A2B2C2 heterohexamers which assemble with high structural specificity. The symmetric homo-oligomers and pseudosymmetric hetero-oligomers generated for each system have identical or nearly identical backbones, and hence are ideal building blocks for generating and functionalizing larger symmetric and pseudosymmetric assemblies.
Collapse
Affiliation(s)
- Ryan D Kibler
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Sangmin Lee
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Madison A Kennedy
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98006, USA
| | - Basile I M Wicky
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Stella M Lai
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Marius M Kostelic
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Ann Carr
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Cameron M Chow
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Tina K Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Barry L Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98006, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
28
|
Micale L, Vourlia A, Fusco C, Pracella R, Karagiannis DC, Nardella G, Vaccaro L, Leone MP, Gramazio A, Dentici ML, Aiello C, Novelli A, Xenou L, Sui Y, Eichler EE, Cacchiarelli D, Mavrothalassitis G, Castori M. Heterozygous variants disrupting the interaction of ERF with activated ERK1/2 cause microcephaly, developmental delay, and skeletal anomalies. Eur J Hum Genet 2024:10.1038/s41431-024-01721-9. [PMID: 39668184 DOI: 10.1038/s41431-024-01721-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/20/2024] [Accepted: 10/17/2024] [Indexed: 12/14/2024] Open
Abstract
Heterozygous deleterious null alleles and specific missense variants in the DNA-binding domain of the ETS2 repressor factor (ERF) cause craniosynostosis, while the recurrent p.(Tyr89Cys) missense variant is associated with Chitayat syndrome. Exome and whole transcriptome sequencing revealed the ERF de novo in-frame indel c.911_913del selectively removing the serine of the FSF motif, which interacts with the extracellular signal-regulated kinases (ERKs), in a 10-year-old girl with microcephaly, multiple congenital joint dislocations, generalized joint hypermobility, and Pierre-Robin sequence. Three additional cases with developmental delay variably associated with microcephaly, Pierre-Robin sequence and minor skeletal anomalies were detected carrying heterozygous de novo non-truncating alleles (two with c.911_913del and one with the missense c.907 T > A change) in the same FSF motif. Protein affinity maps, co-immunoprecipitation experiments and subcellular distribution showed that both the variants impair the interaction between ERF and activated ERK1/2 and increase ERF nuclear localization, affecting ERF repressor activity that may lead to developmental defects. Our work expands the phenotypic spectrum of ERF-related disorders to a pleiotropic condition with microcephaly, developmental delay and skeletal anomalies, that we termed MIDES syndrome, and adds to the understanding of the relevance of the ERF-ERK interaction in human development and disease.
Collapse
Affiliation(s)
- Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy.
| | - Aikaterini Vourlia
- IMBB, FORTH, 71003, Heraklion, Crete, Greece
- Medical School, University of Crete, 71003, Heraklion, Crete, Greece
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy
| | - Riccardo Pracella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy
| | | | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy
| | - Lorenzo Vaccaro
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Pia Leone
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy
| | - Antonio Gramazio
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Lisa Dentici
- Rare Diseases and Medical Genetics, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Chiara Aiello
- Translational Cytogenetics, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenetics, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Lydia Xenou
- IMBB, FORTH, 71003, Heraklion, Crete, Greece
| | - Yang Sui
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Davide Cacchiarelli
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - George Mavrothalassitis
- IMBB, FORTH, 71003, Heraklion, Crete, Greece.
- Medical School, University of Crete, 71003, Heraklion, Crete, Greece.
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Italy
| |
Collapse
|
29
|
Kutz J, Schmietendorf H, Rahman SA, Opel F, Pospiech H. HROB Is Implicated in DNA Replication. Genes (Basel) 2024; 15:1587. [PMID: 39766854 PMCID: PMC11675949 DOI: 10.3390/genes15121587] [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: 10/27/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
DNA replication represents a series of precisely regulated events performed by a complex protein machinery that guarantees accurate duplication of the genetic information. Since DNA replication is permanently faced by a variety of exogenous and endogenous stressors, DNA damage response, repair and replication must be closely coordinated to maintain genomic integrity. HROB has been identified recently as a binding partner and activator of the Mcm8/9 helicase involved in DNA interstrand crosslink (ICL) repair. We identified HROB independently as a nuclear protein whose expression is co-regulated with various DNA replication factors. Accordingly, the HROB protein level showed a maximum in S phase and a downregulation in quiescence. Structural prediction and homology searches revealed that HROB is a largely intrinsically disordered protein bearing a helix-rich region and a canonical oligonucleotide/oligosaccharide-binding-fold motif that originated early in eukaryotic evolution. Employing a flow cytometry Förster resonance energy transfer (FRET) assay, we detected associations between HROB and proteins of the DNA replication machinery. Moreover, ectopic expression of HROB protein led to an almost complete shutdown of DNA replication. The available data imply a function for HROB during DNA replication across barriers such as ICLs.
Collapse
Affiliation(s)
- Julia Kutz
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Hannes Schmietendorf
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Sheikh Anika Rahman
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Franz Opel
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Department of Medical Engineering and Biotechnology, Ernst-Abbe University of Applied Sciences, D-07745 Jena, Germany
| | - Helmut Pospiech
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
- Department of Obstetrics and Gynecology, University Hospital Düsseldorf and Heinrich-Heine University, D-40225 Düsseldorf, Germany
| |
Collapse
|
30
|
Zhang J, Yang Y, Wang B, Qiu W, Zhang H, Qiu Y, Yuan J, Dong R, Zha Y. Developing a universal multi-epitope protein vaccine candidate for enhanced borna virus pandemic preparedness. Front Immunol 2024; 15:1427677. [PMID: 39703502 PMCID: PMC11655343 DOI: 10.3389/fimmu.2024.1427677] [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: 05/04/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Borna disease virus 1 (BoDV-1) is an emerging zoonotic RNA virus that can cause severe acute encephalitis with high mortality. Currently, there are no effective countermeasures, and the potential risk of a future outbreak requires urgent attention. To address this challenge, the complete genome sequence of BoDV-1 was utilized, and immunoinformatics was applied to identify antigenic peptides suitable for vaccine development. Methods Immunoinformatics and antigenicity-focused protein screening were employed to predict B-cell linear epitopes, B-cell conformational epitopes, and cytotoxic T lymphocyte (CTL) epitopes. Only overlapping epitopes with antigenicity greater than 1 and non-toxic, non-allergenic properties were selected for subsequent vaccine construction. The epitopes were linked using GPGPG linkers, incorporating β-defensins at the N-terminus to enhance immune response, and incorporating Hit-6 at the C-terminus to improve protein solubility and aid in protein purification. Computational tools were used to predict the immunogenicity, physicochemical properties, and structural stability of the vaccine. Molecular docking was performed to predict the stability and dynamics of the vaccine in complex with Toll-like receptor 4 (TLR-4) and major histocompatibility complex I (MHC I) receptors. The vaccine construct was cloned through in silico restriction to create a plasmid for expression in a suitable host. Results Among the six BoDV-1 proteins analyzed, five exhibited high antigenicity scores. From these, eight non-toxic, non-allergenic overlapping epitopes with antigenicity scores greater than 1 were selected for vaccine development. Computational predictions indicated favorable immunogenicity, physicochemical properties, and structural stability. Molecular docking analysis showed that the vaccine remained stable in complex with TLR-4 and MHC I receptors, suggesting strong potential for immune recognition. A plasmid construct was successfully generated, providing a foundation for the experimental validation of vaccines in future pandemic scenarios. Discussion These findings demonstrate the potential of the immunoinformatics-designed multi-epitope vaccines for the prevention and treatment of BoDV-1. Relevant preparations were made in advance for possible future outbreaks and could be quickly utilized for experimental verification.
Collapse
Affiliation(s)
- Jingjing Zhang
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Youfang Yang
- Department of Nephrology, The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Binyu Wang
- School of Medicine, Guizhou University, Guiyang, China
| | - Wanting Qiu
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Helin Zhang
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yuyang Qiu
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jing Yuan
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Rong Dong
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yan Zha
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
31
|
Meng C, Hou Y, Zou Q, Shi L, Su X, Ju Y. Rore: robust and efficient antioxidant protein classification via a novel dimensionality reduction strategy based on learning of fewer features. Genomics Inform 2024; 22:29. [PMID: 39633440 PMCID: PMC11616364 DOI: 10.1186/s44342-024-00026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 10/03/2024] [Indexed: 12/07/2024] Open
Abstract
In protein identification, researchers increasingly aim to achieve efficient classification using fewer features. While many feature selection methods effectively reduce the number of model features, they often cause information loss caused by merely selecting or discarding features, which limits classifier performance. To address this issue, we present Rore, an algorithm based on a feature-dimensionality reduction strategy. By mapping the original features to a latent space, Rore retains all relevant feature information while using fewer representations of the latent features. This approach significantly preserves the original information and overcomes the information loss problem associated with previous feature selection. Through extensive experimental validation and analysis, Rore demonstrated excellent performance on an antioxidant protein dataset, achieving an accuracy of 95.88% and MCC of 91.78%, using vectors including only 15 features. The Rore algorithm is available online at http://112.124.26.17:8021/Rore .
Collapse
Affiliation(s)
- Chaolu Meng
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research and Application of Agriculture and Animal Husbandry, Hohhot, China
| | - Yongqi Hou
- School of Computer Science, Inner Mongolia University, Hohhot, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Shi
- Department of Spine Surgery, Changzheng Hospital, Naval Medical University, Huangpu District, No. 415, Fengyang Road, Shanghai, China
| | - Xi Su
- Foshan Women and Children Hospital, Foshan, China
| | - Ying Ju
- School of Informatics, Xiamen University, Xiamen, China.
| |
Collapse
|
32
|
Peeyatu C, Prompat N, Voravuthikunchai SP, Roongsawang N, Sangkhathat S, Khongkow P, Saetang J, Tipmanee V. Role of Non-Binding T63 Alteration in IL-18 Binding. Int J Mol Sci 2024; 25:12992. [PMID: 39684709 DOI: 10.3390/ijms252312992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Engineered interleukin-18 (IL-18) has attracted interest as a cytokine-based treatment. However, knowledge-based mutagenesis of IL-18 has been reported for only a few regions of the protein structures, including binding sites I and II. When coupled with the binding region mutant (E6K), the non-binding residue of IL-18, Thr63 (T63), has been shown to increase the flexibility of the binding loop. Nevertheless, the function of Thr63 in conformational regulation is still unknown. Using homology modeling, molecular dynamics simulation, and structural analysis, we investigated the effects of Thr63 alteration coupling with E6K on conformational change pattern, binding loop flexibility, and the hydrogen bond network. The results indicate that the 63rd residue was significantly associated with hydrogen-bond relaxation at the core β-barrel binding sites I and II Glu85-Ile100 loop. This result provided conformational and flexible effects to binding sites I and III by switching their binding loops and stabilizing the 63rd residue cavity. These findings may pave the way for the conceptualization of a new design for IL-18 proteins by modifying non-binding residues for structure-based drug development.
Collapse
Affiliation(s)
- Chariya Peeyatu
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Napat Prompat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Supayang Piyawan Voravuthikunchai
- Center of Antimicrobial Biomaterial Innovation-Southeast Asia and Natural Product Research Center of Excellent, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Niran Roongsawang
- Microbial Cell Factory Research Team, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Surasak Sangkhathat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Pasarat Khongkow
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Institute of Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Jirakrit Saetang
- EZ-Mol-Design Laboratory, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Songkhla 90112, Thailand
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Institute of Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- EZ-Mol-Design Laboratory, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
33
|
Takata S, Kawano S, Mine A, Mise K, Takano Y, Ohtsu M, Kaido M. Unveiling crucial amino acid residues in the red clover necrotic mosaic virus movement protein for dynamic subcellular localization and viral cell-to-cell movement. Virology 2024; 600:110215. [PMID: 39255728 DOI: 10.1016/j.virol.2024.110215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
Emerging evidence suggests that the localization of viral movement proteins (MPs) to both plasmodesmata (PD) and viral replication complexes (VRCs) is the key to viral cell-to-cell movement. However, the molecular mechanism that establishes the subcellular localization of MPs is not fully understood. Here, we investigated the PD localization pathway of red clover necrotic mosaic virus (RCNMV) MP and the functional regions of MP that are crucial for MP localization to PD and VRCs. Disruption analysis of the transport pathway suggested that RCNMV MP does not rely on the ER-Golgi pathway or the cytoskeleton for the localization to the PD. Furthermore, mutagenesis analysis identified amino acid residues within the alpha helix regions responsible for localization to the PD or VRCs. These α-helix regions were also essential for efficient viral cell-to-cell movement, highlighting the importance of these dynamic localization of the MPs for viral infection.
Collapse
Affiliation(s)
- Shota Takata
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Saho Kawano
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Akira Mine
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Kazuyuki Mise
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Yoshitaka Takano
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan
| | - Mina Ohtsu
- Laboratory of Plant Symbiosis, Graduate School of Science and Technology, Nara Institute of Science and Technology, Nara, 630-0192, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Saitama, 332-0012, Japan
| | - Masanori Kaido
- Laboratory of Plant Pathology, Graduate School of Agriculture, Kyoto University, Kyoto, 606-8502, Japan.
| |
Collapse
|
34
|
Nascimento KS, Morada RCV, Oliveira MV, Martins FWV, Sacramento-Neto JC, Cavada BS. Purification and partial characterization of a new melibiose-specific lectin from Bauhinia catingae Harms. Int J Biol Macromol 2024; 282:136564. [PMID: 39414198 DOI: 10.1016/j.ijbiomac.2024.136564] [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: 06/11/2024] [Revised: 09/28/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Lectins are ubiquitous proteins that selectively bind to carbohydrates, serving as vital models for understanding protein-carbohydrate interactions. While extensively distributed across various life forms, plant lectins, especially from the Leguminosae family, have garnered significant attention. However, limited research exists on lectins from the Caesalpinioideae subfamily, suggesting a source of untapped biotechnological potential. This underscores the imperative for further exploration, particularly in isolating lectins from the Bauhinia genus, which remains relatively understudied, despite harboring lectins with diverse characteristics and promising biotechnological activities. In this study, a novel lectin extracted from Bauhinia catingae Harms seeds (BCL) was isolated in three chromatographic steps. BCL exhibited affinity for galactose and derivatives, akin to other Bauhinia lectins, with SDS-PAGE confirming its molecular weight around 30 kDa. Notably, BCL demonstrated stability across temperature and pH ranges and lacked metalloprotein characteristics. Electrospray ionization mass spectrometry revealed a partial sequence covering 81 % of the total protein sequence with nearly 80 % identity to Bauhinia forficata. Structural analysis suggested a β-sheet-rich secondary structure similar to that of other lectins. Further structural elucidation of BCL is essential to unveil its full potential and applications.
Collapse
Affiliation(s)
- Kyria S Nascimento
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil.
| | - Rebeca Cristian V Morada
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil
| | - Messias V Oliveira
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil
| | - Francisco William V Martins
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil
| | - José Carlos Sacramento-Neto
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil
| | - Benildo S Cavada
- Federal University of Ceara, Department of Biochemistry and Molecular Biology, Laboratory of Biologically Active Molecules, José Aurelio Camara, 60.440-970 Fortaleza, CE, Brazil.
| |
Collapse
|
35
|
Berdieva M, Kalinina V, Palii O, Skarlato S. Putative MutS2 Homologs in Algae: More Goods in Shopping Bag? J Mol Evol 2024; 92:815-833. [PMID: 39365456 DOI: 10.1007/s00239-024-10210-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024]
Abstract
MutS2 proteins are presumably involved in either control of recombination or translation quality control in bacteria. MutS2 homologs have been found in plants and some algae; however, their actual diversity in eukaryotes remains unknown. We found putative MutS2 homologs in various species of photosynthetic eukaryotes and performed a detailed analysis of the revealed amino acid sequences. Three groups of homologs were distinguished depending on their domain composition: MutS2 homologs with full set of specific domains, MutS2-like sequences without endonuclease Smr domain, and MutS2-like homologs lacking Smr and clamp in domain IV, the extreme form of which are proteins with only a complete ATPase domain. We clarified the information about amino acid composition and set of specific motifs in the conserved domains in MutS2 and MutS2-like sequences. The models of the predicted tertiary structure were obtained for each group of homologs. The phylogenetic analysis demonstrated that all eukaryotic sequences split into two large groups. The first group included homologs belonging to species of Archaeplastida and a subset of haptophyte homologs, while the second-sequences of organisms from CASH groups (cryptophytes, alveolates, stramenopiles, haptophytes) and chlorarachniophytes. The cyanobacterial MutS2 clustered together with the first group, and proteins belonging to Deltaproteobacteria (orders Myxococcales and Bradymonadales) showed phylogenetic affinity to the CASH-including group with strong support. The observed tree pattern did not support a clear differentiation of eukaryotes into lineages with red and green algae-derived plastids. The results are discussed in the context of current conceptions of serial endosymbioses and genetic mosaicism in algae with complex plastids.
Collapse
Affiliation(s)
- Mariia Berdieva
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | - Vera Kalinina
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Olga Palii
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Sergei Skarlato
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| |
Collapse
|
36
|
Duan Y, Ma L, Zhao T, Liu J, Zheng C, Song F, Tian L, Cai W, Li H. Conserved A-to-I RNA editing with non-conserved recoding expands the candidates of functional editing sites. Fly (Austin) 2024; 18:2367359. [PMID: 38889318 PMCID: PMC11188811 DOI: 10.1080/19336934.2024.2367359] [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: 03/06/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
Adenosine-to-inosine (A-to-I) RNA editing recodes the genome and confers flexibility for the organisms to adapt to the environment. It is believed that RNA recoding sites are well suited for facilitating adaptive evolution by increasing the proteomic diversity in a temporal-spatial manner. The function and essentiality of a few conserved recoding sites are recognized. However, the experimentally discovered functional sites only make up a small corner of the total sites, and there is still the need to expand the repertoire of such functional sites with bioinformatic approaches. In this study, we define a new category of RNA editing sites termed 'conserved editing with non-conserved recoding' and systematically identify such sites in Drosophila editomes, figuring out their selection pressure and signals of adaptation at inter-species and intra-species levels. Surprisingly, conserved editing sites with non-conserved recoding are not suppressed and are even slightly overrepresented in Drosophila. DNA mutations leading to such cases are also favoured during evolution, suggesting that the function of those recoding events in different species might be diverged, specialized, and maintained. Finally, structural prediction suggests that such recoding in potassium channel Shab might increase ion permeability and compensate the effect of low temperature. In conclusion, conserved editing with non-conserved recoding might be functional as well. Our study provides novel aspects in considering the adaptive evolution of RNA editing sites and meanwhile expands the candidates of functional recoding sites for future validation.
Collapse
Affiliation(s)
| | | | | | - Jiyao Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Caiqing Zheng
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Fan Song
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Li Tian
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Wanzhi Cai
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Hu Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
37
|
Pawar SV, Banini WSK, Shamsuddeen MM, Jumah TA, Dolling NNO, Tiamiyu A, Awe OI. Prostruc: an open-source tool for 3D structure prediction using homology modeling. Front Chem 2024; 12:1509407. [PMID: 39717221 PMCID: PMC11664737 DOI: 10.3389/fchem.2024.1509407] [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: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 12/25/2024] Open
Abstract
Introduction Homology modeling is a widely used computational technique for predicting the three-dimensional (3D) structures of proteins based on known templates,evolutionary relationships to provide structural insights critical for understanding protein function, interactions, and potential therapeutic targets. However, existing tools often require significant expertise and computational resources, presenting a barrier for many researchers. Methods Prostruc is a Python-based homology modeling tool designed to simplify protein structure prediction through an intuitive, automated pipeline. Integrating Biopython for sequence alignment, BLAST for template identification, and ProMod3 for structure generation, Prostruc streamlines complex workflows into a user-friendly interface. The tool enables researchers to input protein sequences, identify homologous templates from databases such as the Protein Data Bank (PDB), and generate high-quality 3D structures with minimal computational expertise. Prostruc implements a two-stage vSquarealidation process: first, it uses TM-align for structural comparison, assessing Root Mean Deviations (RMSD) and TM scores against reference models. Second, it evaluates model quality via QMEANDisCo to ensure high accuracy. Results The top five models are selected based on these metrics and provided to the user. Prostruc stands out by offering scalability, flexibility, and ease of use. It is accessible via a cloud-based web interface or as a Python package for local use, ensuring adaptability across research environments. Benchmarking against existing tools like SWISS-MODEL,I-TASSER and Phyre2 demonstrates Prostruc's competitive performance in terms of structural accuracy and job runtime, while its open-source nature encourages community-driven innovation. Discussion Prostruc is positioned as a significant advancement in homology modeling, making high-quality protein structure prediction more accessible to the scientific community.
Collapse
Affiliation(s)
- Shivani V. Pawar
- Department of Biotechnology and Bioinformatics, Deogiri College, Auranagabad, Maharashtra, India
| | - Wilson Sena Kwaku Banini
- Department of Theoretical and Applied Biology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Musa Muhammad Shamsuddeen
- Department of Public Health, Faculty of Health Sciences, National Open University of Nigeria, Abuja, Nigeria
| | - Toheeb A. Jumah
- School of Collective Intelligence, University Mohammed VI Polytechnic, Rabat, Morocco
| | - Nigel N. O. Dolling
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Abdulwasiu Tiamiyu
- School of Collective Intelligence, University Mohammed VI Polytechnic, Rabat, Morocco
| | - Olaitan I. Awe
- African Society for Bioinformatics and Computational Biology, Cape Town, South Africa
| |
Collapse
|
38
|
Zhang C, Wang Q, Li Y, Teng A, Hu G, Wuyun Q, Zheng W. The Historical Evolution and Significance of Multiple Sequence Alignment in Molecular Structure and Function Prediction. Biomolecules 2024; 14:1531. [PMID: 39766238 PMCID: PMC11673352 DOI: 10.3390/biom14121531] [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: 09/26/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Multiple sequence alignment (MSA) has evolved into a fundamental tool in the biological sciences, playing a pivotal role in predicting molecular structures and functions. With broad applications in protein and nucleic acid modeling, MSAs continue to underpin advancements across a range of disciplines. MSAs are not only foundational for traditional sequence comparison techniques but also increasingly important in the context of artificial intelligence (AI)-driven advancements. Recent breakthroughs in AI, particularly in protein and nucleic acid structure prediction, rely heavily on the accuracy and efficiency of MSAs to enhance remote homology detection and guide spatial restraints. This review traces the historical evolution of MSA, highlighting its significance in molecular structure and function prediction. We cover the methodologies used for protein monomers, protein complexes, and RNA, while also exploring emerging AI-based alternatives, such as protein language models, as complementary or replacement approaches to traditional MSAs in application tasks. By discussing the strengths, limitations, and applications of these methods, this review aims to provide researchers with valuable insights into MSA's evolving role, equipping them to make informed decisions in structural prediction research.
Collapse
Affiliation(s)
- Chenyue Zhang
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China; (C.Z.); (Y.L.); (G.H.)
| | - Qinxin Wang
- Suzhou New & High-Tech Innovation Service Center, Suzhou 215011, China;
| | - Yiyang Li
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China; (C.Z.); (Y.L.); (G.H.)
| | - Anqi Teng
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511453, China;
| | - Gang Hu
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China; (C.Z.); (Y.L.); (G.H.)
| | - Qiqige Wuyun
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Wei Zheng
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin 300071, China; (C.Z.); (Y.L.); (G.H.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
39
|
Kim M, Bezprozvanny I. Biological function of Aβ peptides revealed by analysis of membrane-association properties: Implications for Azheimer's disease pathogenesis. Biochem Biophys Res Commun 2024; 734:150611. [PMID: 39222574 DOI: 10.1016/j.bbrc.2024.150611] [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: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Proteolytic processing of amyloid precursor protein (APP) plays a critical role in the pathogenesis of Azheimer's disease (AD). Sequential cleavage of APP by β and γ secretases leads to generation of Aβ40 (non-amyloidogenic) and Aβ42 (amyloidogenic) peptides. Despite intense studies, the biological function of these peptides and the mechanism of Aβ42 toxicity is poorly understood. In the previous publications we proposed that association of Aβ peptides with the endosomal membranes may have important implications for pathogenesis of AD (Kim and Bezprozvanny, IJMS, 2021, vol 22, 13600; Kim and Bezprozvanny, IJMS, 2023, vol 24, 2092). To understand potential biological importance of such interaction, we focused on the region of Aβ peptides involved in peri-membrane association (E682 to N698). We discovered that association of this region with the membranes is reminiscent of several known anti-microbial peptides (AMP) such as PA13, Aurein1.2 and BP100. Our analysis further revealed that energy of peri-membrane association of Aβ40 is significantly weaker than for Aβ42 or AMP peptides, but it can be increased in the presence of non-amyloidogenic FAD mutations or in the presence of cholesterol in the membrane. Based on similarity with established mechanism of action of AMP peptides, we propose that Aβ peptides affect the curvature of endosomal membranes and shift the balance between endosomal recycling to plasma membrane and late endosomal/lysosomal pathway. We further propose that these effects are enhanced as a result of non-amyloidogenic FAD mutations in the sequence of Aβ peptides or in the presence of cholesterol in the membrane. The proposed model provides potential mechanistic explanation to synaptic defects induced by increased levels of Aβ42, by non-amyloidogenic FAD mutations in APP and by age-related increase in the levels of cholesterol in the brain.
Collapse
Affiliation(s)
- Meewhi Kim
- Dept of Physiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Ilya Bezprozvanny
- Dept of Physiology, UT Southwestern Medical Center, Dallas, TX, 75390, USA; Laboratory of Molecular Neurodegeneration, St Petersburg State Polytechnical Universty, St Petersburg, 195251, Russian Federation.
| |
Collapse
|
40
|
Liu J, Zhang Z, Pu W, Pan X, Li P, Bai Q, Liang S, Li C, Yu Y, Yao H, Ma J. A multi-epitope subunit vaccine providing broad cross-protection against diverse serotypes of Streptococcus suis. NPJ Vaccines 2024; 9:216. [PMID: 39543108 PMCID: PMC11564553 DOI: 10.1038/s41541-024-01015-7] [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: 08/08/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024] Open
Abstract
Streptococcus suis infection represents a major challenge in pig farming and public health due to its zoonotic potential and diverse serotypes, while existing vaccines lack effective cross-protection. This study employed reverse vaccinology and immunoinformatics to identify 8 conserved proteins across 11 prevalent serotypes of S. suis. 16 candidate epitopes were selected to design three multi-epitope antigens against S. suis (designated as MEASs), which fused with a dendritic cell-targeting peptide to improve antigen presentation in host. Purified MEASs displayed favorable cross-reactogenicity against 29 serotype-specific antiserums. Robust humoral and cellular immune responses can be induced by MEAS 1 and MEAS 3 in a mouse model, which provided substantial protection against virulent strains from two different serotypes. In particular, their immune serums exhibited positive opsonization effects within bloodstream and macrophage phagocytosis. Taken together, we identified two promising MEASs with excellent cross-protection, offering potential in preventing S. suis infections in a mouse model.
Collapse
Affiliation(s)
- Jianan Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Zhen Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Wanxia Pu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Xinming Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Pei Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Qiankun Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Song Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Caiying Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China
| | - Yong Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| | - Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, 210095, China.
- WOAH Reference Lab for Swine Streptococcosis, Nanjing, 210095, China.
| |
Collapse
|
41
|
Ramos-Sevillano E, Ercoli G, Betts M, Guerra-Assunção JA, Iverson A, Frank M, Partridge F, Lo SW, Fernandes VE, Nasher F, Wall E, Wren B, Gordon SB, Ferreira DM, Heyderman R, Rosch J, Brown JS. Essential role of proline synthesis and the one-carbon metabolism pathways for systemic virulence of Streptococcus pneumoniae. mBio 2024; 15:e0175824. [PMID: 39422467 PMCID: PMC11559097 DOI: 10.1128/mbio.01758-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 07/09/2024] [Indexed: 10/19/2024] Open
Abstract
Virulence screens have indicated potential roles during Streptococcus pneumoniae infection for the one-carbon metabolism pathway component Fhs and proline synthesis mediated by ProABC. To define how these metabolic pathways affect S. pneumoniae virulence, we have investigated the phenotypes, transcription, and metabolic profiles of Δfhs and ΔproABC mutants. S. pneumoniae capsular serotype 6B BHN418 Δfhs and ΔproABC mutant strains had strongly reduced virulence in mouse sepsis and pneumonia models but could colonize the nasopharynx. Both mutant strains grew normally in complete media but had markedly impaired growth in chemically defined medium, human serum, and human cerebrospinal fluid. The BHN418 ΔproABC strain also had impaired growth under conditions of osmotic and oxidative stress. The virulence role of proABC was strain specific, as the D39 ΔproABC strain could still cause septicemia and grow in serum. Compared to culture in broth, in serum, the BHN418 Δfhs and ΔproABC strains showed considerable derangement in global gene transcription that affected multiple but different metabolic pathways for each mutant strain. Metabolic data suggested that Δfhs had an impaired stringent response, and when cultured in sera, BHN418 Δfhs and ΔproABC were under increased oxidative stress and had altered lipid profiles. Loss of proABC also affected carbohydrate metabolism and the accumulation of peptidoglycan synthesis precursors in the BHN418 but not the D39 background, linking this phenotype to the conditional virulence phenotype. These data identify the S. pneumoniae metabolic functions affected by S. pneumoniae one-carbon metabolism and proline biosynthesis, and the role of these genetic loci for establishing systemic infection.IMPORTANCERapid adaptation to grow within the physiological conditions found in the host environment is an essential but poorly understood virulence requirement for systemic pathogens such as Streptococcus pneumoniae. We have now demonstrated an essential role for the one-carbon metabolism pathway and a conditional role depending on strain background for proline biosynthesis for S. pneumoniae growth in serum or cerebrospinal fluid, and therefore for systemic virulence. RNAseq and metabolomic data demonstrated that the loss of one-carbon metabolism or proline biosynthesis has profound but differing effects on S. pneumoniae metabolism in human serum, identifying the metabolic processes dependent on each pathway during systemic infection. These data provide a more detailed understanding of the adaptations required by systemic bacterial pathogens in order to cause infection and demonstrate that the requirement for some of these adaptations varies between strains from the same species and could therefore underpin strain variations in virulence potential.
Collapse
Affiliation(s)
- Elisa Ramos-Sevillano
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London, Rayne Institute, London, United Kingdom
| | - Giuseppe Ercoli
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London, Rayne Institute, London, United Kingdom
| | - Modupeh Betts
- Research Department of Infection, Division of Infection and Immunity, University College London, Rayne Institute, London, United Kingdom
| | | | - Amy Iverson
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Matthew Frank
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Frederick Partridge
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London, Rayne Institute, London, United Kingdom
- School of Life Sciences, University of Westminster, London, United Kingdom
| | - Stephanie W. Lo
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Vitor E. Fernandes
- Faculdade de Medicina e Ciências Biomédicas and ABC-RI. Faro, Faro, Portugal
| | - Fauzy Nasher
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emma Wall
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London, Rayne Institute, London, United Kingdom
| | - Brendan Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stephen B. Gordon
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme Blantyre, Blantyre, Malawi
| | | | - Rob Heyderman
- Research Department of Infection, Division of Infection and Immunity, University College London, Rayne Institute, London, United Kingdom
| | - Jason Rosch
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremy S. Brown
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College London, Rayne Institute, London, United Kingdom
| |
Collapse
|
42
|
Hota S, Kumar M. Unveiling the impact of Leptospira TolC efflux protein on host tissue adherence, complement evasion, and diagnostic potential. Infect Immun 2024; 92:e0041924. [PMID: 39392312 PMCID: PMC11556070 DOI: 10.1128/iai.00419-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
The TolC family protein of Leptospira is a type I outer membrane efflux protein. Phylogenetic analysis revealed significant sequence conservation among pathogenic Leptospira species (83%-98% identity) compared with intermediate and saprophytic species. Structural modeling indicated a composition of six β-strands and 10 α-helices arranged in two repeats, resembling bacterial outer membrane efflux proteins. Recombinant TolC (rTolC), expressed in a heterologous host and purified via Ni-NTA chromatography, maintained its secondary structural integrity, as verified by circular dichroism spectroscopy. Polyclonal antibodies against rTolC detected native TolC expression in pathogenic Leptospira but not in nonpathogenic ones. Immunoassays and detergent fractionation assays indicated surface localization of TolC. The rTolC's recognition by sera from leptospirosis-infected hosts across species suggests its utility as a diagnostic marker. Notably, rTolC demonstrated binding affinity for various extracellular matrix components, including collagen and chondroitin sulfate A, as well as plasma proteins such as factor H, C3b, and plasminogen, indicating potential roles in tissue adhesion and immune evasion. Functional assays demonstrated that rTolC-bound FH retained cofactor activity for C3b cleavage, highlighting TolC's role in complement regulation. The rTolC protein inhibited both the alternative and the classical pathway-mediated membrane attack complex (MAC) deposition in vitro. Blocking surface-expressed TolC on leptospires using specific antibodies reduced FH acquisition by Leptospira and increased MAC deposition on the spirochete. These findings indicate that TolC contributes to leptospiral virulence by promoting host tissue colonization and evading the immune response, presenting it as a potential target for diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Saswat Hota
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Manish Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
43
|
Li S, Peng L, Chen L, Que L, Kang W, Hu X, Ma J, Di Z, Liu Y. Discovery of Highly Bioactive Peptides through Hierarchical Structural Information and Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:8164-8175. [PMID: 39466714 DOI: 10.1021/acs.jcim.4c01006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Peptide drugs play an essential role in modern therapeutics, but the computational design of these molecules is hindered by several challenges. Traditional methods like molecular docking and molecular dynamics (MD) simulation, as well as recent deep learning approaches, often face limitations related to computational resource demands, complex binding affinity assessments, extensive data requirements, and poor model interpretability. Here, we introduce PepHiRe, an innovative methodology that utilizes the hierarchical structural information in peptide sequences and employs a novel strategy called Ladderpath, rooted in algorithmic information theory, to rapidly generate and enhance the efficiency and clarity of novel peptide design. We applied PepHiRe to develop BH3-like peptide inhibitors targeting myeloid cell leukemia-1, a protein associated with various cancers. By analyzing just eight known bioactive BH3 peptide sequences, PepHiRe effectively derived a hierarchy of subsequences used to create new BH3-like peptides. These peptides underwent screening through MD simulations, leading to the selection of five candidates for synthesis and subsequent in vitro testing. Experimental results demonstrated that these five peptides possess high inhibitory activity, with IC50 values ranging from 28.13 ± 7.93 to 167.42 ± 22.15 nM. Our study explores a white-box model driven technique and a structured screening pipeline for identifying and generating novel peptides with potential bioactivity.
Collapse
Affiliation(s)
- Shu Li
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Lu Peng
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Liuqing Chen
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Linjie Que
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Wenqingqing Kang
- Centre of Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
| | - Xiaojun Hu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Jun Ma
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Zengru Di
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| | - Yu Liu
- Department of Systems Science, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
- International Academic Center of Complex Systems, Beijing Normal University, Zhuhai 519087, China
| |
Collapse
|
44
|
Chowdhury S, Sadhukhan P, Mahata N. Immunoinformatics investigation on pathogenic Escherichia coli proteome to develop an epitope-based peptide vaccine candidate. Mol Divers 2024:10.1007/s11030-024-11034-0. [PMID: 39516450 DOI: 10.1007/s11030-024-11034-0] [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: 08/06/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Escherichia coli (E. coli), a gram-negative bacterium, quickly colonizes in the human gastrointestinal tract after birth and typically sustains a long-term, symbiotic relationship with the host. However, certain virulent strains of E. coli can cause diseases such as urinary tract infections, meningitis, and enteric disorders. The rising antibiotic resistance among these strains has heightened the urgency for an effective vaccine. This study employs immunoinformatics and a reverse vaccinology technique to identify prospective antigens and create an efficient vaccine construct. In this study, we reported the "Attaching and Effacing Protein" a novel outer-membrane protein conserved in all pathogenic E. coli strains, based on proteome screening. We developed an in silico multi-epitope vaccine that includes helper T lymphocyte (HTL), cytotoxic T lymphocyte (CTL), B cell lymphocyte (BCL), and pan HLA DR-binding reactive epitope (PADRE) sequences, along with appropriate linkers and adjuvants. Machine Learning algorithms were used to evaluate antigenicity, solubility, stability, and non-allergenicity of the vaccine construct. Additionally, molecular docking analysis revealed that vaccine construct has a strong predicted binding affinity for human toll-like receptors on the cell surface. In this context, laboratory validations are necessary to demonstrate the effectiveness of the possible vaccine design that showed encouraging findings through computational validation.
Collapse
Affiliation(s)
- Soham Chowdhury
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Pinkan Sadhukhan
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| |
Collapse
|
45
|
Masum MHU, Mahdeen AA, Barua L, Parvin R, Heema HP, Ferdous J. Developing a chimeric multiepitope vaccine against Nipah virus (NiV) through immunoinformatics, molecular docking and dynamic simulation approaches. Microb Pathog 2024; 197:107098. [PMID: 39521154 DOI: 10.1016/j.micpath.2024.107098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Nipah virus (NiV) is a highly lethal zoonotic pathogen that poses a significant threat to human and animal health. Unfortunately, no effective treatments have been developed for this deadly zoonotic disease. Therefore, we designed a chimeric multiepitope vaccine targeting the Nipah virus (NiV) glycoprotein and fusion protein through immunoinformatic approaches. Therefore, the vaccine was developed by combining promising and potential antigenic MHC-I, MHC-II, and B-cell epitopes obtained from the selected proteins. When combined, the MHC-I and MHC-II epitopes offered 100 % global population coverage. The physicochemical characterization also exhibited favorable properties, including solubility and potential functional stability of the vaccine within the body (GRAVY score of -0.308). Structural analyses unveiled a well-stabilized secondary and tertiary structure with a Ramachandran score of 84.4 % and a Z score of -5.02. Findings from docking experiments with TLR-2 (-1089.3 kJ/mol) and TLR-4 (-1016.7 kJ/mol) showed a strong affinity of the vaccine towards the receptor. Molecular dynamics simulations revealed unique conformational dynamics among the "vaccine-apo," "vaccine-TLR-2," and "vaccine-TLR-4″ complexes. Consequently, the complexes exhibited significant compactness, flexibility, and exposure to solvents. The results of the codon optimization were remarkable, as the vaccine showed a significant amount of expression in the E. coli vector (GC content of 45.36 % and a CAI score of 1.0). The results of immune simulations, however, showed evidence of both adaptive and innate immune responses induced by the vaccine. Therefore, we highly recommend further research on this chimeric multiepitope vaccine to establish its efficacy and safety against the Nipah virus (NiV).
Collapse
Affiliation(s)
- Md Habib Ullah Masum
- Department of Genomics and Bioinformatics, Faculty of Biotechnology and Genetic Engineering, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh.
| | - Ahmad Abdullah Mahdeen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Logon Barua
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Rehana Parvin
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh
| | - Homaira Pervin Heema
- Genomics Research Group, Department of Pathology and Parasitology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University (CVASU), Khulshi, 4225, Chattogram, Bangladesh
| | - Jannatul Ferdous
- Department of Obstetrics and Gynecology, Chittagong Medical College Hospital, Chattogram, 4203, Bangladesh
| |
Collapse
|
46
|
Versini R, Baaden M, Cavellini L, Cohen MM, Taly A, Fuchs PFJ. Lys716 in the transmembrane domain of yeast mitofusin Fzo1 modulates anchoring and fusion. Structure 2024; 32:1997-2012.e7. [PMID: 39299234 DOI: 10.1016/j.str.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/06/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Outer mitochondrial membrane fusion, a vital cellular process, is mediated by mitofusins. However, the underlying molecular mechanism remains elusive. We have performed extensive multiscale molecular dynamics simulations to predict a model of the transmembrane (TM) domain of the yeast mitofusin Fzo1. Coarse-grained simulations of the two TM domain helices, TM1 and TM2, reveal a stable interface, which is controlled by the charge status of residue Lys716. Atomistic replica-exchange simulations further tune our model, which is confirmed by a remarkable agreement with an independent AlphaFold2 (AF2) prediction of Fzo1 in complex with its fusion partner Ugo1. Furthermore, the presence of the TM domain destabilizes the membrane, even more if Lys716 is charged, which can be an asset for initiating fusion. The functional role of Lys716 was confirmed with yeast experiments, which show that mutating Lys716 to a hydrophobic residue prevents mitochondrial fusion.
Collapse
Affiliation(s)
- Raphaëlle Versini
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France; Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Marc Baaden
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France
| | - Laetitia Cavellini
- Laboratoire de Biologie Cellulaire et Moléculaire des Eucaryotes, Institut de Biologie Physico-Chimique, UMR 8226, CNRS, Sorbonne Université, Paris, France
| | - Mickaël M Cohen
- Laboratoire de Biologie Cellulaire et Moléculaire des Eucaryotes, Institut de Biologie Physico-Chimique, UMR 8226, CNRS, Sorbonne Université, Paris, France
| | - Antoine Taly
- Laboratoire de Biochimie Théorique, CNRS, Université Paris Cité, 75005 Paris, France.
| | - Patrick F J Fuchs
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France; Université Paris Cité, 75006 Paris, France.
| |
Collapse
|
47
|
Douradinha B. Computational strategies in Klebsiella pneumoniae vaccine design: navigating the landscape of in silico insights. Biotechnol Adv 2024; 76:108437. [PMID: 39216613 DOI: 10.1016/j.biotechadv.2024.108437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/07/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
The emergence of multidrug-resistant Klebsiella pneumoniae poses a grave threat to global public health, necessitating urgent strategies for vaccine development. In this context, computational tools have emerged as indispensable assets, offering unprecedented insights into klebsiellal biology and facilitating the design of effective vaccines. Here, a review of the application of computational methods in the development of K. pneumoniae vaccines is presented, elucidating the transformative impact of in silico approaches. Through a systematic exploration of bioinformatics, structural biology, and immunoinformatics techniques, the complex landscape of K. pneumoniae pathogenesis and antigenicity was unravelled. Key insights into virulence factors, antigen discovery, and immune response mechanisms are discussed, highlighting the pivotal role of computational tools in accelerating vaccine development efforts. Advancements in epitope prediction, antigen selection, and vaccine design optimisation are examined, highlighting the potential of in silico approaches to update vaccine development pipelines. Furthermore, challenges and future directions in leveraging computational tools to combat K. pneumoniae are discussed, emphasizing the importance of multidisciplinary collaboration and data integration. This review provides a comprehensive overview of the current state of computational contributions to K. pneumoniae vaccine development, offering insights into innovative strategies for addressing this urgent global health challenge.
Collapse
|
48
|
Dong S, Chen C, Li J, Liu Y, Bayer EA, Lamed R, Mizrahi I, Cui Q, Feng Y. Unique Fn3-like biosensor in σ I/anti-σ I factors for regulatory expression of major cellulosomal scaffoldins in Pseudobacteroides cellulosolvens. Protein Sci 2024; 33:e5193. [PMID: 39470320 PMCID: PMC11520246 DOI: 10.1002/pro.5193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024]
Abstract
Lignocellulolytic clostridia employ multiple pairs of alternative σ/anti-σ (SigI/RsgI) factors to regulate cellulosomal components for substrate-specific degradation of cellulosic biomass. The current model has proposed that RsgIs use a sensor domain to bind specific extracellular lignocellulosic components and activate cognate SigIs to initiate expression of corresponding cellulosomal enzyme genes, while expression of scaffoldins can be initiated by several different SigIs. Pseudobacteroides cellulosolvens contains the most complex known cellulosome system and the highest number of SigI-RsgI regulons yet discovered. However, the function of many RsgI sensor domains and their relationship with the various enzyme types are not fully understood. Here, we report that RsgI4 from P. cellulosolvens employs a C-terminal module that bears distant similarity to the fibronectin type III (Fn3) domain and serves as the sensor domain. Substrate-binding analysis revealed that the Fn3-like domain of RsgI4 represents a novel carbohydrate-binding module (CBM) that binds to a wide range of polysaccharide types. Structure determination further revealed that the Fn3-like domain belongs to the type B group of CBMs with a predicted concave face for substrate binding. Promoter sequence analysis of cellulosomal genes revealed that SigI4 is responsible for cellulosomal regulation of major scaffoldins rather than enzymes, consistent with the broad substrate specificity of the RsgI4 sensor domain. Notably, scaffoldins are invariably required as cellulosome components regardless of the substrate type. These findings suggest that the intricate cellulosome system of P. cellulosolvens comprises a more elaborate regulation mechanism than other bacteria and thus expands the paradigm of cellulosome regulation.
Collapse
Affiliation(s)
- Sheng Dong
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chao Chen
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jie Li
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
- Present address:
Department of BiochemistryUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Ya‐Jun Liu
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
| | - Edward A. Bayer
- Department of Biomolecular SciencesThe Weizmann Institute of ScienceRehovotIsrael
- Department of Life Sciences and the National Institute for Biotechnology in the NegevBen‐Gurion University of the NegevBeershebaIsrael
| | - Raphael Lamed
- Department of Molecular Microbiology and BiotechnologyTel Aviv UniversityTel AvivIsrael
| | - Itzhak Mizrahi
- Department of Life Sciences and the National Institute for Biotechnology in the NegevBen‐Gurion University of the NegevBeershebaIsrael
| | - Qiu Cui
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoShandongChina
| | - Yingang Feng
- CAS Key Laboratory of Biofuels, Shandong Provincial Key Laboratory of Synthetic Biology, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Qingdao Engineering Laboratory of Single Cell Oil, Shandong Engineering Laboratory of Single Cell Oil, Qingdao New Energy Shandong Laboratory, Qingdao Institute of Bioenergy and Bioprocess TechnologyChinese Academy of SciencesQingdaoChina
- Shandong Energy InstituteQingdaoChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
49
|
Calamari ZT, Song A, Cohen E, Akter M, Das Roy R, Hallikas O, Christensen MM, Li P, Marangoni P, Jernvall J, Klein OD. Bank vole genomics links determinate and indeterminate growth of teeth. BMC Genomics 2024; 25:1000. [PMID: 39472825 PMCID: PMC11523675 DOI: 10.1186/s12864-024-10901-2] [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: 06/23/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Continuously growing teeth are an important innovation in mammalian evolution, yet genetic regulation of continuous growth by stem cells remains incompletely understood. Dental stem cells responsible for tooth crown growth are lost at the onset of tooth root formation. Genetic signaling that initiates this loss is difficult to study with the ever-growing incisor and rooted molars of mice, the most common mammalian dental model species, because signals for root formation overlap with signals that pattern tooth size and shape (i.e., cusp patterns). Bank and prairie voles (Cricetidae, Rodentia, Glires) have evolved rooted and unrooted molars while retaining similar size and shape, providing alternative models for studying roots. RESULTS We assembled a de novo genome of Myodes glareolus, a vole with high-crowned, rooted molars, and performed genomic and transcriptomic analyses in a broad phylogenetic context of Glires (rodents and lagomorphs) to assess differential selection and evolution in tooth forming genes. Bulk transcriptomics comparisons of embryonic molar development between bank voles and mice demonstrated overall conservation of gene expression levels, with species-specific differences corresponding to the accelerated and more extensive patterning of the vole molar. We leverage convergent evolution of unrooted molars across the clade to examine changes that may underlie the evolution of unrooted molars. We identified 15 dental genes with changing synteny relationships and six dental genes undergoing positive selection across Glires, two of which were undergoing positive selection in species with unrooted molars, Dspp and Aqp1. Decreased expression of both genes in prairie voles with unrooted molars compared to bank voles supports the presence of positive selection and may underlie differences in root formation. CONCLUSIONS Our results support ongoing evolution of dental genes across Glires and identify candidate genes for mechanistic studies of root formation. Comparative research using the bank vole as a model species can reveal the complex evolutionary background of convergent evolution for ever-growing molars.
Collapse
Affiliation(s)
- Zachary T Calamari
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY, 10010, USA.
- The Graduate Center, City University of New York, 365 Fifth Ave, New York, NY, 10016, USA.
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Division of Paleontology, American Museum of Natural History, Central Park West at 79th Street, New York, NY, 10024, USA.
| | - Andrew Song
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY, 10010, USA
- Cornell University, 616 Thurston Ave, Ithaca, NY, 14853, USA
| | - Emily Cohen
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY, 10010, USA
- New York University College of Dentistry, 345 E 34th St, New York, NY, 10010, USA
| | - Muspika Akter
- Baruch College, City University of New York, One Bernard Baruch Way, New York, NY, 10010, USA
| | - Rishi Das Roy
- Institute of Biotechnology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Outi Hallikas
- Institute of Biotechnology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Mona M Christensen
- Institute of Biotechnology, University of Helsinki, Helsinki, FI-00014, Finland
| | - Pengyang Li
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA, 90048, USA
- Department of Bioengineering, Stanford University, 443 Via Ortega, Rm 119, Stanford, CA, 94305, USA
| | - Pauline Marangoni
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA, 90048, USA
| | - Jukka Jernvall
- Institute of Biotechnology, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Geosciences and Geography, University of Helsinki, Helsinki, FI-00014, Finland
| | - Ophir D Klein
- Program in Craniofacial Biology, Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Pediatrics, Cedars-Sinai Guerin Children's, 8700 Beverly Blvd., Suite 2416, Los Angeles, CA, 90048, USA.
| |
Collapse
|
50
|
Verma P, Thakur D, Pandit SB. Exon nomenclature and classification of transcripts database (ENACTdb): a resource for analyzing alternative splicing mediated proteome diversity. BIOINFORMATICS ADVANCES 2024; 4:vbae157. [PMID: 39569321 PMCID: PMC11576355 DOI: 10.1093/bioadv/vbae157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/20/2024] [Accepted: 10/27/2024] [Indexed: 11/22/2024]
Abstract
Motivation Gene transcripts are distinguished by the composition of their exons, and this different exon composition may contribute to advancing proteome complexity. Despite the availability of alternative splicing information documented in various databases, a ready association of exonic variations to the protein sequence remains a mammoth task. Results To associate exonic variation(s) with the protein systematically, we designed the Exon Nomenclature and Classification of Transcripts (ENACT) framework for uniquely annotating exons that tracks their loci in gene architecture context with encapsulating variations in splice site(s) and amino acid coding status. After ENACT annotation, predicted protein features (secondary structure/disorder/Pfam domains) are mapped to exon attributes. Thus, ENACTdb provides trackable exonic variation(s) association to isoform(s) and protein features, enabling the assessment of functional variation due to changes in exon composition. Such analyses can be readily performed through multiple views supported by the server. The exon-centric visualizations of ENACT annotated isoforms could provide insights on the functional repertoire of genes due to alternative splicing and its related processes and can serve as an important resource for the research community. Availability and implementation The database is publicly available at https://www.iscbglab.in/enactdb/. It contains protein-coding genes and isoforms for Caenorhabditis elegans, Drosophila melanogaster, Danio rerio, Mus musculus, and Homo sapiens.
Collapse
Affiliation(s)
- Paras Verma
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, Punjab, 140306, India
| | - Deeksha Thakur
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, Punjab, 140306, India
| | - Shashi B Pandit
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)-Mohali, Punjab, 140306, India
| |
Collapse
|