1
|
Najar Najafi N, Karbassian R, Hajihassani H, Azimzadeh Irani M. Unveiling the influence of fastest nobel prize winner discovery: alphafold's algorithmic intelligence in medical sciences. J Mol Model 2025; 31:163. [PMID: 40387957 DOI: 10.1007/s00894-025-06392-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
CONTEXT AlphaFold's advanced AI technology has transformed protein structure interpretation. By predicting three-dimensional protein structures from amino acid sequences, AlphaFold has solved the complex protein-folding problem, previously challenging for experimental methods due to numerous possible conformations. Since its inception, AlphaFold has introduced several versions, including AlphaFold2, AlphaFold DB, AlphaFold Multimer, Alpha Missense, and AlphaFold3, each further enhancing protein structure prediction. Remarkably, AlphaFold is recognized as the fastest Nobel Prize winner in science history. This technology has extensive applications, potentially transforming treatment and diagnosis in medical sciences by reducing drug design costs and time, while elucidating structural pathways of human body systems. Numerous studies have demonstrated how AlphaFold aids in understanding health conditions by providing critical information about protein mutations, abnormal protein-protein interactions, and changes in protein dynamics. Researchers have also developed new technologies and pipelines using different versions of AlphaFold to amplify its potential. However, addressing existing limitations is crucial to maximizing AlphaFold's capacity to redefine medical research. This article reviews AlphaFold's impact on five key aspects of medical sciences: protein mutation, protein-protein interaction, molecular dynamics, drug design, and immunotherapy. METHODS This review examines the contributions of various AlphaFold versions AlphaFold2, AlphaFold DB, AlphaFold Multimer, Alpha Missense, and AlphaFold3 to protein structure prediction. The methods include an extensive analysis of computational techniques and software used in interpreting and predicting protein structures, emphasizing advances in AI technology and its applications in medical research.
Collapse
Affiliation(s)
- Niki Najar Najafi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Reyhaneh Karbassian
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Helia Hajihassani
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
2
|
Micsonai A, Wien F, Murvai N, Nyiri MP, Balatoni B, Lee YH, Molnár T, Goto Y, Jamme F, Kardos J. BeStSel: analysis site for protein CD spectra-2025 update. Nucleic Acids Res 2025:gkaf378. [PMID: 40357643 DOI: 10.1093/nar/gkaf378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/18/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
Circular dichroism (CD) spectroscopy is a widely used technique to characterize the secondary structure composition of proteins. We have developed the Beta Structure Selection (BeStSel) method (PNAS, 112, E3095), which solves the main problem of protein CD spectroscopy-namely, the spectral variability of β-structures. The BeStSel web server utilizes this method to provide tools to the community for CD spectrum analysis. BeStSel uniquely provides information on eight secondary structure components, including parallel β-structure and antiparallel β-sheets with three different twist groups. It outperforms all available methods in accuracy and information content, and is also able to predict protein folds down to the topology/homology level of the CATH classification. The algorithm has been further developed, and the accuracy of the estimation of the secondary structure elements is improved by 0.7% as an average on the reference dataset. A new module of the web server calculates protein stability from the thermal denaturation profile followed by CD. Secondary structure calculations of uploaded PDB and mmcif files support the experimental verification of MD simulations and AlphaFold models by CD spectroscopy. Well-proven modules for disorder-order classification and extinction coefficient calculation continue to work. The BeStSel server is freely accessible at https://bestsel.elte.hu.
Collapse
Affiliation(s)
- András Micsonai
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
- ELTE-Functional Nucleic Acid Motifs Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Frank Wien
- Synchrotron SOLEIL, Gif-sur-Yvette 91192, France
| | - Nikoletta Murvai
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
- ELTE-Functional Nucleic Acid Motifs Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Márton Péter Nyiri
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Bori Balatoni
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Young-Ho Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Gyeonggi 17546, Republic of Korea
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Tamás Molnár
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| | - Yuji Goto
- Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | | | - József Kardos
- Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest H-1117, Hungary
| |
Collapse
|
3
|
Chi H, Wan J, Melin AD, DeCasien AR, Wang S, Zhang Y, Cui Y, Guo X, Zhao L, Williamson J, Zhang T, Li Q, Zhan Y, Li N, Guo J, Xu Z, Hou W, Cao Y, Yuan J, Zheng J, Shao Y, Wang J, Chen W, Song S, Lu X, Qi X, Zhang G, Rossiter SJ, Wu DD, Liu Y, Lu H, Li G. Genomic and phenotypic evidence support visual and olfactory shifts in primate evolution. Nat Ecol Evol 2025; 9:721-733. [PMID: 40021902 DOI: 10.1038/s41559-025-02651-5] [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: 08/27/2023] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Sensory trade-offs between vision and olfaction in the evolution and radiation of primates have long been debated. However, insights have been limited by a lack of sensory gene sequences and accompanying functional predictions. Here we conduct large-scale functional analyses of visual and olfactory receptors and related brain regions across extant primates. Our results reveal a visual shift from ultraviolet to violet colour sensitivity in early haplorrhine primates, followed by acceleration in the rhodopsin retinal release rates at the origin of anthropoids, both of which are expected to greatly enhance visual acuity under brighter light conditions. Additionally, we find that the sensitivity of olfactory receptors shifted from narrowly to broadly tuned early in anthropoid evolution. In contrast, strepsirrhines appear to have retained sensitive dim-light vision and underwent functional enhancement of narrowly tuned olfactory receptors. Our models indicate that this would have enhanced odorant discrimination and facilitated olfaction-mediated physiology and behaviour. These differences in tuning patterns of olfactory receptors between major primate lineages mirror well-established morphological differences in external anatomy and brain structures, revealing new mechanisms of olfactory adaptation and evolutionary plasticity. Our multisystem analyses reveal patterns of co-evolution in genomic, molecular and neuroanatomical traits that are consistent with a sensory 'reallocation' rather than strict trade-offs.
Collapse
Affiliation(s)
- Hai Chi
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jiahui Wan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Amanda D Melin
- Department of Anthropology and Archaeology, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Alex R DeCasien
- Computational and Evolutionary Neurogenomics Unit, National Institute on Aging, Bethesda, MD, USA
| | - Sufang Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yudan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yimeng Cui
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Xin Guo
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Le Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
- QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C., School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, China
| | - Joseph Williamson
- School of Biological and Behavioural Sciences, Queen Mary, University of London, London, UK
| | - Tianmin Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Qian Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yue Zhan
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Na Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jinqu Guo
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zhe Xu
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wenhui Hou
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yumin Cao
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jiaqing Yuan
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jiangmin Zheng
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yong Shao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jinhong Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Wu Chen
- Guangzhou Zoo & Guangzhou Wildlife Research Center, Guangzhou, China
| | - Shengjing Song
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xiaoli Lu
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Xiaoguang Qi
- Shaanxi Key Laboratory for Animal Conservation, College of Life Sciences, Northwest University, Xi'an, China
| | - Guojie Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- BGI-Shenzhen, Shenzhen, China
- Villum Center for Biodiversity Genomics, Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stephen J Rossiter
- School of Biological and Behavioural Sciences, Queen Mary, University of London, London, UK
| | - Dong-Dong Wu
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Natural History Museum of Zoology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| | - Yang Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, China.
| | - Huimeng Lu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.
| | - Gang Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, China.
- QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C., School of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, China.
| |
Collapse
|
4
|
Mirinezhad MR, Mirzaei F, Salmaninejad A, Esfehani RJ, Seyedtaghia MR, Farahmand S, Toosi MB, Hashemian S, Lewis MES. Reporting a Homozygous Case of Neurodevelopmental Disorder Associated With a Novel PRPF8 Variant. Mol Genet Genomic Med 2025; 13:e70084. [PMID: 40066647 PMCID: PMC11894437 DOI: 10.1002/mgg3.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND While recently identified heterozygous PRPF8 variants have been linked to various human diseases, their role in neurodevelopmental disorders (NDDs) remains ambiguous. This study investigates the potential association between homozygous PRPF8 variants and NDDs. Most PRPF8 variants are primarily associated with retinal diseases; however, we analyze a family with multiple members diagnosed with NDDs. METHODS Using exome sequencing (ES), the cause of behavioral problems and intellectual disabilities (IDs) of two sisters from a consanguineous parents was solved, and the results confirmed by direct sanger sequencing method likewise protein modeling to assess the structural impact of the identified variant on the PRPF8 protein has been done. RESULTS ES identified a novel homozygous variant, PRPF8 c.257G>T, p.R86M. To the best of our knowledge at the time of writing this manuscript, the mentioned variant has not been reported in relation to NDDs. Protein modeling provided another line of evidence proving the pathogenicity of the novel variant. CONCLUSION Our findings indicate that the p.R86M variant may disrupt normal protein function by changing its structure and probably its interaction, potentially leading to the observed neurodevelopmental phenotypes. This study highlights the first link between the PRPF8 variant and NDDs, suggesting a distinct role for specific PRPF8 variants in the etiology of NDDs. These results warrant further investigation into the mechanisms by which PRPF8 variants contribute to NDDs, emphasizing the need for comprehensive genetic screening in families with unexplained neurodevelopmental conditions.
Collapse
Affiliation(s)
- Mohammad Reza Mirinezhad
- Department of Medical GeneticsFaculty of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - Farzaneh Mirzaei
- Department of Medical GeneticsFaculty of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - Arash Salmaninejad
- Department of Medical GeneticsFaculty of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - Reza Jafarzadeh Esfehani
- Department of Medical GeneticsFaculty of Medicine, Mashhad University of Medical SciencesMashhadIran
- Blood Borne Infections Research Center, Academic Center for Education, Culture & Research (ACECR)Razavi Khorasan BranchMashhadIran
| | - Mohammad Reza Seyedtaghia
- Department of Medical Genetics, Faculty of MedicineHormozgan University of Medical SciencesBandar AbbasIran
| | - Sheyda Farahmand
- Department of BiologyMashhad Branch, Islamic Azad UniversityMashhadIran
| | - Mehran Beiraghi Toosi
- Pediatric WardSchool of Medicine, Mashhad University of Medical SciencesMashhadIran
- Pediatric Neurology Research CenterMashhad University of Medical SciencesMashhadIran
| | | | - M. E. Suzzane Lewis
- Department of Medical GeneticsUniversity of British Columbia (UBC)VancouverBritish ColumbiaCanada
- BC Children's Hospital Research InstituteVancouverBritish ColumbiaCanada
| |
Collapse
|
5
|
Rosignoli S, Pacelli M, Manganiello F, Paiardini A. An outlook on structural biology after AlphaFold: tools, limits and perspectives. FEBS Open Bio 2025; 15:202-222. [PMID: 39313455 PMCID: PMC11788754 DOI: 10.1002/2211-5463.13902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/19/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
AlphaFold and similar groundbreaking, AI-based tools, have revolutionized the field of structural bioinformatics, with their remarkable accuracy in ab-initio protein structure prediction. This success has catalyzed the development of new software and pipelines aimed at incorporating AlphaFold's predictions, often focusing on addressing the algorithm's remaining challenges. Here, we present the current landscape of structural bioinformatics shaped by AlphaFold, and discuss how the field is dynamically responding to this revolution, with new software, methods, and pipelines. While the excitement around AI-based tools led to their widespread application, it is essential to acknowledge that their practical success hinges on their integration into established protocols within structural bioinformatics, often neglected in the context of AI-driven advancements. Indeed, user-driven intervention is still as pivotal in the structure prediction process as in complementing state-of-the-art algorithms with functional and biological knowledge.
Collapse
Affiliation(s)
- Serena Rosignoli
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Maddalena Pacelli
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Francesca Manganiello
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| | - Alessandro Paiardini
- Department of Biochemical sciences “A. Rossi Fanelli”Sapienza Università di RomaItaly
| |
Collapse
|
6
|
Shen X, Zhang Y, Li J, Zhou Y, Butensky S, Zhang Y, Cai Z, DeWan AT, Khan SA, Yan H, Johnson CH, Zhu F. OncoSexome: the landscape of sex-based differences in oncologic diseases. Nucleic Acids Res 2025; 53:D1443-D1459. [PMID: 39535034 PMCID: PMC11701605 DOI: 10.1093/nar/gkae1003] [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: 08/06/2024] [Revised: 09/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The NIH policy on sex as biological variable (SABV) emphasized the importance of sex-based differences in precision oncology. Over 50% of clinically actionable oncology genes are sex-biased, indicating differences in drug efficacy. Research has identified sex differences in non-reproductive cancers, highlighting the need for comprehensive sex-based cancer data. We therefore developed OncoSexome, a multidimensional knowledge base describing sex-based differences in cancer (https://idrblab.org/OncoSexome/) across four key topics: antineoplastic drugs and responses (SDR), oncology-related biomarkers (SBM), risk factors (SRF) and microbial landscape (SML). SDR covers sex-based differences in 2051 anticancer drugs; SBM describes 12 551 sex-differential biomarkers; SRF illustrates 350 sex-dependent risk factors; SML demonstrates 1386 microbes with sex-differential abundances associated with cancer development. OncoSexome is unique in illuminating multifaceted influences of biological sex on cancer, providing both external and endogenous contributors to cancer development and describing sex-based differences for the broadest oncological classes. Given the increasing global research interest in sex-based differences, OncoSexome is expected to impact future precision oncology practices significantly.
Collapse
Affiliation(s)
- Xinyi Shen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Jiamin Li
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | | | - Yechi Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
- School of Public Health, Zhejiang University, Hangzhou 310058, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Sajid A Khan
- Yale School of Medicine, Yale University, New Haven 06510, USA
- Division of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven 06510, USA
| | - Hong Yan
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong 999077, China
| | - Caroline H Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
7
|
Li Y, Li F, Duan Z, Liu R, Jiao W, Wu H, Zhu F, Xue W. SYNBIP 2.0: epitopes mapping, sequence expansion and scaffolds discovery for synthetic binding protein innovation. Nucleic Acids Res 2025; 53:D595-D603. [PMID: 39413165 PMCID: PMC11701522 DOI: 10.1093/nar/gkae893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/18/2024] Open
Abstract
Synthetic binding proteins (SBPs) represent a pivotal class of artificially engineered proteins, meticulously crafted to exhibit targeted binding properties and specific functions. Here, the SYNBIP database, a comprehensive resource for SBPs, has been significantly updated. These enhancements include (i) featuring 3D structures of 899 SBP-target complexes to illustrate the binding epitopes of SBPs, (ii) using the structures of SBPs in the monomer or complex forms with target proteins, their sequence space has been expanded five times to 12 025 by integrating a structure-based protein generation framework and a protein property prediction tool, (iii) offering detailed information on 78 473 newly identified SBP-like scaffolds from the RCSB Protein Data Bank, and an additional 16 401 555 ones from the AlphaFold Protein Structure Database, and (iv) the database is regularly updated, incorporating 153 new SBPs. Furthermore, the structural models of all SBPs have been enhanced through the application of the AlphaFold2, with their clinical statuses concurrently refreshed. Additionally, the design methods employed for each SBP are now prominently featured in the database. In sum, SYNBIP 2.0 is designed to provide researchers with essential SBP data, facilitating their innovation in research, diagnosis and therapy. SYNBIP 2.0 is now freely accessible at https://idrblab.org/synbip/.
Collapse
Affiliation(s)
- Yanlin Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Fengcheng Li
- Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, Zhejiang 310052, China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Zixin Duan
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Ruihan Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Wantong Jiao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Haibo Wu
- School of Life Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Weiwei Xue
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, No. 55 South University Town Road, High-tech Zone, Chongqing 401331, China
| |
Collapse
|
8
|
Li F, Mou M, Li X, Xu W, Yin J, Zhang Y, Zhu F. DrugMAP 2.0: molecular atlas and pharma-information of all drugs. Nucleic Acids Res 2025; 53:D1372-D1382. [PMID: 39271119 PMCID: PMC11701670 DOI: 10.1093/nar/gkae791] [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: 08/03/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
The escalating costs and high failure rates have decelerated the pace of drug development, which amplifies the research interests in developing combinatorial/repurposed drugs and understanding off-target adverse drug reaction (ADR). In other words, it is demanded to delineate the molecular atlas and pharma-information for the combinatorial/repurposed drugs and off-target interactions. However, such invaluable data were inadequately covered by existing databases. In this study, a major update was thus conducted to the DrugMAP, which accumulated (a) 20831 combinatorial drugs and their interacting atlas involving 1583 pharmacologically important molecules; (b) 842 repurposed drugs and their interacting atlas with 795 molecules; (c) 3260 off-targets relevant to the ADRs of 2731 drugs and (d) various types of pharmaceutical information, including diverse ADMET properties, versatile diseases, and various ADRs/off-targets. With the growing demands for discovering combinatorial/repurposed therapies and the rapidly emerging interest in AI-based drug discovery, DrugMAP was highly expected to act as an indispensable supplement to existing databases facilitating drug discovery, which was accessible at: https://idrblab.org/drugmap/.
Collapse
Affiliation(s)
- Fengcheng Li
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
- State Key Lab of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Xiaoyi Li
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Weize Xu
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
| | - Jiayi Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yang Zhang
- School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Children's Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Zhejiang University, Hangzhou 310058, China
- State Key Lab of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
9
|
Fu S, Chen Z, Luo Z, Nie M, Fu T, Zhou Y, Yang Q, Zhu F, Ni F. Chem(Pro)2: the atlas of chemoproteomic probes labelling human proteins. Nucleic Acids Res 2025; 53:D1651-D1662. [PMID: 39436046 PMCID: PMC11701659 DOI: 10.1093/nar/gkae943] [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: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Chemoproteomic probes (CPPs) have been widely considered as powerful molecular biological tools that enable the highly efficient discovery of both binding proteins and modes of action for the studied compounds. They have been successfully used to validate targets and identify binders. The design of CPP has been considered extremely challenging, which asks for the generalization using a large number of probe data. However, none of the existing databases gives such valuable data of CPPs. Herein, a database entitled 'Chem(Pro)2' was therefore developed to systematically describe the atlas of diverse types of CPPs labelling human protein in living cell/lysate. With the booming application of chemoproteomic technique and artificial intelligence in current chemical biology study, Chem(Pro)2 was expected to facilitate the AI-based learning of interacting pattern among molecules for discovering innovative targets and new drugs. Till now, Chem(Pro)2 has been open to all users without any login requirement at: https://idrblab.org/chemprosquare/.
Collapse
Affiliation(s)
- Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhiming Luo
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Meiyun Nie
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| |
Collapse
|
10
|
Menacer R, Bouchekioua S, Meliani S, Belattar N. New combined Inverse-QSAR and molecular docking method for scaffold-based drug discovery. Comput Biol Med 2024; 180:108992. [PMID: 39128176 DOI: 10.1016/j.compbiomed.2024.108992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/14/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Computer-aided drug discovery plays a vital role in developing novel medications for various diseases. The COVID-19 pandemic has heightened the need for innovative approaches to design lead compounds with the potential to become effective drugs. Specifically, designing promising inhibitors of the SARS-CoV-2 main protease (Mpro) is crucial, as it plays a key role in viral replication. Phytochemicals, primarily flavonoids and flavonols from medicinal plants, were screened. Fifty small molecules were selected for molecular docking analysis against SARS-CoV-2 Mpro (PDB ID: 6LU7). Binding energies and interactions were analyzed and compared to those of the anti-SARS-CoV-2 inhibitor Nirmatrelvir. Using these 50 structures as a training set, a QSAR model was built employing simple, reversible topological descriptors. An inverse-QSAR analysis was then performed on 2⁹ = 512 hydroxyl combinations at nine possible positions on the flavone and flavonol scaffold. The model predicted three novel, promising compounds exhibiting the most favorable binding energies (-8.5 kcal/mol) among the 512 possible hydroxyl combinations: 3,6,7,2',4'-pentahydroxyflavone (PF9), 6,7,2',4'-tetrahydroxyflavone (PF11), and 3,6,7,4'-tetrahydroxyflavone (PF15). Molecular dynamics (MD) simulations demonstrated the stability of the PF9/Mpro complex over 300 ns of simulation. These predicted structures, reported here for the first time, warrant synthesis and further evaluation of their biological activity through in vitro and in vivo studies.
Collapse
Affiliation(s)
- Rafik Menacer
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria; Centre de Recherche Scientifique et Technique en Analyses Physico-Chimiques CRAPC, BP 384, Zone Industrielle, Bou-ismail, Tipaza, RP, 42004, Algeria.
| | - Saad Bouchekioua
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| | - Saida Meliani
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| | - Nadjah Belattar
- Centre de Recherche en Sciences Pharmaceutiques, Constantine, 25000, Algeria
| |
Collapse
|
11
|
Zhang Z, Li F, Duan Z, Shi C, Wang X, Zhu F, Xue W. OPTICS: An interactive online platform for photosensory and bio-functional proteins in optogenetic systems. Comput Biol Med 2024; 178:108687. [PMID: 38870722 DOI: 10.1016/j.compbiomed.2024.108687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/25/2024] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
High-precise modulation of bio-functional proteins related to signaling is crucial in life sciences and human health. The cutting-edge technology of optogenetics, which combines optical method with genetically encoded protein expression, pioneered new pathways for the control of cellular bio-functional proteins (CPs) using optogenetic tools (OTs) in spatial and temporal. Over the past decade, hundreds of optogenetic systems (OSs) have been developed for various applications from living cells to freely moving organisms. However, no database has been constructed to comprehensively provide the valuable information of OSs yet. In this work, a new database named OPTICS (an interactive online platform for photosensory and bio-functional proteins in optogenetic systems) is introduced. Our OPTICS is unique in (i) systematically describing diverse OSs from the perspective of photoreceptor-based classification and mechanism of action, (ii) featuring the detailed biophysical properties and functional data of OSs, (iii) providing the interaction between OT and CP for each OS referring to distinct applications in research, diagnosis, and therapy, and (iv) enabling a light response property-based search against all OSs in the database. Since the information on OSs is essential for rapid and predictable design of optogenetic controls, the comprehensive data provided in OPTICS lay a solid foundation for the future development of novel OSs. OPTICS is freely accessible without login requirement at https://idrblab.org/optics/.
Collapse
Affiliation(s)
- Zhao Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zixin Duan
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, China
| | - Chaoqun Shi
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, China
| | - Xiaona Wang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
12
|
Zhou S, Zhang H, Li J, Li W, Su M, Ren Y, Ge F, Zhang H, Shang H. Potential anti-liver cancer targets and mechanisms of kaempferitrin based on network pharmacology, molecular docking and experimental verification. Comput Biol Med 2024; 178:108693. [PMID: 38850960 DOI: 10.1016/j.compbiomed.2024.108693] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 06/10/2024]
Abstract
AIM Kaempferitrin is an active component in Chenopodium ambrosioides, showing medicinal functions against liver cancer. This study aimed to identify the potential targets and pathways of kaempferitrin against liver cancer using network pharmacology and molecular docking, and verify the essential hub targets and pathway in mice model of SMMC-7721 cells xenografted tumors and SMMC-7721 cells. METHODS Kaempferitrin therapeutical targets were obtained by searching SwissTargetPrediction, PharmMapper, STITCH, DrugBank, and TTD databases. Liver cancer specific genes were obtained by searching GeneCards, DrugBank, TTD, OMIM, and DisGeNET databases. PPI network of "kaempferitrin-targets-liver cancer" was constructed to screen the hub targets. GO, KEGG pathway and MCODE clustering analyses were performed to identify possible enrichment of genes with specific biological subjects. Molecular docking and molecular dynamics simulation were employed to determine the docking pose, potential and stability of kaempferitrin with hub targets. The potential anti-liver cancer mechanisms of kaempferitrin, as predicted by network pharmacology analyses, were verified by in vitro and in vivo experiments. RESULTS 228 kaempferitrin targets and 2186 liver cancer specific targets were identified, of which 50 targets were overlapped. 8 hub targets were identified through network topology analysis, and only SIRT1 and TP53 had a potent binding activity with kaempferitrin as indicated by molecular docking and molecular dynamics simulation. MCODE clustering analysis revealed the most significant functional module of PPI network including SIRT1 and TP53 was mainly related to cell apoptosis. GO and KEGG enrichment analyses suggested that kaempferitrin exerted therapeutic effects on liver cancer possibly by promoting apoptosis via p21/Bcl-2/Caspase 3 signaling pathway, which were confirmed by in vivo and in vitro experiments, such as HE staining of tumor tissues, CCK-8, qRT-PCR and Western blot. CONCLUSION This study provided not only insight into how kaempferitrin could act against liver cancer by identifying hub targets and their associated signaling pathways, but also experimental evidence for the clinical use of kaempferitrin in liver cancer treatment.
Collapse
Affiliation(s)
- Siyu Zhou
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Huidong Zhang
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Jiao Li
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China.
| | - Wei Li
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Min Su
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Yao Ren
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Fanglan Ge
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Hong Zhang
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| | - Hongli Shang
- College of Life Science, Sichuan Normal University, Chengdu, 610101, China
| |
Collapse
|
13
|
El-Assaad AM, Hamieh T. SARS-CoV-2: Prediction of critical ionic amino acid mutations. Comput Biol Med 2024; 178:108688. [PMID: 38870723 DOI: 10.1016/j.compbiomed.2024.108688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that caused coronavirus disease 2019 (COVID-19), has been studied thoroughly, and several variants are revealed across the world with their corresponding mutations. Studies and vaccines development focus on the genetic mutations of the S protein due to its vital role in allowing the virus attach and fuse with the membrane of a host cell. In this perspective, we study the effects of all ionic amino acid mutations of the SARS-CoV-2 viral spike protein S1 when bound to Antibody CC12.1 within the SARS-CoV-2:CC12.1 complex model. Binding free energy calculations between SARS-CoV-2 and antibody CC12.1 are based on the Analysis of Electrostatic Similarities of Proteins (AESOP) framework, where the electrostatic potentials are calculated using Adaptive Poisson-Boltzmann Solver (APBS). The atomic radii and charges that feed into the APBS calculations are calculated using the PDB2PQR software. Our results are the first to propose in silico potential life-threatening mutations of SARS-CoV-2 beyond the present mutations found in the five common variants worldwide. We find each of the following mutations: K378A, R408A, K424A, R454A, R457A, K458A, and K462A, to play significant roles in the binding to Antibody CC12.1, since they are turned into strong inhibitors on both chains of the S1 protein, whereas the mutations D405A, D420A, and D427A, show to play important roles in this binding, as they are turned into mild inhibitors on both chains of the S1 protein.
Collapse
Affiliation(s)
- Atlal M El-Assaad
- Department of Electrical Engineering & Computer Science, University of Toledo (UT), Toledo OH 43606, USA; Department of Computer Science, Lebanese International University (LIU), Bekaa, Lebanon.
| | - Tayssir Hamieh
- Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Laboratory of Materials, Catalysis, Environment and Analytical Methods (MCEMA), Faculty of Sciences, Lebanese University, Hadath, Lebanon.
| |
Collapse
|
14
|
Ranteh O, Tedasen A, Rahman MA, Ibrahim MA, Sama-Ae I. Bioactive compounds from Ocimum tenuiflorum and Poria cocos: A novel natural Compound for insomnia treatment based on A computational approach. Comput Biol Med 2024; 175:108491. [PMID: 38657467 DOI: 10.1016/j.compbiomed.2024.108491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Insomnia, a widespread public health issue, is associated with substantial distress and daytime functionality impairments and can predispose to depression and cardiovascular disease. Cognitive Behavioral Anti-insomnia therapies including benzodiazepines often face limitations due to patient adherence or potential adverse effects. This study focused on identifying novel bioactive compounds from medicinal plants, aiming to discover and develop new therapeutic agents with low risk-to-benefit ratios using computational drug discovery methods. Through a systematic framework involving compound library preparation, evaluation of drug-likeness and pharmacokinetics, toxicity prediction, molecular docking, and molecular dynamic simulations, two natural compounds such as 2-(4-hydroxy-3-methoxyphenyl)-8-methoxy-6-prop-2-enyl-3,4-dihydro-2H-chromen-3-ol from Ocimum tenuiflorum and 7-(2-hydroxypropan-2-yl)-1,4a-dimethyl-9-oxo-3,4,10,10a-tetrahydro-2H-phenanthrene-1-carboxylic acid from Poria cocos exhibited high binding affinity with orexin receptor type 1 (OX1R) and type 2 (OX2R), surpassing commercial drugs used in insomnia treatment. Additionally, they showed interactions with critical amino acid residues within the receptors that play crucial roles in competitive inhibitor activity, like commercial drugs such as Suvorexant, Lemborexant, and Daridorexant. Further, molecular dynamics simulations of the protein-ligand complexes under conditions that mimic the in vivo environment revealed both compounds' sustained and robust interactions with the OX1R and OX2R, reinforcing their potential as effective therapeutic candidates. Furthermore, upon evaluating both compounds' drug-likeness, pharmacokinetics, and toxicity profiles, it was discerned that they displayed considerable drug-like properties and favorable pharmacokinetics, along with diminished toxicity. The research provides a solid foundation for further exploring and validating these compounds as potential anti-insomnia therapeutics.
Collapse
Affiliation(s)
- Onggan Ranteh
- Department of Community Public Health, School of Public Health, Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand; Excellent Centre of Dengue and Community Public Health (EC for DACH), Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand.
| | - Aman Tedasen
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand; Research Excellence Center for Innovation and Health Products, Walailak University, Tha Sala, Nakhon Si Thammarat, 80161, Thailand.
| | - Md Atiar Rahman
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand; Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong-4331, Bangladesh.
| | | | - Imran Sama-Ae
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand; Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Tha Sala District, Nakhon Si Thammarat, 80160, Thailand.
| |
Collapse
|
15
|
da Silva LSA, Seman LO, Camponogara E, Mariani VC, Dos Santos Coelho L. Bilinear optimization of protein structure prediction: An exact approach via AB off-lattice model. Comput Biol Med 2024; 176:108558. [PMID: 38754216 DOI: 10.1016/j.compbiomed.2024.108558] [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: 02/27/2024] [Revised: 04/25/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
Protein structure prediction (PSP) remains a central challenge in computational biology due to its inherent complexity and high dimensionality. While numerous heuristic approaches have appeared in the literature, their success varies. The AB off-lattice model, which characterizes proteins as sequences of A (hydrophobic) and B (hydrophilic) beads, presents a simplified perspective on PSP. This work presents a mathematical optimization-based methodology capitalizing on the off-lattice AB model. Dissecting the inherent non-linearities of the energy landscape of protein folding allowed for formulating the PSP as a bilinear optimization problem. This formulation was achieved by introducing auxiliary variables and constraints that encapsulate the nuanced relationship between the protein's conformational space and its energy landscape. The proposed bilinear model exhibited notable accuracy in pinpointing the global minimum energy conformations on a benchmark dataset presented by the Protein Data Bank (PDB). Compared to traditional heuristic-based methods, this bilinear approach yielded exact solutions, reducing the likelihood of local minima entrapment. This research highlights the potential of reframing the traditionally non-linear protein structure prediction problem into a bilinear optimization problem through the off-lattice AB model. Such a transformation offers a route toward methodologies that can determine the global solution, challenging current PSP paradigms. Exploration into hybrid models, merging bilinear optimization and heuristic components, might present an avenue for balancing accuracy with computational efficiency.
Collapse
Affiliation(s)
- Luiza Scapinello Aquino da Silva
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil.
| | - Laio Oriel Seman
- Department of Automation and Systems Engineering, Federal University of Santa Catarina (UFSC), Engenheiro Agronômico Andrei Cristian Ferreira, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Eduardo Camponogara
- Department of Automation and Systems Engineering, Federal University of Santa Catarina (UFSC), Engenheiro Agronômico Andrei Cristian Ferreira, Florianópolis, 88040-900, Santa Catarina, Brazil
| | - Viviana Cocco Mariani
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil; Mechanical Engineering Graduate Program (PGMec), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil
| | - Leandro Dos Santos Coelho
- Electrical Engineering Graduate Program (PPGEE), Federal University of Parana (UFPR), Coronel Francisco Heraclito dos Santos, Curitiba, 81530-000, Paraná, Brazil
| |
Collapse
|
16
|
Zhou Y, Chen Z, Yang M, Chen F, Yin J, Zhang Y, Zhou X, Sun X, Ni Z, Chen L, Lv Q, Zhu F, Liu S. FERREG: ferroptosis-based regulation of disease occurrence, progression and therapeutic response. Brief Bioinform 2024; 25:bbae223. [PMID: 38742521 PMCID: PMC11091744 DOI: 10.1093/bib/bbae223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 04/21/2024] [Indexed: 05/16/2024] Open
Abstract
Ferroptosis is a non-apoptotic, iron-dependent regulatory form of cell death characterized by the accumulation of intracellular reactive oxygen species. In recent years, a large and growing body of literature has investigated ferroptosis. Since ferroptosis is associated with various physiological activities and regulated by a variety of cellular metabolism and mitochondrial activity, ferroptosis has been closely related to the occurrence and development of many diseases, including cancer, aging, neurodegenerative diseases, ischemia-reperfusion injury and other pathological cell death. The regulation of ferroptosis mainly focuses on three pathways: system Xc-/GPX4 axis, lipid peroxidation and iron metabolism. The genes involved in these processes were divided into driver, suppressor and marker. Importantly, small molecules or drugs that mediate the expression of these genes are often good treatments in the clinic. Herein, a newly developed database, named 'FERREG', is documented to (i) providing the data of ferroptosis-related regulation of diseases occurrence, progression and drug response; (ii) explicitly describing the molecular mechanisms underlying each regulation; and (iii) fully referencing the collected data by cross-linking them to available databases. Collectively, FERREG contains 51 targets, 718 regulators, 445 ferroptosis-related drugs and 158 ferroptosis-related disease responses. FERREG can be accessed at https://idrblab.org/ferreg/.
Collapse
Affiliation(s)
- Yuan Zhou
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Mengjie Yang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Fengyun Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiayi Yin
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine
| | - Yintao Zhang
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xuheng Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xiuna Sun
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Lu Chen
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| | - Qun Lv
- Department of Respiratory, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, 311121, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, 330110, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, and Department of Respiratory Medicine of Affiliated Hospital, Hangzhou Normal University, Hangzhou, 311121, China
| |
Collapse
|
17
|
Gorostiola González M, Rakers PRJ, Jespers W, IJzerman AP, Heitman LH, van Westen GJP. Computational Characterization of Membrane Proteins as Anticancer Targets: Current Challenges and Opportunities. Int J Mol Sci 2024; 25:3698. [PMID: 38612509 PMCID: PMC11011372 DOI: 10.3390/ijms25073698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer remains a leading cause of mortality worldwide and calls for novel therapeutic targets. Membrane proteins are key players in various cancer types but present unique challenges compared to soluble proteins. The advent of computational drug discovery tools offers a promising approach to address these challenges, allowing for the prioritization of "wet-lab" experiments. In this review, we explore the applications of computational approaches in membrane protein oncological characterization, particularly focusing on three prominent membrane protein families: receptor tyrosine kinases (RTKs), G protein-coupled receptors (GPCRs), and solute carrier proteins (SLCs). We chose these families due to their varying levels of understanding and research data availability, which leads to distinct challenges and opportunities for computational analysis. We discuss the utilization of multi-omics data, machine learning, and structure-based methods to investigate aberrant protein functionalities associated with cancer progression within each family. Moreover, we highlight the importance of considering the broader cellular context and, in particular, cross-talk between proteins. Despite existing challenges, computational tools hold promise in dissecting membrane protein dysregulation in cancer. With advancing computational capabilities and data resources, these tools are poised to play a pivotal role in identifying and prioritizing membrane proteins as personalized anticancer targets.
Collapse
Affiliation(s)
- Marina Gorostiola González
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
- Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Pepijn R. J. Rakers
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
| | - Willem Jespers
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
| | - Adriaan P. IJzerman
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
| | - Laura H. Heitman
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
- Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Gerard J. P. van Westen
- Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands; (M.G.G.); (P.R.J.R.); (W.J.); (A.P.I.); (L.H.H.)
| |
Collapse
|