1
|
Carr KD, Zambrano DED, Weidle C, Goodson A, Eisenach HE, Pyles H, Courbet A, King NP, Borst AJ. Protein identification using Cryo-EM and artificial intelligence guides improved sample purification. J Struct Biol X 2025; 11:100120. [PMID: 39958810 PMCID: PMC11830286 DOI: 10.1016/j.yjsbx.2025.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
Protein purification is essential in protein biochemistry, structural biology, and protein design, enabling the determination of protein structures, the study of biological mechanisms, and the characterization of both natural and de novo designed proteins. However, standard purification strategies often encounter challenges, such as unintended co-purification of contaminants alongside the target protein. This issue is particularly problematic for self-assembling protein nanomaterials, where unexpected geometries may reflect novel assembly states, cross-contamination, or native proteins originating from the expression host. Here, we used an automated structure-to-sequence pipeline to first identify an unknown co-purifying protein found in several purified designed protein samples. By integrating cryo-electron microscopy (Cryo-EM), ModelAngelo's sequence-agnostic model-building, and Protein BLAST, we identified the contaminant as dihydrolipoamide succinyltransferase (DLST). This identification was validated through comparisons with DLST structures in the Protein Data Bank, AlphaFold 3 predictions based on the DLST sequence from our E. coli expression vector, and traditional biochemical methods. The identification informed subsequent modifications to our purification protocol, which successfully excluded DLST from future preparations. To explore the potential broader utility of this approach, we benchmarked four computational methods for DLST identification across varying resolution ranges. This study demonstrates the successful application of a structure-to-sequence protein identification workflow, integrating Cryo-EM, ModelAngelo, Protein BLAST, and AlphaFold 3 predictions, to identify and ultimately help guide the removal of DLST from sample purification efforts. It highlights the potential of combining Cryo-EM with AI-driven tools for accurate protein identification and addressing purification challenges across diverse contexts in protein science.
Collapse
Affiliation(s)
- Kenneth D. Carr
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Dane Evan D. Zambrano
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Connor Weidle
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alex Goodson
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Helen E. Eisenach
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Harley Pyles
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alexis Courbet
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Andrew J. Borst
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
Wang Y, Song H, Teng Y, Huang G, Qian J, Wang H, Dong S, Ha J, Ma Y, Chang M, Jeong SD, Deng W, Schrank BR, Grippin A, Wu A, Edwards JL, Zhang Y, Lin Y, Poon W, Wilhelm S, Bi Y, Teng L, Wang Z, Kim BYS, Jiang W. A Generative Artificial Intelligence Copilot for Biomedical Nanoengineering. ACS NANO 2025. [PMID: 40367350 DOI: 10.1021/acsnano.5c03454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
The recent success of large language models (LLMs) in performing natural language processing tasks has increased interest in applying generative artificial intelligence (AI) to scientific research. However, a common problem of LLMs is their tendency to produce inaccurate and sometimes "hallucinated" outputs. Here, we established a generative AI tool, NanoSafari, to automatically extract knowledge from the biomedical nanoscience literature and address scientific queries. We developed the Grouped Iterative Validation based Information Extraction (GIVE) method to extract contextual information on nanoparticle characteristics from >20,000 published articles and established a database that was incorporated into the generative LLM to provide accurate nanomaterial design parameters. Blinded evaluation by biomedical nanoscientists showed that NanoSafari outperformed the baseline model in providing more reliable parameters for nanomaterial design tasks, as further validated by bench experiments. Together, these findings demonstrate the utility of AI-based methods for automated learning from "real-world" published work to provide accurate and reliable scientific references for biomaterial and bioengineering applications.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Haitao Song
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Yue Teng
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Guan Huang
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Jingzhe Qian
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Hongyu Wang
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - JongHoon Ha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Mengyu Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Seong Dong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Weiye Deng
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Benjamin R Schrank
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Adam Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Annette Wu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Jared L Edwards
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Yixiang Zhang
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
| | - Yuanyuan Lin
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Wilson Poon
- Department of Metallurgical, Materials and Biomedical Engineering, The University of Texas El Paso, El Paso, Texas 79968, United States
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
- Institute for Biomedical Engineering, Science, and Technology (IBEST), Norman, Oklahoma 73019, United States
- Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States
- Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma 73104, United States
| | - Ye Bi
- The Public Experimental Center, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Lesheng Teng
- School of Life Science, Jilin University, Changchun 130012, China
| | - Zikai Wang
- SigmaSafari Project, Shanghai Artificial Intelligence Research Institute Co., Ltd., Shanghai 201109, China
- Xiangfu Laboratory, Jiashan 314100, China
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
3
|
Li Y, Zhang Y. Flexing protein assemblies. Nat Struct Mol Biol 2025:10.1038/s41594-025-01560-2. [PMID: 40355748 DOI: 10.1038/s41594-025-01560-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Affiliation(s)
- Yang Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yang Zhang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Department of Computer Science, School of Computing, National University of Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
Lin M, Guo J, Gu Z, Tang W, Tao H, You S, Jia D, Sun Y, Jia P. Machine learning and multi-omics integration: advancing cardiovascular translational research and clinical practice. J Transl Med 2025; 23:388. [PMID: 40176068 PMCID: PMC11966820 DOI: 10.1186/s12967-025-06425-2] [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: 08/06/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
The global burden of cardiovascular diseases continues to rise, making their prevention, diagnosis and treatment increasingly critical. With advancements and breakthroughs in omics technologies such as high-throughput sequencing, multi-omics approaches can offer a closer reflection of the complex physiological and pathological changes in the body from a molecular perspective, providing new microscopic insights into cardiovascular diseases research. However, due to the vast volume and complexity of data, accurately describing, utilising, and translating these biomedical data demands substantial effort. Researchers and clinicians are actively developing artificial intelligence (AI) methods for data-driven knowledge discovery and causal inference using various omics data. These AI approaches, integrated with multi-omics research, have shown promising outcomes in cardiovascular studies. In this review, we outline the methods for integrating machine learning, one of the most successful applications of AI, with omics data and summarise representative AI models developed that leverage various omics data to facilitate the exploration of cardiovascular diseases from underlying mechanisms to clinical practice. Particular emphasis is placed on the effectiveness of using AI to extract potential molecular information to address current knowledge gaps. We discuss the challenges and opportunities of integrating omics with AI into routine diagnostic and therapeutic practices and anticipate the future development of novel AI models for wider application in the field of cardiovascular diseases.
Collapse
Affiliation(s)
- Mingzhi Lin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhilin Gu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Wenyi Tang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hongqian Tao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Shilong You
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
5
|
Li S, Liu Y, Yang B, Lv S, Zou X, Tang L, Zhou L, Jiang Z, Liu Z. An Improved Monte Carlo Method for Quantitative Analysis of Transparency Degradation Caused by Corneal Edema. Transl Vis Sci Technol 2025; 14:15. [PMID: 40208891 PMCID: PMC11993133 DOI: 10.1167/tvst.14.4.15] [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: 10/02/2024] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
Purpose To address the impact of corneal edema on transparency, we proposed an improved Monte Carlo (MC) method that uses a cost-effective and easily applicable approach to extract fibril features. This method aims to enhance computational accuracy and elucidate microscopic mechanisms of corneal transparency. Methods Fresh ex vivo porcine corneas were immersed in deionized water to simulate various states of corneal edema. For each edema state, we carefully analyzed its transparency and fibrous conditions. The diameter and spatial distribution of corneal fibrils were measured using image recognition algorithms. An improved MC method, analyzing the multiple scattering of each incident light, was proposed to simulate the propagation of incident light in the cornea. Results Higher edema reduces transparency. After 10 hours of hydration, corneal water content increased by 7% and transparency decreased by over 30%. Cross-sectional observations indicated that the radius increases from the original 15.1 nm to 16.8 nm, the fibril-to-base material area ratio decreases from the initial 15% to roughly 7%, and the minimum gap between fibrils expanded by 35%. The proposed MC method provides more accurate transparency estimates compared to the direct summation of field theory. Conclusions Corneal transparency decreases as a result of multiple factors, including fibril and cornea thickening, variations in the refractive index, and changes in the fibril distribution. Translational Relevance The MC method is cost-effective, easily applicable, and accurate in predicting the transparency of corneas with varying water contents. Moreover, this method intuitively demonstrates the scattering of light during the propagation process.
Collapse
Affiliation(s)
- Shuaiyi Li
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Yiping Liu
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Bao Yang
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Sulei Lv
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Xiulan Zou
- General Hospital of Southern Theater Command of the People's Liberation Army of China, Guangzhou, China
| | - Liqun Tang
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Licheng Zhou
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Zhenyu Jiang
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| | - Zejia Liu
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, China
| |
Collapse
|
6
|
Wang Y, Han S, Wang Y, Liang Q, Luo W. Artificial Intelligence Technology Assists Enzyme Prediction and Rational Design. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7065-7073. [PMID: 40066931 DOI: 10.1021/acs.jafc.4c13201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Since the structure of enzymes determines their function, elucidating the structure of enzymes lays a solid foundation for deciphering their catalytic mechanism and enabling rational design. The development of artificial intelligence (AI) has sparked a technological revolution, infusing new vitality into theoretical studies of enzymology and the advancement of enzyme engineering techniques. This Review outlines the development process and main methods of AI applied in the structural elucidation and functional prediction of enzymes. Furthermore, it emphasizes AI-based rational design of enzymes and provides a detailed exposition of representative AI algorithms and case studies. With the support of AI technology, the comprehension of enzyme structure and function and their relationship will become deeper and more efficient, thereby promoting the widespread application of enzyme engineering in various fields.
Collapse
Affiliation(s)
- Yuhang Wang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Shuangxin Han
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| | - Yi Wang
- Department of Biological and Agricultural Engineering, University of California, Davis, 1 Shields Ave, Davis, California 95616, United States
| | - Quanfeng Liang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, P. R. China
| | - Wei Luo
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214126, China
| |
Collapse
|
7
|
He XH, Li JR, Xu J, Shan H, Shen SY, Gao SH, Xu HE. AI-driven antibody design with generative diffusion models: current insights and future directions. Acta Pharmacol Sin 2025; 46:565-574. [PMID: 39349764 PMCID: PMC11845702 DOI: 10.1038/s41401-024-01380-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/15/2024] [Indexed: 02/23/2025]
Abstract
Therapeutic antibodies are at the forefront of biotherapeutics, valued for their high target specificity and binding affinity. Despite their potential, optimizing antibodies for superior efficacy presents significant challenges in both monetary and time costs. Recent strides in computational and artificial intelligence (AI), especially generative diffusion models, have begun to address these challenges, offering novel approaches for antibody design. This review delves into specific diffusion-based generative methodologies tailored for antibody design tasks, de novo antibody design, and optimization of complementarity-determining region (CDR) loops, along with their evaluation metrics. We aim to provide an exhaustive overview of this burgeoning field, making it an essential resource for leveraging diffusion-based generative models in antibody design endeavors.
Collapse
Affiliation(s)
- Xin-Heng He
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun-Rui Li
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - James Xu
- Cascade Pharma, Shanghai, 201318, China
| | - Hong Shan
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shi-Yi Shen
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Si-Han Gao
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - H Eric Xu
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Cascade Pharma, Shanghai, 201318, China.
| |
Collapse
|
8
|
Khmelinskaia A, Bethel NP, Fatehi F, Mallik BB, Antanasijevic A, Borst AJ, Lai SH, Chim HY, Wang JY'J, Miranda MC, Watkins AM, Ogohara C, Caldwell S, Wu M, Heck AJR, Veesler D, Ward AB, Baker D, Twarock R, King NP. Local structural flexibility drives oligomorphism in computationally designed protein assemblies. Nat Struct Mol Biol 2025:10.1038/s41594-025-01490-z. [PMID: 40011747 DOI: 10.1038/s41594-025-01490-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/14/2025] [Indexed: 02/28/2025]
Abstract
Many naturally occurring protein assemblies have dynamic structures that allow them to perform specialized functions. Although computational methods for designing novel self-assembling proteins have advanced substantially over the past decade, they primarily focus on designing static structures. Here we characterize three distinct computationally designed protein assemblies that exhibit unanticipated structural diversity arising from flexibility in their subunits. Cryo-EM single-particle reconstructions and native mass spectrometry reveal two distinct architectures for two assemblies, while six cryo-EM reconstructions for the third likely represent a subset of its solution-phase structures. Structural modeling and molecular dynamics simulations indicate that constrained flexibility within the subunits of each assembly promotes a defined range of architectures rather than nonspecific aggregation. Redesigning the flexible region in one building block rescues the intended monomorphic assembly. These findings highlight structural flexibility as a powerful design principle, enabling exploration of new structural and functional spaces in protein assembly design.
Collapse
Affiliation(s)
- Alena Khmelinskaia
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Transdisciplinary Research Areas 'Building Blocks of Matter and Fundamental Interactions', University of Bonn, Bonn, Germany.
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
- Department of Chemistry, Ludwig Maximilian University of Munich, Munich, Germany.
| | - Neville P Bethel
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Farzad Fatehi
- Department of Mathematics, University of York, York, UK
- York Cross-Disciplinary Center for Systems Analysis, University of York, York, UK
| | - Bhoomika Basu Mallik
- Transdisciplinary Research Areas 'Building Blocks of Matter and Fundamental Interactions', University of Bonn, Bonn, Germany
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Department of Chemistry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aleksandar Antanasijevic
- Department of Integrative, Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Szu-Hsueh Lai
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | - Ho Yeung Chim
- Department of Chemistry, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jing Yang 'John' Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Marcos C Miranda
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Cassandra Ogohara
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Shane Caldwell
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mengyu Wu
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Albert J R Heck
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Andrew B Ward
- Scripps Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Reidun Twarock
- Department of Mathematics, University of York, York, UK
- York Cross-Disciplinary Center for Systems Analysis, University of York, York, UK
- Department of Biology, University of York, York, UK
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
9
|
Kim J, Kim JY, Kim J, Hyeong Y, Neseli B, You JB, Shim J, Shin J, Park HH, Kurt H. Inverse design of nanophotonic devices enabled by optimization algorithms and deep learning: recent achievements and future prospects. NANOPHOTONICS (BERLIN, GERMANY) 2025; 14:121-151. [PMID: 39927200 PMCID: PMC11806510 DOI: 10.1515/nanoph-2024-0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
Nanophotonics, which explores significant light-matter interactions at the nanoscale, has facilitated significant advancements across numerous research fields. A key objective in this area is the design of ultra-compact, high-performance nanophotonic devices to pave the way for next-generation photonics. While conventional brute-force, intuition-based forward design methods have produced successful nanophotonic solutions over the past several decades, recent developments in optimization methods and artificial intelligence offer new potential to expand these capabilities. In this review, we delve into the latest progress in the inverse design of nanophotonic devices, where AI and optimization methods are leveraged to automate and enhance the design process. We discuss representative methods commonly employed in nanophotonic design, including various meta-heuristic algorithms such as trajectory-based, evolutionary, and swarm-based approaches, in addition to adjoint-based optimization. Furthermore, we explore state-of-the-art deep learning techniques, involving discriminative models, generative models, and reinforcement learning. We also introduce and categorize several notable inverse-designed nanophotonic devices and their respective design methodologies. Additionally, we summarize the open-source inverse design tools and commercial foundries. Finally, we provide our perspectives on the current challenges of inverse design, while offering insights into future directions that could further advance this rapidly evolving field.
Collapse
Affiliation(s)
- Junhyeong Kim
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jae-Yong Kim
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jungmin Kim
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI53706, USA
| | - Yun Hyeong
- The School of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Berkay Neseli
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jong-Bum You
- National Nanofab Center (NNFC), Daejeon, Republic of Korea
| | - Joonsup Shim
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Jonghwa Shin
- The School of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyo-Hoon Park
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hamza Kurt
- The School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| |
Collapse
|
10
|
Flood R, Thureau A, Crowley PB. A Macrocycle-Mediated Protein Cage. ACS Macro Lett 2024; 13:1686-1690. [PMID: 39592260 PMCID: PMC11656731 DOI: 10.1021/acsmacrolett.4c00656] [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: 09/26/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
Engineered protein cages are of great interest considering their diverse applications in delivery and catalysis. Here, we describe macrocycle-triggered icosahedral cage assembly of a designed β-propeller. Cage assembly was evidenced by small-angle X-ray scattering and X-ray crystallography.
Collapse
Affiliation(s)
- Ronan
J. Flood
- SSPC,
Science Foundation Ireland Research Centre for Pharmaceuticals, School
of Biological and Chemical Sciences, University
of Galway, University
Road, Galway H91 TK33, Ireland
| | - Aurélien Thureau
- Synchrotron
SOLEIL, L’Orme des Merisiers, Saint-Aubin BP 48, 91192 Cedex Gif-sur-Yvette, France
| | - Peter B. Crowley
- SSPC,
Science Foundation Ireland Research Centre for Pharmaceuticals, School
of Biological and Chemical Sciences, University
of Galway, University
Road, Galway H91 TK33, Ireland
| |
Collapse
|
11
|
Kuang Z, Yan X, Yuan Y, Wang R, Zhu H, Wang Y, Li J, Ye J, Yue H, Yang X. Advances in stress-tolerance elements for microbial cell factories. Synth Syst Biotechnol 2024; 9:793-808. [PMID: 39072145 PMCID: PMC11277822 DOI: 10.1016/j.synbio.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Microorganisms, particularly extremophiles, have evolved multiple adaptation mechanisms to address diverse stress conditions during survival in unique environments. Their responses to environmental coercion decide not only survival in severe conditions but are also an essential factor determining bioproduction performance. The design of robust cell factories should take the balance of their growing and bioproduction into account. Thus, mining and redesigning stress-tolerance elements to optimize the performance of cell factories under various extreme conditions is necessary. Here, we reviewed several stress-tolerance elements, including acid-tolerant elements, saline-alkali-resistant elements, thermotolerant elements, antioxidant elements, and so on, providing potential materials for the construction of cell factories and the development of synthetic biology. Strategies for mining and redesigning stress-tolerance elements were also discussed. Moreover, several applications of stress-tolerance elements were provided, and perspectives and discussions for potential strategies for screening stress-tolerance elements were made.
Collapse
Affiliation(s)
- Zheyi Kuang
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830017, China
| | - Xiaofang Yan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yanfei Yuan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Ruiqi Wang
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830017, China
| | - Haifan Zhu
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830017, China
| | - Youyang Wang
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830017, China
| | - Jianfeng Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Jianwen Ye
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Haitao Yue
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830017, China
- Laboratory of Synthetic Biology, School of Life Science and Technology, Xinjiang University, Urumqi, 830017, China
| | - Xiaofeng Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
12
|
Le Y, Zhang M, Wu P, Wang H, Ni J. Biofuel production from lignocellulose via thermophile-based consolidated bioprocessing. ENGINEERING MICROBIOLOGY 2024; 4:100174. [PMID: 39628591 PMCID: PMC11610967 DOI: 10.1016/j.engmic.2024.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 12/06/2024]
Abstract
The depletion of fossil fuels and their impact on the environment have led to efforts to develop alternative sustainable fuels. While biofuel derived from lignocellulose is considered a sustainable, renewable, and green energy source, enhancing biofuel production and achieving a cost-effective bioconversion of lignocellulose at existing bio-refineries remains a challenge. Consolidated bioprocessing (CBP) using thermophiles can simplify this operation by integrating multiple processes, such as hydrolytic enzyme production, lignocellulose degradation, biofuel fermentation, and product distillation. This paper reviews recent developments in the conversion of lignocellulose to biofuel using thermophile-based CBP. First, advances in thermostable enzyme and thermophilic lignocellulolytic microorganism discovery and development for lignocellulosic biorefinery use are outlined. Then, several thermophilic CBP candidates and thermophilic microbes engineered to drive CBP of lignocellulose are reviewed. CRISPR/Cas-based genome editing tools developed for thermophiles are also highlighted. The potential applications of the Design-Build-Test-Learn (DBTL) synthetic biology strategy for designing and constructing thermophilic CBP hosts are also discussed in detail. Overall, this review illustrates how to develop highly sophisticated thermophilic CBP hosts for use in lignocellulosic biorefinery applications.
Collapse
Affiliation(s)
- Yilin Le
- Biofuels Institute, School of Emergency Management, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Mengqi Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Pengju Wu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| | - Huilei Wang
- Biofuels Institute, School of Emergency Management, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Jinfeng Ni
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, PR China
| |
Collapse
|
13
|
Luu MT, Berengut JF, Li J, Chen JB, Daljit Singh JK, Coffi Dit Glieze K, Turner M, Skipper K, Meppat S, Fowler H, Close W, Doye JPK, Abbas A, Wickham SFJ. Reconfigurable nanomaterials folded from multicomponent chains of DNA origami voxels. Sci Robot 2024; 9:eadp2309. [PMID: 39602517 DOI: 10.1126/scirobotics.adp2309] [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: 03/14/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
In cells, proteins rapidly self-assemble into sophisticated nanomachines. Bioinspired self-assembly approaches, such as DNA origami, have been used to achieve complex three-dimensional (3D) nanostructures and devices. However, current synthetic systems are limited by low yields in hierarchical assembly and challenges in rapid and efficient reconfiguration between diverse structures. Here, we developed a modular system of DNA origami "voxels" with programmable 3D connections. We demonstrate multifunctional pools of up to 12 unique voxels that can assemble into many shapes, prototyping 50 structures. Programmable switching of local connections between flexible and rigid states achieved rapid and reversible reconfiguration of global structures in three dimensions. Multistep assembly pathways were then explored to increase the yield. Voxels were assembled via flexible chain intermediates into rigid structures, increasing yield up to 100-fold. We envision that foldable chains of DNA origami voxels can achieve increased complexity in reconfigurable nanomaterials, providing modular components for the assembly of nanorobotic systems with future applications in synthetic biology, assembly of inorganic materials, and nanomedicine.
Collapse
Affiliation(s)
- Minh Tri Luu
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia
- School of Chemical and Biomolecular Engineering, University of Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan F Berengut
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
- EMBL Australia Node for Single Molecule Science, School of Biomedical Sciences, University of New South Wales, Sydney 2052, Australia
- ARC Centre of Excellence in Synthetic Biology, University of New South Wales, Sydney, Australia
| | - Jiahe Li
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
| | - Jing-Bing Chen
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
| | - Jasleen Kaur Daljit Singh
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- School of Chemical and Biomolecular Engineering, University of Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Kanako Coffi Dit Glieze
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Matthew Turner
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
| | - Karuna Skipper
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Sreelakshmi Meppat
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Hannah Fowler
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - William Close
- Australian Centre for Microscopy & Microanalysis, University of Sydney, Sydney, NSW 2006, Australia
| | - Jonathan P K Doye
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
| | - Ali Abbas
- School of Chemical and Biomolecular Engineering, University of Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Shelley F J Wickham
- School of Chemistry, University of Sydney, Sydney, NSW 2006, Australia
- School of Physics, University of Sydney, Sydney, NSW 2006, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Zhu C, Mu J, Liang L. Nanocarriers for intracellular delivery of proteins in biomedical applications: strategies and recent advances. J Nanobiotechnology 2024; 22:688. [PMID: 39523313 PMCID: PMC11552240 DOI: 10.1186/s12951-024-02969-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Protein drugs are of great importance in maintaining the normal functioning of living organisms. Indeed, they have been instrumental in combating tumors and genetic diseases for decades. Among these pharmaceutical agents, those that target intracellular components necessitate the use of therapeutic proteins to exert their effects within the targeted cells. However, the use of protein drugs is limited by their short half-life and potential adverse effects in the physiological environment. The advent of nanoparticles offers a promising avenue for prolonging the half-life of protein drugs. This is achieved by encapsulating proteins, thereby safeguarding their biological activity and ensuring precise delivery into cells. This nanomaterial-based intracellular protein drug delivery system mitigates the rapid hydrolysis and unwarranted diffusion of proteins, thereby minimizing potential side effects and circumventing the limitations inherent in traditional techniques like electroporation. This review examines established protein drug delivery systems, including those based on polymers, liposomes, and protein nanoparticles. We delve into the operational principles and transport mechanisms of nanocarriers, discussing the various considerations essential for designing cutting-edge delivery platforms. Additionally, we investigate innovative designs and applications of traditional cytosolic protein delivery systems in medical research and clinical practice, particularly in areas like tumor treatment, gene editing and fluorescence imaging. This review sheds light on the current restrictions of protein delivery systems and anticipates future research avenues, aiming to foster the continued advancement in this field.
Collapse
Affiliation(s)
- Chuanda Zhu
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P.R. China.
| | - Ling Liang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China.
| |
Collapse
|
15
|
Zhang P, Wei L, Li J, Wang X. Artificial intelligence-guided strategies for next-generation biological sequence design. Natl Sci Rev 2024; 11:nwae343. [PMID: 39606146 PMCID: PMC11601974 DOI: 10.1093/nsr/nwae343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Pengcheng Zhang
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, China
| | - Lei Wei
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, China
| | - Jiaqi Li
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, China
| | - Xiaowo Wang
- Ministry of Education Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Department of Automation, Tsinghua University, China
| |
Collapse
|
16
|
Gao S, Fang A, Huang Y, Giunchiglia V, Noori A, Schwarz JR, Ektefaie Y, Kondic J, Zitnik M. Empowering biomedical discovery with AI agents. Cell 2024; 187:6125-6151. [PMID: 39486399 DOI: 10.1016/j.cell.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/16/2024] [Accepted: 09/12/2024] [Indexed: 11/04/2024]
Abstract
We envision "AI scientists" as systems capable of skeptical learning and reasoning that empower biomedical research through collaborative agents that integrate AI models and biomedical tools with experimental platforms. Rather than taking humans out of the discovery process, biomedical AI agents combine human creativity and expertise with AI's ability to analyze large datasets, navigate hypothesis spaces, and execute repetitive tasks. AI agents are poised to be proficient in various tasks, planning discovery workflows and performing self-assessment to identify and mitigate gaps in their knowledge. These agents use large language models and generative models to feature structured memory for continual learning and use machine learning tools to incorporate scientific knowledge, biological principles, and theories. AI agents can impact areas ranging from virtual cell simulation, programmable control of phenotypes, and the design of cellular circuits to developing new therapies.
Collapse
Affiliation(s)
- Shanghua Gao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Ada Fang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA; Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA
| | - Yepeng Huang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Valentina Giunchiglia
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Department of Brain Sciences, Imperial College London, London, UK
| | - Ayush Noori
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Harvard College, Cambridge, MA, USA
| | | | - Yasha Ektefaie
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jovana Kondic
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Allston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Data Science Initiative, Cambridge, MA, USA.
| |
Collapse
|
17
|
Han Z, Shen Z, Pei J, You Q, Zhang Q, Wang L. Transformation of peptides to small molecules in medicinal chemistry: Challenges and opportunities. Acta Pharm Sin B 2024; 14:4243-4265. [PMID: 39525591 PMCID: PMC11544290 DOI: 10.1016/j.apsb.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/14/2024] [Accepted: 06/11/2024] [Indexed: 11/16/2024] Open
Abstract
Peptides are native binders involved in numerous physiological life procedures, such as cellular signaling, and serve as ready-made regulators of biochemical processes. Meanwhile, small molecules compose many drugs owing to their outstanding advantages of physiochemical properties and synthetic convenience. A novel field of research is converting peptides into small molecules, providing a convenient portable solution for drug design or peptidomic research. Endowing properties of peptides onto small molecules can evolutionarily combine the advantages of both moieties and improve the biological druggability of molecules. Herein, we present eight representative recent cases in this conversion and elaborate on the transformation process of each case. We discuss the innovative technological methods and research approaches involved, and analyze the applicability conditions of the approaches and methods in each case, guiding further modifications of peptides to small molecules. Finally, based on the aforementioned cases, we summarize a general procedure for peptide-to-small molecule modifications, listing the technological methods available for each transformation step and providing our insights on the applicable scenarios for these methods. This review aims to present the progress of peptide-to-small molecule modifications and propose our thoughts and perspectives for future research in this field.
Collapse
Affiliation(s)
- Zeyu Han
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zekai Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiayue Pei
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiuyue Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
18
|
Ouyang B, Shan C, Shen S, Dai X, Chen Q, Su X, Cao Y, Qin X, He Y, Wang S, Xu R, Hu R, Shi L, Lu T, Yang W, Peng S, Zhang J, Wang J, Li D, Pang Z. AI-powered omics-based drug pair discovery for pyroptosis therapy targeting triple-negative breast cancer. Nat Commun 2024; 15:7560. [PMID: 39215014 PMCID: PMC11364624 DOI: 10.1038/s41467-024-51980-9] [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: 08/27/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Due to low success rates and long cycles of traditional drug development, the clinical tendency is to apply omics techniques to reveal patient-level disease characteristics and individualized responses to treatment. However, the heterogeneous form of data and uneven distribution of targets make drug discovery and precision medicine a non-trivial task. This study takes pyroptosis therapy for triple-negative breast cancer (TNBC) as a paradigm and uses data mining of a large TNBC cohort and drug databases to establish a biofactor-regulated neural network for rapidly screening and optimizing compound pyroptosis drug pairs. Subsequently, biomimetic nanococrystals are prepared using the preferred combination of mitoxantrone and gambogic acid for rational drug delivery. The unique mechanism of obtained nanococrystals regulating pyroptosis genes through ribosomal stress and triggering pyroptosis cascade immune effects are revealed in TNBC models. In this work, a target omics-based intelligent compound drug discovery framework explores an innovative drug development paradigm, which repurposes existing drugs and enables precise treatment of refractory diseases.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, P. R. China
| | - Caihua Shan
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Shun Shen
- Pharmacy Department & Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, P. R. China
| | - Xinnan Dai
- Microsoft Research Asia, Shanghai, 200232, P. R. China
| | - Qingwang Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xiaomin Su
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Yongbin Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Xifeng Qin
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ying He
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Siyu Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruizhe Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Shanghai Cancer Center, Fudan University, Shanghai, 200438, P. R. China
| | - Tun Lu
- School of Computer Science, Fudan University, Shanghai, 200438, P. R. China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438, P. R. China
| | - Shaojun Peng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University); Zhuhai, Guangdong, 519000, P. R. China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| | - Dongsheng Li
- Microsoft Research Asia, Shanghai, 200232, P. R. China.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, P. R. China.
| |
Collapse
|
19
|
Albanese KI, Petrenas R, Pirro F, Naudin EA, Borucu U, Dawson WM, Scott DA, Leggett GJ, Weiner OD, Oliver TAA, Woolfson DN. Rationally seeded computational protein design of ɑ-helical barrels. Nat Chem Biol 2024; 20:991-999. [PMID: 38902458 PMCID: PMC11288890 DOI: 10.1038/s41589-024-01642-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 05/09/2024] [Indexed: 06/22/2024]
Abstract
Computational protein design is advancing rapidly. Here we describe efficient routes starting from validated parallel and antiparallel peptide assemblies to design two families of α-helical barrel proteins with central channels that bind small molecules. Computational designs are seeded by the sequences and structures of defined de novo oligomeric barrel-forming peptides, and adjacent helices are connected by loop building. For targets with antiparallel helices, short loops are sufficient. However, targets with parallel helices require longer connectors; namely, an outer layer of helix-turn-helix-turn-helix motifs that are packed onto the barrels. Throughout these computational pipelines, residues that define open states of the barrels are maintained. This minimizes sequence sampling, accelerating the design process. For each of six targets, just two to six synthetic genes are made for expression in Escherichia coli. On average, 70% of these genes express to give soluble monomeric proteins that are fully characterized, including high-resolution structures for most targets that match the design models with high accuracy.
Collapse
Affiliation(s)
- Katherine I Albanese
- School of Chemistry, University of Bristol, Bristol, UK
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol, Bristol, UK
| | | | - Fabio Pirro
- School of Chemistry, University of Bristol, Bristol, UK
| | | | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Medical Sciences Building, Bristol, UK
| | | | - D Arne Scott
- Rosa Biotech, Science Creates St Philips, Bristol, UK
| | | | - Orion D Weiner
- Cardiovascular Research Institute, Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | | | - Derek N Woolfson
- School of Chemistry, University of Bristol, Bristol, UK.
- Max Planck-Bristol Centre for Minimal Biology, University of Bristol, Bristol, UK.
- School of Biochemistry, University of Bristol, Medical Sciences Building, Bristol, UK.
- Bristol BioDesign Institute, University of Bristol, Bristol, UK.
| |
Collapse
|
20
|
Vijayakumar S, Alberstein RG, Zhang Z, Lu YS, Chan A, Wahl CE, Ha JS, Hunka DE, Boss GR, Sailor MJ, Tezcan FA. Designed 2D protein crystals as dynamic molecular gatekeepers for a solid-state device. Nat Commun 2024; 15:6326. [PMID: 39068153 PMCID: PMC11283500 DOI: 10.1038/s41467-024-50567-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
The sensitivity and responsiveness of living cells to environmental changes are enabled by dynamic protein structures, inspiring efforts to construct artificial supramolecular protein assemblies. However, despite their sophisticated structures, designed protein assemblies have yet to be incorporated into macroscale devices for real-life applications. We report a 2D crystalline protein assembly of C98/E57/E66L-rhamnulose-1-phosphate aldolase (CEERhuA) that selectively blocks or passes molecular species when exposed to a chemical trigger. CEERhuA crystals are engineered via cobalt(II) coordination bonds to undergo a coherent conformational change from a closed state (pore dimensions <1 nm) to an ajar state (pore dimensions ~4 nm) when exposed to an HCN(g) trigger. When layered onto a mesoporous silicon (pSi) photonic crystal optical sensor configured to detect HCN(g), the 2D CEERhuA crystal layer effectively blocks interferents that would otherwise result in a false positive signal. The 2D CEERhuA crystal layer opens in selective response to low-ppm levels of HCN(g), allowing analyte penetration into the pSi sensor layer for detection. These findings illustrate that designed protein assemblies can function as dynamic components of solid-state devices in non-aqueous environments.
Collapse
Affiliation(s)
- Sanahan Vijayakumar
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert G Alberstein
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Zhiyin Zhang
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yi-Sheng Lu
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Adriano Chan
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - James S Ha
- Leidos, 4161 Campus Point Ct, San Diego, CA, 92121, USA
- Battelle, 505 King Ave Columbus, Ohio, OH, 43201, USA
| | | | - Gerry R Boss
- Department of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael J Sailor
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - F Akif Tezcan
- Materials Science and Engineering Program, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Chemistry & Biochemistry, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
21
|
Wu F, Huang Y, Yang G, Ye S, Mukamel S, Jiang J. Unraveling dynamic protein structures by two-dimensional infrared spectra with a pretrained machine learning model. Proc Natl Acad Sci U S A 2024; 121:e2409257121. [PMID: 38917009 PMCID: PMC11228460 DOI: 10.1073/pnas.2409257121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Dynamic protein structures are crucial for deciphering their diverse biological functions. Two-dimensional infrared (2DIR) spectroscopy stands as an ideal tool for tracing rapid conformational evolutions in proteins. However, linking spectral characteristics to dynamic structures poses a formidable challenge. Here, we present a pretrained machine learning model based on 2DIR spectra analysis. This model has learned signal features from approximately 204,300 spectra to establish a "spectrum-structure" correlation, thereby tracing the dynamic conformations of proteins. It excels in accurately predicting the dynamic content changes of various secondary structures and demonstrates universal transferability on real folding trajectories spanning timescales from microseconds to milliseconds. Beyond exceptional predictive performance, the model offers attention-based spectral explanations of dynamic conformational changes. Our 2DIR-based pretrained model is anticipated to provide unique insights into the dynamic structural information of proteins in their native environments.
Collapse
Affiliation(s)
- Fan Wu
- Key Laboratory of Precision and Intelligent Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei230026, Anhui, China
| | - Yan Huang
- Key Laboratory of Precision and Intelligent Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei230026, Anhui, China
| | - Guokun Yang
- Key Laboratory of Precision and Intelligent Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei230026, Anhui, China
| | - Sheng Ye
- Anhui Provincial Engineering Research Center for Unmanned System and Intelligent Technology, School of Artificial Intelligence, Anhui University, Hefei230601, Anhui, China
| | - Shaul Mukamel
- Department of Chemistry and of Physics & Astronomy, University of California, Irvine, CA92697
| | - Jun Jiang
- Key Laboratory of Precision and Intelligent Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei230026, Anhui, China
| |
Collapse
|
22
|
Laakko T, Korkealaakso A, Yildirir BF, Batys P, Liljeström V, Hokkanen A, Nonappa, Penttilä M, Laukkanen A, Miserez A, Södergård C, Mohammadi P. Accelerated Engineering of ELP-Based Materials through Hybrid Biomimetic-De Novo Predictive Molecular Design. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312299. [PMID: 38710202 DOI: 10.1002/adma.202312299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/28/2024] [Indexed: 05/08/2024]
Abstract
Efforts to engineer high-performance protein-based materials inspired by nature have mostly focused on altering naturally occurring sequences to confer the desired functionalities, whereas de novo design lags significantly behind and calls for unconventional innovative approaches. Here, using partially disordered elastin-like polypeptides (ELPs) as initial building blocks this work shows that de novo engineering of protein materials can be accelerated through hybrid biomimetic design, which this work achieves by integrating computational modeling, deep neural network, and recombinant DNA technology. This generalizable approach involves incorporating a series of de novo-designed sequences with α-helical conformation and genetically encoding them into biologically inspired intrinsically disordered repeating motifs. The new ELP variants maintain structural conformation and showed tunable supramolecular self-assembly out of thermal equilibrium with phase behavior in vitro. This work illustrates the effective translation of the predicted molecular designs in structural and functional materials. The proposed methodology can be applied to a broad range of partially disordered biomacromolecules and potentially pave the way toward the discovery of novel structural proteins.
Collapse
Affiliation(s)
- Timo Laakko
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| | | | - Burcu Firatligil Yildirir
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 6, Tampere, FI-33720, Finland
| | - Piotr Batys
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Niezapominajek 8, Krakow, PL-30239, Poland
| | - Ville Liljeström
- Department of Applied Physics, School of Science, Aalto University, Aalto, FI-00076, Finland
| | - Ari Hokkanen
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| | - Nonappa
- Faculty of Engineering and Natural Sciences, Tampere University, Korkeakoulunkatu 6, Tampere, FI-33720, Finland
| | - Merja Penttilä
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| | - Anssi Laukkanen
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| | - Ali Miserez
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University (NTU), Singapore, 637553, Singapore
- School of Biological Sciences, NTU, Singapore, 637551, Singapore
| | - Caj Södergård
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| | - Pezhman Mohammadi
- VTT Technical Research Centre of Finland Ltd., VTT, FI-02044, Finland
| |
Collapse
|
23
|
Winnifrith A, Outeiral C, Hie BL. Generative artificial intelligence for de novo protein design. Curr Opin Struct Biol 2024; 86:102794. [PMID: 38663170 DOI: 10.1016/j.sbi.2024.102794] [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: 12/21/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 05/19/2024]
Abstract
Engineering new molecules with desirable functions and properties has the potential to extend our ability to engineer proteins beyond what nature has so far evolved. Advances in the so-called 'de novo' design problem have recently been brought forward by developments in artificial intelligence. Generative architectures, such as language models and diffusion processes, seem adept at generating novel, yet realistic proteins that display desirable properties and perform specified functions. State-of-the-art design protocols now achieve experimental success rates nearing 20%, thus widening the access to de novo designed proteins. Despite extensive progress, there are clear field-wide challenges, for example, in determining the best in silico metrics to prioritise designs for experimental testing, and in designing proteins that can undergo large conformational changes or be regulated by post-translational modifications. With an increase in the number of models being developed, this review provides a framework to understand how these tools fit into the overall process of de novo protein design. Throughout, we highlight the power of incorporating biochemical knowledge to improve performance and interpretability.
Collapse
Affiliation(s)
- Adam Winnifrith
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford, OX1 3QU, United Kingdom; Evolvere Biosciences, Innovation Building, Old Road Campus, Oxford, OX3 7FZ, United Kingdom.
| | - Carlos Outeiral
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford OX1 3LB, United Kingdom.
| | - Brian L Hie
- Department of Chemical Engineering, Stanford University, 443 Via Ortega, Stanford, CA 94305, USA; Stanford Data Science, 475 Via Ortega, Stanford CA 94305, USA; Arc Institute, 3181 Porter Dr, Palo Alto, CA, USA.
| |
Collapse
|
24
|
de Haas RJ, Brunette N, Goodson A, Dauparas J, Yi SY, Yang EC, Dowling Q, Nguyen H, Kang A, Bera AK, Sankaran B, de Vries R, Baker D, King NP. Rapid and automated design of two-component protein nanomaterials using ProteinMPNN. Proc Natl Acad Sci U S A 2024; 121:e2314646121. [PMID: 38502697 PMCID: PMC10990136 DOI: 10.1073/pnas.2314646121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
The design of protein-protein interfaces using physics-based design methods such as Rosetta requires substantial computational resources and manual refinement by expert structural biologists. Deep learning methods promise to simplify protein-protein interface design and enable its application to a wide variety of problems by researchers from various scientific disciplines. Here, we test the ability of a deep learning method for protein sequence design, ProteinMPNN, to design two-component tetrahedral protein nanomaterials and benchmark its performance against Rosetta. ProteinMPNN had a similar success rate to Rosetta, yielding 13 new experimentally confirmed assemblies, but required orders of magnitude less computation and no manual refinement. The interfaces designed by ProteinMPNN were substantially more polar than those designed by Rosetta, which facilitated in vitro assembly of the designed nanomaterials from independently purified components. Crystal structures of several of the assemblies confirmed the accuracy of the design method at high resolution. Our results showcase the potential of deep learning-based methods to unlock the widespread application of designed protein-protein interfaces and self-assembling protein nanomaterials in biotechnology.
Collapse
Affiliation(s)
- Robbert J. de Haas
- Department of Physical Chemistry and Soft Matter, Wageningen University and Research, Wageningen6078 WE, The Netherlands
| | - Natalie Brunette
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Alex Goodson
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Sue Y. Yi
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Erin C. Yang
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Quinton Dowling
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Hannah Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Asim K. Bera
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| | - Renko de Vries
- Department of Physical Chemistry and Soft Matter, Wageningen University and Research, Wageningen6078 WE, The Netherlands
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
- HHMI, Seattle, WA98195
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, WA98195
- Institute for Protein Design, University of Washington, Seattle, WA98195
| |
Collapse
|
25
|
Li M, Li J, Liu K, Zhang H. Artificial structural proteins: Synthesis, assembly and material applications. Bioorg Chem 2024; 144:107162. [PMID: 38308999 DOI: 10.1016/j.bioorg.2024.107162] [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: 10/30/2023] [Revised: 01/14/2024] [Accepted: 01/27/2024] [Indexed: 02/05/2024]
Abstract
Structural proteins have evolved over billions of years and offer outstanding mechanical properties, such as resilience, toughness and stiffness. Advances in modular protein engineering, polypeptide modification, and synthetic biology have led to the development of novel biomimetic structural proteins to perform in biomedical and military fields. However, the development of customized structural proteins and assemblies with superior performance remains a major challenge, due to the inherent limitations of biosynthesis, difficulty in mimicking the complexed macroscale assembly, etc. This review summarizes the approaches for the design and production of biomimetic structural proteins, and their chemical modifications for multiscale assembly. Furthermore, we discuss the function tailoring and current applications of biomimetic structural protein assemblies. A perspective of future research is to reveal how the mechanical properties are encoded in the sequences and conformations. This review, therefore, provides an important reference for the development of structural proteins-mimetics from replication of nature to even outperforming nature.
Collapse
Affiliation(s)
- Ming Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China; Engineering Research Center of Advanced Rare Earth Materials, Ministry of Education, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China; Engineering Research Center of Advanced Rare Earth Materials, Ministry of Education, Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
26
|
Zheng W, Xu YF, Hu ZM, Li K, Xu ZQ, Sun JL, Wei JF. Artificial intelligence-driven design of the assembled major cat allergen Fel d 1 to improve its spatial folding and IgE-reactivity. Int Immunopharmacol 2024; 128:111488. [PMID: 38185034 DOI: 10.1016/j.intimp.2024.111488] [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: 10/16/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND Cat-derived allergens are considered as one of the most common causes of allergic diseases worldwide. Fel d 1 is a major cat allergen and plays an important role in immunoglobulin E (IgE)-reaction diagnosis. However, the two separate chains of Fel d 1 exhibited lower IgE-reactivity than its complete molecule of an assembled form, which makes it difficult to efficiently prepare and limits the application of Fel d 1 in molecular diagnosis of cat allergy. METHODS We first applied artificial intelligence (AI) based tool AlphaFold2 to build the 3-dimensional structures of Fel d 1 with different connection modes between two chains, which were evaluated by ERRAT program and were expressed in Escherichia coli. We then calculated the expression ratios of soluble form/inclusion bodies form of optimized Fel d 1. The Circular Dichroism (CD), High Performance Liquid Chromatography-Size Exclusion Chromatography (HPLC-SEC) and reducing/non-reducing SDS-PAGE were performed to characterize the folding status and dimerization of the optimized fusion Fel d 1. The improvement of specific-IgE reactivity to optimized fusion Fel d 1 was investigated by enzyme linked immunosorbent assay (ELISA). RESULTS Among several linkers, 2 × GGGGS got the highest scores, with an overall quality factor of 100. The error value of the residues around the junction of 2 × GGGGS was lower than others. It exhibited highest proportion of soluble protein than other Fel d 1 constructs with ERRAT (GGGGS, KK as well as direct fusion Fel d 1). The results of CD and HPLC-SEC showed the consistent folding and dimerization of two fused subunits between the optimized fusion Fel d 1 and previously well-defined direct fusion Fel d 1. The overall IgE-binding absorbance of optimized fusion Fel d 1 tested by ELISA was improved compared with that of the direct fusion Fel d 1. CONCLUSION We firstly provided an AI-design strategy to optimize the Fel d 1, which could spontaneously fold into its native-like structure without additional refolding process or eukaryotic folding factors. The improved IgE-binding activity and simplified preparation method could greatly facilitate it to be a robust allergen material for molecular diagnosis of cat allergy.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Fei Xu
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Ming Hu
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ke Li
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Qiang Xu
- Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Jin-Lyu Sun
- Department of Allergy, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China; Research Division of Clinical Pharmacology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
27
|
McCafferty CL, Klumpe S, Amaro RE, Kukulski W, Collinson L, Engel BD. Integrating cellular electron microscopy with multimodal data to explore biology across space and time. Cell 2024; 187:563-584. [PMID: 38306982 DOI: 10.1016/j.cell.2024.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Biology spans a continuum of length and time scales. Individual experimental methods only glimpse discrete pieces of this spectrum but can be combined to construct a more holistic view. In this Review, we detail the latest advancements in volume electron microscopy (vEM) and cryo-electron tomography (cryo-ET), which together can visualize biological complexity across scales from the organization of cells in large tissues to the molecular details inside native cellular environments. In addition, we discuss emerging methodologies for integrating three-dimensional electron microscopy (3DEM) imaging with multimodal data, including fluorescence microscopy, mass spectrometry, single-particle analysis, and AI-based structure prediction. This multifaceted approach fills gaps in the biological continuum, providing functional context, spatial organization, molecular identity, and native interactions. We conclude with a perspective on incorporating diverse data into computational simulations that further bridge and extend length scales while integrating the dimension of time.
Collapse
Affiliation(s)
| | - Sven Klumpe
- Research Group CryoEM Technology, Max-Planck-Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany.
| | - Rommie E Amaro
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Wanda Kukulski
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Benjamin D Engel
- Biozentrum, University of Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
28
|
Notin P, Rollins N, Gal Y, Sander C, Marks D. Machine learning for functional protein design. Nat Biotechnol 2024; 42:216-228. [PMID: 38361074 DOI: 10.1038/s41587-024-02127-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 01/05/2024] [Indexed: 02/17/2024]
Abstract
Recent breakthroughs in AI coupled with the rapid accumulation of protein sequence and structure data have radically transformed computational protein design. New methods promise to escape the constraints of natural and laboratory evolution, accelerating the generation of proteins for applications in biotechnology and medicine. To make sense of the exploding diversity of machine learning approaches, we introduce a unifying framework that classifies models on the basis of their use of three core data modalities: sequences, structures and functional labels. We discuss the new capabilities and outstanding challenges for the practical design of enzymes, antibodies, vaccines, nanomachines and more. We then highlight trends shaping the future of this field, from large-scale assays to more robust benchmarks, multimodal foundation models, enhanced sampling strategies and laboratory automation.
Collapse
Affiliation(s)
- Pascal Notin
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Department of Computer Science, University of Oxford, Oxford, UK.
| | | | - Yarin Gal
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Chris Sander
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Debora Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
29
|
Hamilton S, Kingston BR. Applying artificial intelligence and computational modeling to nanomedicine. Curr Opin Biotechnol 2024; 85:103043. [PMID: 38091874 DOI: 10.1016/j.copbio.2023.103043] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 02/09/2024]
Abstract
Achieving specific and targeted delivery of nanomedicines to diseased tissues is a major challenge. This is because the process of designing, formulating, testing, and selecting a nanoparticle delivery vehicle for a specific disease target is governed by complex multivariate interactions. Computational modeling and artificial intelligence are well-suited for analyzing and modeling large multivariate datasets in short periods of time. Computational approaches can be applied to help design nanomedicine formulations, interpret nanoparticle-biological interactions, and create models from high-throughput screening techniques to improve the selection of the ideal nanoparticle carrier. In the future, many steps in the nanomedicine development process will be done computationally, reducing the number of experiments and time needed to select the ideal nanomedicine formulation.
Collapse
Affiliation(s)
- Sean Hamilton
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, 2720 S. Moody Avenue, Portland, OR 97201, United States
| | - Benjamin R Kingston
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, 2720 S. Moody Avenue, Portland, OR 97201, United States.
| |
Collapse
|
30
|
Min J, Rong X, Zhang J, Su R, Wang Y, Qi W. Computational Design of Peptide Assemblies. J Chem Theory Comput 2024; 20:532-550. [PMID: 38206800 DOI: 10.1021/acs.jctc.3c01054] [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: 01/13/2024]
Abstract
With the ongoing development of peptide self-assembling materials, there is growing interest in exploring novel functional peptide sequences. From short peptides to long polypeptides, as the functionality increases, the sequence space is also expanding exponentially. Consequently, attempting to explore all functional sequences comprehensively through experience and experiments alone has become impractical. By utilizing computational methods, especially artificial intelligence enhanced molecular dynamics (MD) simulation and de novo peptide design, there has been a significant expansion in the exploration of sequence space. Through these methods, a variety of supramolecular functional materials, including fibers, two-dimensional arrays, nanocages, etc., have been designed by meticulously controlling the inter- and intramolecular interactions. In this review, we first provide a brief overview of the current main computational methods and then focus on the computational design methods for various self-assembled peptide materials. Additionally, we introduce some representative protein self-assemblies to offer guidance for the design of self-assembling peptides.
Collapse
Affiliation(s)
- Jiwei Min
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xi Rong
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Jiaxing Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin 300072, P. R. China
| |
Collapse
|
31
|
Teng F, Cui T, Zhou L, Gao Q, Zhou Q, Li W. Programmable synthetic receptors: the next-generation of cell and gene therapies. Signal Transduct Target Ther 2024; 9:7. [PMID: 38167329 PMCID: PMC10761793 DOI: 10.1038/s41392-023-01680-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/22/2023] [Accepted: 10/11/2023] [Indexed: 01/05/2024] Open
Abstract
Cell and gene therapies hold tremendous promise for treating a range of difficult-to-treat diseases. However, concerns over the safety and efficacy require to be further addressed in order to realize their full potential. Synthetic receptors, a synthetic biology tool that can precisely control the function of therapeutic cells and genetic modules, have been rapidly developed and applied as a powerful solution. Delicately designed and engineered, they can be applied to finetune the therapeutic activities, i.e., to regulate production of dosed, bioactive payloads by sensing and processing user-defined signals or biomarkers. This review provides an overview of diverse synthetic receptor systems being used to reprogram therapeutic cells and their wide applications in biomedical research. With a special focus on four synthetic receptor systems at the forefront, including chimeric antigen receptors (CARs) and synthetic Notch (synNotch) receptors, we address the generalized strategies to design, construct and improve synthetic receptors. Meanwhile, we also highlight the expanding landscape of therapeutic applications of the synthetic receptor systems as well as current challenges in their clinical translation.
Collapse
Affiliation(s)
- Fei Teng
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Tongtong Cui
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Zhou
- University of Chinese Academy of Sciences, Beijing, 101408, China
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingqin Gao
- University of Chinese Academy of Sciences, Beijing, 101408, China
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Zhou
- University of Chinese Academy of Sciences, Beijing, 101408, China.
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Wei Li
- University of Chinese Academy of Sciences, Beijing, 101408, China.
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| |
Collapse
|
32
|
Aguilera-Puga MDC, Cancelarich NL, Marani MM, de la Fuente-Nunez C, Plisson F. Accelerating the Discovery and Design of Antimicrobial Peptides with Artificial Intelligence. Methods Mol Biol 2024; 2714:329-352. [PMID: 37676607 DOI: 10.1007/978-1-0716-3441-7_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Peptides modulate many processes of human physiology targeting ion channels, protein receptors, or enzymes. They represent valuable starting points for the development of new biologics against communicable and non-communicable disorders. However, turning native peptide ligands into druggable materials requires high selectivity and efficacy, predictable metabolism, and good safety profiles. Machine learning models have gradually emerged as cost-effective and time-saving solutions to predict and generate new proteins with optimal properties. In this chapter, we will discuss the evolution and applications of predictive modeling and generative modeling to discover and design safe and effective antimicrobial peptides. We will also present their current limitations and suggest future research directions, applicable to peptide drug design campaigns.
Collapse
Affiliation(s)
- Mariana D C Aguilera-Puga
- Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Irapuato, Guanajuato, Mexico
- CINVESTAV-IPN, Unidad Irapuato, Departamento de Biotecnología y Bioquímica, Irapuato, Guanajuato, Mexico
| | - Natalia L Cancelarich
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales (IPEEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Puerto Madryn, Argentina
| | - Mariela M Marani
- Instituto Patagónico para el Estudio de los Ecosistemas Continentales (IPEEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Puerto Madryn, Argentina
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| | - Fabien Plisson
- Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Unidad de Genómica Avanzada, Laboratorio Nacional de Genómica para la Biodiversidad (Langebio), Irapuato, Guanajuato, Mexico.
- CINVESTAV-IPN, Unidad Irapuato, Departamento de Biotecnología y Bioquímica, Irapuato, Guanajuato, Mexico.
| |
Collapse
|
33
|
Ellis D, Dosey A, Boyoglu-Barnum S, Park YJ, Gillespie R, Syeda H, Hutchinson GB, Tsybovsky Y, Murphy M, Pettie D, Matheson N, Chan S, Ueda G, Fallas JA, Carter L, Graham BS, Veesler D, Kanekiyo M, King NP. Antigen spacing on protein nanoparticles influences antibody responses to vaccination. Cell Rep 2023; 42:113552. [PMID: 38096058 PMCID: PMC10801709 DOI: 10.1016/j.celrep.2023.113552] [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: 06/20/2023] [Revised: 09/28/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Immunogen design approaches aim to control the specificity and quality of antibody responses elicited by next-generation vaccines. Here, we use computational protein design to generate a nanoparticle vaccine platform based on the receptor-binding domain (RBD) of influenza hemagglutinin (HA) that enables precise control of antigen conformation and spacing. HA RBDs are presented as either monomers or native-like closed trimers that are connected to the underlying nanoparticle by a rigid linker that is modularly extended to precisely control antigen spacing. Nanoparticle immunogens with decreased spacing between trimeric RBDs elicit antibodies with improved hemagglutination inhibition and neutralization potency as well as binding breadth across diverse H1 HAs. Our "trihead" nanoparticle immunogen platform provides insights into anti-HA immunity, establishes antigen spacing as an important parameter in structure-based vaccine design, and embodies several design features that could be used in next-generation vaccines against influenza and other viruses.
Collapse
Affiliation(s)
- Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195, USA
| | - Annie Dosey
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Rebecca Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hubza Syeda
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Geoffrey B Hutchinson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Michael Murphy
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Deleah Pettie
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Nick Matheson
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jorge A Fallas
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
34
|
Tian J, Zhang J, Francis F. The role and pathway of VQ family in plant growth, immunity, and stress response. PLANTA 2023; 259:16. [PMID: 38078967 DOI: 10.1007/s00425-023-04292-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
MAIN CONCLUSION This review provides a detailed description of the function and mechanism of VQ family gene, which is helpful for further research and application of VQ gene resources to improve crops. Valine-glutamine (VQ) motif-containing proteins are a large class of transcriptional regulatory cofactors. VQ proteins have their own unique molecular characteristics. Amino acids are highly conserved only in the VQ domain, while other positions vary greatly. Most VQ genes do not contain introns and the length of their proteins is less than 300 amino acids. A majority of VQ proteins are predicted to be localized in the nucleus. The promoter of many VQ genes contains stress or growth related elements. Segment duplication and tandem duplication are the main amplification mechanisms of the VQ gene family in angiosperms and gymnosperms, respectively. Purification selection plays a crucial role in the evolution of many VQ genes. By interacting with WRKY, MAPK, and other proteins, VQ proteins participate in the multiple signaling pathways to regulate plant growth and development, as well as defense responses to biotic and abiotic stresses. Although there have been some reports on the VQ gene family in plants, most of them only identify family members, with little functional verification, and there is also a lack of complete, detailed, and up-to-date review of research progress. Here, we comprehensively summarized the research progress of VQ genes that have been published so far, mainly including their molecular characteristics, biological functions, importance of VQ motif, and working mechanisms. Finally, the regulatory network and model of VQ genes were drawn, a precise molecular breeding strategy based on VQ genes was proposed, and the current problems and future prospects were pointed out, providing a powerful reference for further research and utilization of VQ genes in plant improvement.
Collapse
Affiliation(s)
- Jinfu Tian
- Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, 5030, Gembloux, Belgium.
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100081, China.
| | - Jiahui Zhang
- Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, 5030, Gembloux, Belgium
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100081, China
| | - Frédéric Francis
- Functional and Evolutionary Entomology, Gembloux Agro-Bio Tech, University of Liège, 5030, Gembloux, Belgium
| |
Collapse
|
35
|
Kouba P, Kohout P, Haddadi F, Bushuiev A, Samusevich R, Sedlar J, Damborsky J, Pluskal T, Sivic J, Mazurenko S. Machine Learning-Guided Protein Engineering. ACS Catal 2023; 13:13863-13895. [PMID: 37942269 PMCID: PMC10629210 DOI: 10.1021/acscatal.3c02743] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Indexed: 11/10/2023]
Abstract
Recent progress in engineering highly promising biocatalysts has increasingly involved machine learning methods. These methods leverage existing experimental and simulation data to aid in the discovery and annotation of promising enzymes, as well as in suggesting beneficial mutations for improving known targets. The field of machine learning for protein engineering is gathering steam, driven by recent success stories and notable progress in other areas. It already encompasses ambitious tasks such as understanding and predicting protein structure and function, catalytic efficiency, enantioselectivity, protein dynamics, stability, solubility, aggregation, and more. Nonetheless, the field is still evolving, with many challenges to overcome and questions to address. In this Perspective, we provide an overview of ongoing trends in this domain, highlight recent case studies, and examine the current limitations of machine learning-based methods. We emphasize the crucial importance of thorough experimental validation of emerging models before their use for rational protein design. We present our opinions on the fundamental problems and outline the potential directions for future research.
Collapse
Affiliation(s)
- Petr Kouba
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
- Faculty of
Electrical Engineering, Czech Technical
University in Prague, Technicka 2, 166 27 Prague 6, Czech Republic
| | - Pavel Kohout
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Faraneh Haddadi
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Anton Bushuiev
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Raman Samusevich
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 160 00 Prague 6, Czech Republic
| | - Jiri Sedlar
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Jiri Damborsky
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Tomas Pluskal
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 160 00 Prague 6, Czech Republic
| | - Josef Sivic
- Czech Institute
of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Jugoslavskych partyzanu 1580/3, 160 00 Prague 6, Czech Republic
| | - Stanislav Mazurenko
- Loschmidt
Laboratories, Department of Experimental Biology and RECETOX, Faculty
of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech
Republic
- International
Clinical Research Center, St. Anne’s
University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
36
|
Ryu G, Kim GB, Yu T, Lee SY. Deep learning for metabolic pathway design. Metab Eng 2023; 80:130-141. [PMID: 37734652 DOI: 10.1016/j.ymben.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/23/2023]
Abstract
The establishment of a bio-based circular economy is imperative in tackling the climate crisis and advancing sustainable development. In this realm, the creation of microbial cell factories is central to generating a variety of chemicals and materials. The design of metabolic pathways is crucial in shaping these microbial cell factories, especially when it comes to producing chemicals with yet-to-be-discovered biosynthetic routes. To aid in navigating the complexities of chemical and metabolic domains, computer-supported tools for metabolic pathway design have emerged. In this paper, we evaluate how digital strategies can be employed for pathway prediction and enzyme discovery. Additionally, we touch upon the recent strides made in using deep learning techniques for metabolic pathway prediction. These computational tools and strategies streamline the design of metabolic pathways, facilitating the development of microbial cell factories. Leveraging the capabilities of deep learning in metabolic pathway design is profoundly promising, potentially hastening the advent of a bio-based circular economy.
Collapse
Affiliation(s)
- Gahyeon Ryu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, 34141, Republic of Korea
| | - Gi Bae Kim
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, 34141, Republic of Korea
| | - Taeho Yu
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, 34141, Republic of Korea
| | - Sang Yup Lee
- Metabolic and Biomolecular Engineering National Research Laboratory, Department of Chemical and Biomolecular Engineering (BK21 Four), KAIST Institute for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea; Systems Metabolic Engineering and Systems Healthcare Cross-Generation Collaborative Laboratory, KAIST, Daejeon, 34141, Republic of Korea; BioProcess Engineering Research Center and BioInformatics Research Center, KAIST, Daejeon, 34141, Republic of Korea; Graduate School of Engineering Biology, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
37
|
Khmelinskaia A, Bethel NP, Fatehi F, Antanasijevic A, Borst AJ, Lai SH, Wang JYJ, Mallik BB, Miranda MC, Watkins AM, Ogohara C, Caldwell S, Wu M, Heck AJR, Veesler D, Ward AB, Baker D, Twarock R, King NP. Local structural flexibility drives oligomorphism in computationally designed protein assemblies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562842. [PMID: 37905007 PMCID: PMC10614843 DOI: 10.1101/2023.10.18.562842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Many naturally occurring protein assemblies have dynamic structures that allow them to perform specialized functions. For example, clathrin coats adopt a wide variety of architectures to adapt to vesicular cargos of various sizes. Although computational methods for designing novel self-assembling proteins have advanced substantially over the past decade, most existing methods focus on designing static structures with high accuracy. Here we characterize the structures of three distinct computationally designed protein assemblies that each form multiple unanticipated architectures, and identify flexibility in specific regions of the subunits of each assembly as the source of structural diversity. Cryo-EM single-particle reconstructions and native mass spectrometry showed that only two distinct architectures were observed in two of the three cases, while we obtained six cryo-EM reconstructions that likely represent a subset of the architectures present in solution in the third case. Structural modeling and molecular dynamics simulations indicated that the surprising observation of a defined range of architectures, instead of non-specific aggregation, can be explained by constrained flexibility within the building blocks. Our results suggest that deliberate use of structural flexibility as a design principle will allow exploration of previously inaccessible structural and functional space in designed protein assemblies.
Collapse
|
38
|
Horns F, Martinez JA, Fan C, Haque M, Linton JM, Tobin V, Santat L, Maggiolo AO, Bjorkman PJ, Lois C, Elowitz MB. Engineering RNA export for measurement and manipulation of living cells. Cell 2023; 186:3642-3658.e32. [PMID: 37437570 PMCID: PMC10528933 DOI: 10.1016/j.cell.2023.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/08/2023] [Accepted: 06/19/2023] [Indexed: 07/14/2023]
Abstract
A system for programmable export of RNA molecules from living cells would enable both non-destructive monitoring of cell dynamics and engineering of cells capable of delivering executable RNA programs to other cells. We developed genetically encoded cellular RNA exporters, inspired by viruses, that efficiently package and secrete cargo RNA molecules from mammalian cells within protective nanoparticles. Exporting and sequencing RNA barcodes enabled non-destructive monitoring of cell population dynamics with clonal resolution. Further, by incorporating fusogens into the nanoparticles, we demonstrated the delivery, expression, and functional activity of exported mRNA in recipient cells. We term these systems COURIER (controlled output and uptake of RNA for interrogation, expression, and regulation). COURIER enables measurement of cell dynamics and establishes a foundation for hybrid cell and gene therapies based on cell-to-cell delivery of RNA.
Collapse
Affiliation(s)
- Felix Horns
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Joe A Martinez
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Chengcheng Fan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mehernaz Haque
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - James M Linton
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Victoria Tobin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Leah Santat
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ailiena O Maggiolo
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Carlos Lois
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
39
|
Samineni L, Acharya B, Behera H, Oh H, Kumar M, Chowdhury R. Protein engineering of pores for separation, sensing, and sequencing. Cell Syst 2023; 14:676-691. [PMID: 37591205 DOI: 10.1016/j.cels.2023.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/13/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Proteins are critical to cellular function and survival. They are complex molecules with precise structures and chemistries, which allow them to serve diverse functions for maintaining overall cell homeostasis. Since the discovery of the first enzyme in 1833, a gamut of advanced experimental and computational tools has been developed and deployed for understanding protein structure and function. Recent studies have demonstrated the ability to redesign/alter natural proteins for applications in industrial processes of interest and to make customized, novel synthetic proteins in the laboratory through protein engineering. We comprehensively review the successes in engineering pore-forming proteins and correlate the amino acid-level biochemistry of different pore modification strategies to the intended applications limited to nucleotide/peptide sequencing, single-molecule sensing, and precise molecular separations.
Collapse
Affiliation(s)
- Laxmicharan Samineni
- Department of Civil, Architectural and Environmental Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Bibek Acharya
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Harekrushna Behera
- Department of Civil, Architectural and Environmental Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Hyeonji Oh
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Manish Kumar
- Department of Civil, Architectural and Environmental Engineering, University of Texas at Austin, Austin, TX 78712, USA; McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Ratul Chowdhury
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
40
|
de Haas RJ, Brunette N, Goodson A, Dauparas J, Yi SY, Yang EC, Dowling Q, Nguyen H, Kang A, Bera AK, Sankaran B, de Vries R, Baker D, King NP. Rapid and automated design of two-component protein nanomaterials using ProteinMPNN. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.551935. [PMID: 37577478 PMCID: PMC10418170 DOI: 10.1101/2023.08.04.551935] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The design of novel protein-protein interfaces using physics-based design methods such as Rosetta requires substantial computational resources and manual refinement by expert structural biologists. A new generation of deep learning methods promises to simplify protein-protein interface design and enable its application to a wide variety of problems by researchers from various scientific disciplines. Here we test the ability of a deep learning method for protein sequence design, ProteinMPNN, to design two-component tetrahedral protein nanomaterials and benchmark its performance against Rosetta. ProteinMPNN had a similar success rate to Rosetta, yielding 13 new experimentally confirmed assemblies, but required orders of magnitude less computation and no manual refinement. The interfaces designed by ProteinMPNN were substantially more polar than those designed by Rosetta, which facilitated in vitro assembly of the designed nanomaterials from independently purified components. Crystal structures of several of the assemblies confirmed the accuracy of the design method at high resolution. Our results showcase the potential of deep learning-based methods to unlock the widespread application of designed protein-protein interfaces and self-assembling protein nanomaterials in biotechnology.
Collapse
|
41
|
Ren H, Jia W, Xie Y, Yu M, Chen Y. Adjuvant physiochemistry and advanced nanotechnology for vaccine development. Chem Soc Rev 2023; 52:5172-5254. [PMID: 37462107 DOI: 10.1039/d2cs00848c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vaccines comprising innovative adjuvants are rapidly reaching advanced translational stages, such as the authorized nanotechnology adjuvants in mRNA vaccines against COVID-19 worldwide, offering new strategies to effectively combat diseases threatening human health. Adjuvants are vital ingredients in vaccines, which can augment the degree, extensiveness, and longevity of antigen specific immune response. The advances in the modulation of physicochemical properties of nanoplatforms elevate the capability of adjuvants in initiating the innate immune system and adaptive immunity, offering immense potential for developing vaccines against hard-to-target infectious diseases and cancer. In this review, we provide an essential introduction of the basic principles of prophylactic and therapeutic vaccination, key roles of adjuvants in augmenting and shaping immunity to achieve desired outcomes and effectiveness, and the physiochemical properties and action mechanisms of clinically approved adjuvants for humans. We particularly focus on the preclinical and clinical progress of highly immunogenic emerging nanotechnology adjuvants formulated in vaccines for cancer treatment or infectious disease prevention. We deliberate on how the immune system can sense and respond to the physicochemical cues (e.g., chirality, deformability, solubility, topology, and chemical structures) of nanotechnology adjuvants incorporated in the vaccines. Finally, we propose possible strategies to accelerate the clinical implementation of nanotechnology adjuvanted vaccines, such as in-depth elucidation of nano-immuno interactions, antigen identification and optimization by the deployment of high-dimensional multiomics analysis approaches, encouraging close collaborations among scientists from different scientific disciplines and aggressive exploration of novel nanotechnologies.
Collapse
Affiliation(s)
- Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Yujie Xie
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
42
|
Huang T, Li Y. Current progress, challenges, and future perspectives of language models for protein representation and protein design. Innovation (N Y) 2023; 4:100446. [PMID: 37485078 PMCID: PMC10362512 DOI: 10.1016/j.xinn.2023.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/18/2023] [Indexed: 07/25/2023] Open
Abstract
The sequence-structure-function paradigm of protein is the basis of molecular biology. What is the underlying mechanism of such sequence and structure/function corresponding relationship? We reviewed the methods for protein representation and protein design. With these protein representation models, we can accurately predict many properties of proteins, such as stability and binding affinity. Progen, Chroma, RF Diffusion, SCUBA, and other protein design models have demonstrated how human-designed artificial proteins can have desired biological functions. The protein design will revolutionize drug development. And more efficient artificial enzymes that break down industrial waste or plastics will contribute to carbon neutrality. We also discussed the three greatest challenges of protein design in future and possible solutions.
Collapse
Affiliation(s)
- Tao Huang
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yixue Li
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Guangzhou Laboratory, Guangzhou 510005, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai 200433, China
| |
Collapse
|
43
|
Ellis D, Dosey A, Boyoglu-Barnum S, Park YJ, Gillespie R, Syeda H, Tsybovsky Y, Murphy M, Pettie D, Matheson N, Chan S, Ueda G, Fallas JA, Carter L, Graham BS, Veesler D, Kanekiyo M, King NP. Antigen spacing on protein nanoparticles influences antibody responses to vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541980. [PMID: 37292995 PMCID: PMC10245855 DOI: 10.1101/2023.05.23.541980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunogen design approaches aim to control the specificity and quality of antibody responses to enable the creation of next-generation vaccines with improved potency and breadth. However, our understanding of the relationship between immunogen structure and immunogenicity is limited. Here we use computational protein design to generate a self-assembling nanoparticle vaccine platform based on the head domain of influenza hemagglutinin (HA) that enables precise control of antigen conformation, flexibility, and spacing on the nanoparticle exterior. Domain-based HA head antigens were presented either as monomers or in a native-like closed trimeric conformation that prevents exposure of trimer interface epitopes. These antigens were connected to the underlying nanoparticle by a rigid linker that was modularly extended to precisely control antigen spacing. We found that nanoparticle immunogens with decreased spacing between closed trimeric head antigens elicited antibodies with improved hemagglutination inhibition (HAI) and neutralization potency as well as binding breadth across diverse HAs within a subtype. Our "trihead" nanoparticle immunogen platform thus enables new insights into anti-HA immunity, establishes antigen spacing as an important parameter in structure-based vaccine design, and embodies several design features that could be used to generate next-generation vaccines against influenza and other viruses.
Collapse
Affiliation(s)
- Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195, USA
- These authors contributed equally: Daniel Ellis and Annie Dosey
| | - Annie Dosey
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- These authors contributed equally: Daniel Ellis and Annie Dosey
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Rebecca Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hubza Syeda
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Michael Murphy
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Deleah Pettie
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Nick Matheson
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jorge A. Fallas
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|