1
|
Li Y, Zhu J, Zhang Z, Wei J, Wang F, Meisl G, Knowles TPJ, Egelman EH, Tezcan FA. Transforming an ATP-dependent enzyme into a dissipative, self-assembling system. Nat Chem Biol 2025; 21:883-893. [PMID: 39806067 PMCID: PMC12124957 DOI: 10.1038/s41589-024-01811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025]
Abstract
Nucleoside triphosphate (NTP)-dependent protein assemblies such as microtubules and actin filaments have inspired the development of diverse chemically fueled molecular machines and active materials but their functional sophistication has yet to be matched by design. Given this challenge, we asked whether it is possible to transform a natural adenosine 5'-triphosphate (ATP)-dependent enzyme into a dissipative self-assembling system, thereby altering the structural and functional mode in which chemical energy is used. Here we report that FtsH (filamentous temperature-sensitive protease H), a hexameric ATPase involved in membrane protein degradation, can be readily engineered to form one-dimensional helical nanotubes. FtsH nanotubes require constant energy input to maintain their integrity and degrade over time with the concomitant hydrolysis of ATP, analogous to natural NTP-dependent cytoskeletal assemblies. Yet, in contrast to natural dissipative systems, ATP hydrolysis is catalyzed by free FtsH protomers and FtsH nanotubes serve to conserve ATP, leading to transient assemblies whose lifetimes can be tuned from days to minutes through the inclusion of external ATPases in solution.
Collapse
Affiliation(s)
- Yiying Li
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Jie Zhu
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Zhiyin Zhang
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| | - Jiapeng Wei
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, AL, USA
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Edward H Egelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - F Akif Tezcan
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Hatstat AK, Kormos R, Xu V, Du G, Liu L, Zhang SQ, DeGrado WF. A Designed Zn 2+ Sensor Domain Transmits Binding Information to Transmembrane Histidine Kinases. J Am Chem Soc 2025. [PMID: 40388352 DOI: 10.1021/jacs.5c02273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Generating stimulus-responsive allosteric signaling de novo is a significant challenge in protein design. In natural systems like bacterial histidine kinases (HKs), signal transduction occurs when ligand binding initiates a signal that is amplified across biological membranes over long distances to induce large-scale rearrangements and phosphorylation relays. Here, we ask whether our understanding of protein design and multidomain, intramolecular signaling has progressed sufficiently to enable engineering of a HK with tunable de novo components. We generated de novo metal-binding sensor domains and substituted them for the native sensor domain of a transmembrane HK, affording chimeras that transduce signals initiated from a de novo sensor. Signaling depended on the designed sensor's stability and the interdomain linker's phase and length. These results show the usefulness of de novo design to elucidate the biochemical mechanisms and principles of transmembrane signaling.
Collapse
Affiliation(s)
- A Katherine Hatstat
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| | - Rian Kormos
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
- Biophysics Graduate Program, University of California, San Francisco, California 94158-9001, United States
| | - Vee Xu
- Biotechnology Program, City College of San Francisco, San Francisco, California 94112, United States
| | - Guoming Du
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lijun Liu
- Protein Structure and X-ray Crystallography Laboratory, Structural Biology Center, University of Kansas, Lawrence, Kansas 66047, United States
| | - Shao-Qing Zhang
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158-9001, United States
- The Cardiovascular Research Institute, University of California at San Francisco, San Francisco, California 94158-9001, United States
| |
Collapse
|
3
|
Chen W, Choi J. Molecular circuits for genomic recording of cellular events. Trends Genet 2025:S0168-9525(25)00079-4. [PMID: 40335327 DOI: 10.1016/j.tig.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Advances in precise genome editing are enabling genomic recordings of cellular events. Since the initial demonstration of CRISPR-based genome editing, the field of genomic recording has witnessed key strides in lineage recording, where clonal lineage relationships among cells are indirectly recorded as synthetic mutations. However, methods for directly recording and reconstructing past cellular events are still limited, and their potential for revealing new insights into cell fate decisions has yet to be realized. The field needs new sensing modules and genetic circuit architectures that faithfully encode past cellular states into genomic DNA recordings to achieve such goals. Here we review recently developed strategies to construct diverse sensors and explore how emerging synthetic biology tools may help to build molecular circuits for genomic recording of diverse cellular events.
Collapse
Affiliation(s)
- Wei Chen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| | - Junhong Choi
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, NY 10065, USA.
| |
Collapse
|
4
|
Notari E, Wood CW, Michel J. Assessment of the Topology and Oligomerisation States of Coiled Coils Using Metadynamics with Conformational Restraints. J Chem Theory Comput 2025; 21:3260-3276. [PMID: 40042175 PMCID: PMC11948332 DOI: 10.1021/acs.jctc.4c01695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/04/2025] [Accepted: 02/17/2025] [Indexed: 03/26/2025]
Abstract
Coiled-coil proteins provide an excellent scaffold for multistate de novo protein design due to their established sequence-to-structure relationships and ability to switch conformations in response to external stimuli, such as changes in pH or temperature. However, the computational design of multistate coiled-coil protein assemblies is challenging, as it requires accurate estimates of the free energy differences between multiple alternative coiled-coil conformations. Here, we demonstrate how this challenge can be tackled using metadynamics simulations with orientational, positional and conformational restraints. We show that, even for subtle sequence variations, our protocol can predict the preferred topology of coiled-coil dimers and trimers, the preferred oligomerization states of coiled-coil dimers, trimers, and tetramers, as well as the switching behavior of a pH-dependent multistate system. Our approach provides a method for predicting the stability of coiled-coil designs and offers a new framework for computing binding free energies in protein-protein and multiprotein complexes.
Collapse
Affiliation(s)
- Evangelia Notari
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, U.K.
| | - Christopher W. Wood
- School
of Biological Sciences, University of Edinburgh, Roger Land Building, Edinburgh EH9 3FF, U.K.
| | - Julien Michel
- EaStCHEM
School of Chemistry, University of Edinburgh, David Brewster Road, Edinburgh EH9 3FJ, U.K.
| |
Collapse
|
5
|
Grosvirt-Dramen A, Urbach ZJ, Hurst PJ, Kwok CE, Patterson JP, Wang F, Hochbaum AI. Hierarchical Assembly of Conductive Fibers from Coiled-Coil Peptide Building Blocks. ACS NANO 2025; 19:10162-10172. [PMID: 40052932 DOI: 10.1021/acsnano.4c17248] [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] [Indexed: 03/19/2025]
Abstract
Biology provides many sources of inspiration for synthetic and multifunctional nanomaterials. Naturally evolved proteins exhibit specialized, sequence-defined functions and self-assembly behavior. Recapitulating their molecularly defined self-assembly behavior, however, is challenging in de novo proteins. Peptides, on the other hand, represent a more well-defined and rationally designable space with the potential for sequence-programmable, stimuli-responsive design for structure and function, making them ideal building blocks of bioelectronic interfaces. In this work, we design peptides that exhibit stimuli-responsive self-assembly and the capacity to transport electrical current over micrometer-long distances. A lysine-lysine (KK) motif inserted at solvent-exposed positions of a coiled-coil-forming peptide sequence introduces pH-dependent control over a transition from unordered to α-helical peptide structure. The ordered state of the peptide serves as a building block for the assembly of coiled coils and higher-order assemblies. Cryo-EM structures of these structures reveal a hierarchical organization of α-helical peptides in a cross coiled coil (CCC) arrangement. Structural analysis also reveals a β-sheet fiber phase under certain conditions and placements of the KK motif, revealing a complex and sensitive self-assembly pathway. Both solid-state and solution-based electrochemical characterizations show that CCC fibers are electronically conductive. Single-fiber conductive AFM measurement indicates that the solid-state electrical conductivity is comparable with bacterial cytochrome filaments. Solution-deposited fiber films approximately doubled the electroactive surface area of the electrode, confirming their conductivity in aqueous environments. This work establishes a stimuli-responsive peptide sequence element for balancing the order-disorder transitions in peptides to control their self-assembly into highly organized electronically conductive nanofibers.
Collapse
Affiliation(s)
- Adam Grosvirt-Dramen
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Zachary J Urbach
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Paul J Hurst
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Claire E Kwok
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Joseph P Patterson
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
| | - Fengbin Wang
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Allon I Hochbaum
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
- Department of Chemistry, University of California, Irvine, Irvine, California 92697, United States
- Department of Biochemistry and Molecular Biology, University of California, Irvine, Irvine, California 92697, United States
| |
Collapse
|
6
|
Ikeda T, Nojima T, Yamamoto S, Yamada R, Niwa T, Konno H, Taguchi H. Seesaw protein: Design of a protein that adopts interconvertible alternative functional conformations and its dynamics. Proc Natl Acad Sci U S A 2025; 122:e2412117122. [PMID: 39928865 PMCID: PMC11848303 DOI: 10.1073/pnas.2412117122] [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: 06/19/2024] [Accepted: 01/07/2025] [Indexed: 02/12/2025] Open
Abstract
According to classical Anfinsen's dogma, a protein folds into a single unique conformation with minimal Gibbs energy under physiological conditions. However, certain proteins may fold into two or more conformations from single amino acid sequences. Here, we designed a protein that adopts interconvertible alternative functional conformations, termed "seesaw" protein (SSP). An SSP was engineered by fusing GFP lacking the C-terminal β-strand and dihydrofolate reductase (DHFR) lacking the N-terminal β-strand with an overlapping linker, which can be competitively incorporated into either the GFP or the DHFR moiety. In vivo and biochemical analyses, including atomic force microscopy (AFM) imaging, demonstrated that the SSP adopts two alternative conformations, which can be biased by point mutations and ligand binding. The drastic conformational change upon the ligand binding was directly visualized by high-speed AFM. Furthermore, the balance of the seesaw can be reversibly changed depending on buffer conditions. In summary, our design strategy for SSP provides a unique direction for creating artificial proteins with on-off behaviors.
Collapse
Affiliation(s)
- Toma Ikeda
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama226-8501, Japan
| | - Tatsuya Nojima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
| | - Souma Yamamoto
- College of Science and Engineering, School of Biological Science and Technology, Kanazawa University, Kanazawa920-1192, Japan
| | - Ryusei Yamada
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa920-1192, Japan
| | - Tatsuya Niwa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
| | - Hiroki Konno
- World Premier International Research Center Initiative Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa920-1192, Japan
| | - Hideki Taguchi
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama226-8501, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama226-8503, Japan
| |
Collapse
|
7
|
Meng JL, Dong ZX, Chen YR, Lin MH, Liu YC, Roffler SR, Lin WW, Chang CY, Tzou SC, Cheng TL, Huang HC, Li ZQ, Lin YC, Su YC. pH-Responsive Polyethylene Glycol Engagers for Enhanced Brain Delivery of PEGylated Nanomedicine to Treat Glioblastoma. ACS NANO 2025; 19:307-321. [PMID: 39749925 PMCID: PMC11752499 DOI: 10.1021/acsnano.4c05906] [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: 05/04/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The blood-brain barrier (BBB) remains a major obstacle for effective delivery of therapeutics to treat central nervous system (CNS) disorders. Although transferrin receptor (TfR)-mediated transcytosis is widely employed for brain drug delivery, the inefficient release of therapeutic payload hinders their efficacy from crossing the BBB. Here, we developed a pH-responsive anti-polyethylene glycol (PEG) × anti-TfR bispecific antibody (pH-PEG engagerTfR) that can complex with PEGylated nanomedicine at physiological pH to trigger TfR-mediated transcytosis in the brain microvascular endothelial cells, while rapidly dissociating from PEGylated nanomedicine at acidic endosomes for efficient release of PEGylated nanomedicine to cross the BBB. The pH-PEG engagerTfR significantly increased the accumulation of PEGylated nanomedicine in the mouse brain compared to wild-type PEG engagerTfR (WT-PEG engagerTfR). pH-PEG engagerTfR-decorated PEGylated liposomal doxorubicin exhibited an enhanced antitumor effect and extended survival in a human glioblastoma (GBM) orthotopic xenograft mice model. Conditional release of PEGylated nanomedicine during BBB-related receptor-mediated transcytosis by pH-PEG engagerTfR is promising for enhanced brain drug delivery to treat CNS disorders.
Collapse
Affiliation(s)
- Jun-Lun Meng
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zi-Xuan Dong
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yan-Ru Chen
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Meng-Hsuan Lin
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ching Liu
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Steve R. Roffler
- Institute
of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Graduate
Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Wei Lin
- School
of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Yuan Chang
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Shey-Cherng Tzou
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| | - Tian-Lu Cheng
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| | - Hsiao-Chen Huang
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zhi-Qin Li
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yen-Cheng Lin
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Cheng Su
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Hou S, He H, Yang H, Chen C, Wang Q, Wu Z, Li S, Xie J. The receptor binding mechanism of mouse sPLA2 group IIE. Biochem Biophys Res Commun 2025; 742:151103. [PMID: 39672005 DOI: 10.1016/j.bbrc.2024.151103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 12/15/2024]
Abstract
Secreted phospholipase A2s (sPLA2s) participate in physiological function by their enzyme and receptor binding activity. Muscle-type phospholipase A2 receptor (M-type PLA2R) is the sPLA2 binding protein with the highest affinity so far, and also inhibits the enzyme activity of sPLA2. There is species specificity and pH dependence for the binding of M-type PLA2R to sPLA2. Mouse sPLA2 Group IIE (mGIIE) has been verified to have a high affinity for mouse M-type PLA2R (M-type mPLA2R) at the nanomolar scale. For further exploration of the receptor binding mechanism of GIIE, in this study, we use Alphafold Multimer to generate complex models of mGIIE with the M-type mPLA2R ectodomain, wild-type CTLD5 domain of mPLA2R, and three CTLD5 mutants, respectively. mPLA2R-mGIIE models exhibit heterogeneous extended mPLA2R conformations with uncovered sPLA2-binding surface of CTLD5 domain. Complexed models of mGIIE with wild-type and mutated mCTLD5 further confirm that helix α1 of mCTLD5, especially essential residues F838 and W842, interact with the substrate pocket of mGIIE and thus inhibit its enzyme activity. Peptides from helix α1 of mCTLD5 are verified to inhibit the enzymatic activity of mGIIE. This AI-guided research would substantially accelerate our understanding of the functional study of GIIE, and provide the lead-peptide for the further inhibitor design of sPLA2.
Collapse
Affiliation(s)
- Shulin Hou
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| | - Huili He
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Haishan Yang
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Chunrong Chen
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qian Wang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhifang Wu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Sijin Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
9
|
Zhang Y, Zhao Q, Zhang J, Wei S, Tao F, Yang P. Bio-Inspired Adaptive and Responsive Protein-Based Materials. Chempluschem 2024; 89:e202400309. [PMID: 39116292 DOI: 10.1002/cplu.202400309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
In nature, the inherent adaptability and responsiveness of proteins play a crucial role in the survival and reproduction of organisms, enabling them to adjust to ever-changing environments. A comprehensive understanding of protein structure and function is essential for unraveling the complex biological adaptive processes, providing new insights for the design of protein-based materials in advanced fields. Recently, materials derived from proteins with specific properties and functions have been engineered. These protein-based materials, distinguished by their engineered adaptability and responsiveness, range from the nanoscale to the macroscale through meticulous control of protein structure. First, the review introduces the natural adaptability and responsiveness of proteins in organisms, encompassing biological adhesion and the responses of organisms to light, magnetic fields, and temperature. Next, it discusses the achievements in protein-engineered adaptability and adhesion through protein assembly and nanotechnology, emphasizing precise control over protein bioactivity. Finally, the review briefly addresses the application of protein engineering techniques and the self-assembly capabilities of proteins to achieve responsiveness in protein-based materials to humidity, light, magnetism, temperature, and other factors. We hope this review will foster a multidimensional understanding of protein adaptability and responsiveness, thereby advancing the interdisciplinary integration of biomedical science, materials science, and biotechnology.
Collapse
Affiliation(s)
- Yingying Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| | - Qi Zhao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| | - Jingjiao Zhang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| | - Shuo Wei
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| | - Fei Tao
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| | - Peng Yang
- Key Laboratory of Applied Surface and Colloid Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, Shaanxi, 710119, P. R. China
| |
Collapse
|
10
|
McElroy CA, Ihms EC, Kumar Yadav D, Holmquist ML, Wadhwa V, Wysocki VH, Gollnick P, Foster MP. Solution structure, dynamics and tetrahedral assembly of Anti-TRAP, a homo-trimeric triskelion-shaped regulator of tryptophan biosynthesis in Bacillus subtilis. J Struct Biol X 2024; 10:100103. [PMID: 39035014 PMCID: PMC11255114 DOI: 10.1016/j.yjsbx.2024.100103] [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: 11/12/2023] [Revised: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/23/2024] Open
Abstract
Cellular production of tryptophan is metabolically expensive and tightly regulated. The small Bacillus subtilis zinc binding Anti-TRAP protein (AT), which is the product of the yczA/rtpA gene, is upregulated in response to accumulating levels of uncharged tRNATrp through a T-box antitermination mechanism. AT binds to the undecameric axially symmetric ring-shaped protein TRAP (trp RNA Binding Attenuation Protein), thereby preventing it from binding to the trp leader RNA. This reverses the inhibitory effect of TRAP on transcription and translation of the trp operon. AT principally adopts two symmetric oligomeric states, a trimer (AT3) featuring three-fold axial symmetry or a dodecamer (AT12) comprising a tetrahedral assembly of trimers, whereas only the trimeric form binds and inhibits TRAP. We apply native mass spectrometry (nMS) and small-angle x-ray scattering (SAXS), together with analytical ultracentrifugation (AUC) to monitor the pH and concentration-dependent equilibrium between the trimeric and dodecameric structural forms of AT. In addition, we use solution nuclear magnetic resonance (NMR) spectroscopy to determine the solution structure of AT3, while heteronuclear 15N relaxation measurements on both oligomeric forms of AT provide insights into the dynamic properties of binding-active AT3 and binding-inactive AT12, with implications for TRAP binding and inhibition.
Collapse
Affiliation(s)
- Craig A. McElroy
- Ohio State Biochemistry Program, USA
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Elihu C. Ihms
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, USA
| | - Deepak Kumar Yadav
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Melody L. Holmquist
- Ohio State Biochemistry Program, USA
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Vibhuti Wadhwa
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- National Resource for Native MS-Guided Structural Biology, USA
| | - Paul Gollnick
- Department of Biological Sciences, State University of New York, Buffalo, NY 14260, USA
| | - Mark P. Foster
- Ohio State Biochemistry Program, USA
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, USA
| |
Collapse
|
11
|
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
|
12
|
Hatstat AK, Kormos R, Xu V, DeGrado WF. A designed Zn 2+ sensor domain transmits binding information to transmembrane histidine kinases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621206. [PMID: 39553995 PMCID: PMC11565981 DOI: 10.1101/2024.10.30.621206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Generating stimulus-responsive, allosteric signaling de novo is a significant challenge in protein design. In natural systems like bacterial histidine kinases (HKs), signal transduction occurs when ligand binding initiates a signal that is amplified across biological membranes over long distances to induce large-scale rearrangements and phosphorylation relays. Here, we ask whether our understanding of protein design and multi-domain, intramolecular signaling has progressed sufficiently to enable engineering of a HK with tunable de novo components. We generated de novo metal-binding sensor domains and substituted them for the native sensor domain of a transmembrane HK, affording chimeras that transduce signals initiated from a de novo sensor. Signaling depended on the designed sensor's stability and the interdomain linker's phase and length. These results show the usefulness of de novo design to elucidate biochemical mechanisms and principles for design of new signaling systems.
Collapse
|
13
|
Wang S, Favor A, Kibler R, Lubner J, Borst AJ, Coudray N, Redler RL, Chiang HT, Sheffler W, Hsia Y, Li Z, Ekiert DC, Bhabha G, Pozzo LD, Baker D. Bond-centric modular design of protein assemblies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617872. [PMID: 39416012 PMCID: PMC11483063 DOI: 10.1101/2024.10.11.617872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
We describe a modular bond-centric approach to protein nanomaterial design inspired by the rich diversity of chemical structures that can be generated from the small number of atomic valencies and bonding interactions. We design protein building blocks with regular coordination geometries and bonding interactions that enable the assembly of a wide variety of closed and opened nanomaterials using simple geometrical principles. Experimental characterization confirms successful formation of more than twenty multi-component polyhedral protein cages, 2D arrays, and 3D protein lattices, with a high (10-50 %) success rate and electron microscopy data closely matching the corresponding design models. Because of the modularity, individual building blocks can assemble with different partners to generate distinct regular assemblies, resulting in an economy of parts and enabling the construction of reconfigurable systems.
Collapse
Affiliation(s)
- Shunzhi Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Andrew Favor
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Ryan Kibler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Joshua Lubner
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Andrew J. Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Nicolas Coudray
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, Division of Precision Medicine, NYU Grossman School of Medicine, New York, USA
| | - Rachel L. Redler
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Huat Thart Chiang
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - William Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Yang Hsia
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Zhe Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Damian C. Ekiert
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Gira Bhabha
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Lilo D Pozzo
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - 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, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Kephart SM, Hom N, Lee KK. Visualizing intermediate stages of viral membrane fusion by cryo-electron tomography. Trends Biochem Sci 2024; 49:916-931. [PMID: 39054240 PMCID: PMC11455608 DOI: 10.1016/j.tibs.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Protein-mediated membrane fusion is the dynamic process where specialized protein machinery undergoes dramatic conformational changes that drive two membrane bilayers together, leading to lipid mixing and opening of a fusion pore between previously separate membrane-bound compartments. Membrane fusion is an essential stage of enveloped virus entry that results in viral genome delivery into host cells. Recent studies applying cryo-electron microscopy techniques in a time-resolved fashion provide unprecedented glimpses into the interaction of viral fusion proteins and membranes, revealing fusion intermediate states from the initiation of fusion to release of the viral genome. In combination with complementary structural, biophysical, and computation modeling approaches, these advances are shedding new light on the mechanics and dynamics of protein-mediated membrane fusion.
Collapse
Affiliation(s)
- Sally M Kephart
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Nancy Hom
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA; Biological Structure Physics and Design Graduate Program, University of Washington, Seattle, WA, USA.
| |
Collapse
|
15
|
Liu F, Li X, Li Y, Xu S, Guo C, Wang L. Visualization of drug release in a chemo-immunotherapy nanoplatform via ratiometric 19F magnetic resonance imaging. Chem Sci 2024:d4sc03643c. [PMID: 39364076 PMCID: PMC11446317 DOI: 10.1039/d4sc03643c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Visualization of drug release in vivo is crucial for improving therapeutic efficacy and preventing inappropriate medication dosing, yet, challenging. Herein, we report a pH-activated chemo-immunotherapy nanoplatform with visualization of drug release in vivo by ratiometric 19F magnetic resonance imaging (19F MRI). This nanoplatform consists of ultra-small histamine-modified perfluoro-15-crown-5-ether (PFCE) nanodroplets loaded with doxorubicin (Dox), which are packaged in trifluoromethyl-containing metal-organic assemblies via coordination-driven self-assembly. The chemical shifts of two types of 19F atoms in the nanoplatform are significantly different in 19F nuclear magnetic resonance (NMR) spectra, which facilitates the implementation of ratiometric 19F MRI without any signal crosstalk. In an acidic tumor microenvironment, this nanoplatform gradually degrades, which results in a sustained drug release with a real-time change in the ratiometric 19F MRI signal. Therefore, a linear correlation between the Dox release profile and ratiometric 19F MRI signal is established to visualize Dox release. Moreover, the pH-triggered disassembly of the nanoplatform leads to cell pyroptosis, which evokes immunogenic cell death (ICD), resulting in the regression of the primary tumor and inhibition of distal tumor growth. This study provides the proof-of-concept application of ratiometric 19F MRI to visualize drug release in vivo.
Collapse
Affiliation(s)
- Fanqi Liu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| | - Xindi Li
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| | - Yumin Li
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| | - Suying Xu
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| | - Chang Guo
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| | - Leyu Wang
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology Beijing 100029 China
| |
Collapse
|
16
|
Custom protein nanoparticles for targeted drug delivery. Nat Struct Mol Biol 2024; 31:1317-1318. [PMID: 38724719 DOI: 10.1038/s41594-024-01289-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
|
17
|
Yang EC, Divine R, Miranda MC, Borst AJ, Sheffler W, Zhang JZ, Decarreau J, Saragovi A, Abedi M, Goldbach N, Ahlrichs M, Dobbins C, Hand A, Cheng S, Lamb M, Levine PM, Chan S, Skotheim R, Fallas J, Ueda G, Lubner J, Somiya M, Khmelinskaia A, King NP, Baker D. Computational design of non-porous pH-responsive antibody nanoparticles. Nat Struct Mol Biol 2024; 31:1404-1412. [PMID: 38724718 PMCID: PMC11402598 DOI: 10.1038/s41594-024-01288-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/22/2024] [Indexed: 05/21/2024]
Abstract
Programming protein nanomaterials to respond to changes in environmental conditions is a current challenge for protein design and is important for targeted delivery of biologics. Here we describe the design of octahedral non-porous nanoparticles with a targeting antibody on the two-fold symmetry axis, a designed trimer programmed to disassemble below a tunable pH transition point on the three-fold axis, and a designed tetramer on the four-fold symmetry axis. Designed non-covalent interfaces guide cooperative nanoparticle assembly from independently purified components, and a cryo-EM density map closely matches the computational design model. The designed nanoparticles can package protein and nucleic acid payloads, are endocytosed following antibody-mediated targeting of cell surface receptors, and undergo tunable pH-dependent disassembly at pH values ranging between 5.9 and 6.7. The ability to incorporate almost any antibody into a non-porous pH-dependent nanoparticle opens up new routes to antibody-directed targeted delivery.
Collapse
Affiliation(s)
- Erin C Yang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure & Design, University of Washington, Seattle, WA, USA
| | - Robby Divine
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biochemistry, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Marcos C Miranda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Andrew J Borst
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Will Sheffler
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jason Z Zhang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Justin Decarreau
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Amijai Saragovi
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mohamad Abedi
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Nicolas Goldbach
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Technical University of Munich, Munich, Germany
| | - Maggie Ahlrichs
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexis Hand
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Suna Cheng
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mila Lamb
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Paul M Levine
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rebecca Skotheim
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Jorge Fallas
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Joshua Lubner
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Masaharu Somiya
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- SANKEN, Osaka University, Osaka, Japan
| | - Alena Khmelinskaia
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Transdisciplinary Research Area 'Building Blocks of Matter and Fundamental Interactions (TRA Matter)', University of Bonn, Bonn, Germany
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
18
|
Levenson R, Malady B, Lee T, Al Sabeh Y, Gordon MJ, Morse DE. Protein Charge Neutralization Is the Proximate Driver Dynamically Tuning Reflectin Assembly. Int J Mol Sci 2024; 25:8954. [PMID: 39201640 PMCID: PMC11354490 DOI: 10.3390/ijms25168954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Reflectin is a cationic, block copolymeric protein that mediates the dynamic fine-tuning of color and brightness of light reflected from nanostructured Bragg reflectors in iridocyte skin cells of squids. In vivo, the neuronally activated phosphorylation of reflectin triggers its assembly, driving osmotic dehydration of the membrane-bounded Bragg lamellae containing the protein to simultaneously shrink the lamellar thickness and spacing while increasing their refractive index contrast, thus tuning the wavelength and increasing the brightness of reflectance. In vitro, we show that the reduction in repulsive net charge of the purified, recombinant reflectin-either (for the first time) by generalized anionic screening with salt or by pH titration-drives a finely tuned, precisely calibrated increase in the size of the resulting multimeric assemblies. The calculated effects of phosphorylation in vivo are consistent with these effects observed in vitro. The precise proportionality between the assembly size and charge neutralization is enabled by the demonstrated rapid dynamic arrest of multimer growth by a continual, equilibrium tuning of the balance between the protein's Coulombic repulsion and short-range interactive forces. The resulting stability of reflectin assemblies with time ensures a reciprocally precise control of the particle number concentration, encoding a precise calibration between the extent of neuronal signaling, osmotic pressure, and the resulting optical changes. The charge regulation of reflectin assembly precisely fine-tunes a colligative property-based nanostructured biological machine. A physical mechanism is proposed.
Collapse
Affiliation(s)
- Robert Levenson
- Life Sciences, Soka University of America, Aliso Viejo, CA 92656, USA
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106-5100, USA
| | - Brandon Malady
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106-5100, USA
| | - Tyler Lee
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106-5100, USA
| | - Yahya Al Sabeh
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106-5100, USA
| | - Michael J. Gordon
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106-5080, USA
| | - Daniel E. Morse
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106-5100, USA
| |
Collapse
|
19
|
Pillai A, Idris A, Philomin A, Weidle C, Skotheim R, Leung PJY, Broerman A, Demakis C, Borst AJ, Praetorius F, Baker D. De novo design of allosterically switchable protein assemblies. Nature 2024; 632:911-920. [PMID: 39143214 PMCID: PMC11338832 DOI: 10.1038/s41586-024-07813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Allosteric modulation of protein function, wherein the binding of an effector to a protein triggers conformational changes at distant functional sites, plays a central part in the control of metabolism and cell signalling1-3. There has been considerable interest in designing allosteric systems, both to gain insight into the mechanisms underlying such 'action at a distance' modulation and to create synthetic proteins whose functions can be regulated by effectors4-7. However, emulating the subtle conformational changes distributed across many residues, characteristic of natural allosteric proteins, is a significant challenge8,9. Here, inspired by the classic Monod-Wyman-Changeux model of cooperativity10, we investigate the de novo design of allostery through rigid-body coupling of peptide-switchable hinge modules11 to protein interfaces12 that direct the formation of alternative oligomeric states. We find that this approach can be used to generate a wide variety of allosterically switchable systems, including cyclic rings that incorporate or eject subunits in response to peptide binding and dihedral cages that undergo effector-induced disassembly. Size-exclusion chromatography, mass photometry13 and electron microscopy reveal that these designed allosteric protein assemblies closely resemble the design models in both the presence and absence of peptide effectors and can have ligand-binding cooperativity comparable to classic natural systems such as haemoglobin14. Our results indicate that allostery can arise from global coupling of the energetics of protein substructures without optimized side-chain-side-chain allosteric communication pathways and provide a roadmap for generating allosterically triggerable delivery systems, protein nanomachines and cellular feedback control circuitry.
Collapse
Affiliation(s)
- Arvind Pillai
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| | - Abbas Idris
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Annika Philomin
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Connor Weidle
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Rebecca Skotheim
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Philip J Y Leung
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Program in Molecular Engineering, University of Washington, Seattle, WA, USA
| | - Adam Broerman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Cullen Demakis
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure, and Design, University of Washington, Seattle, WA, USA
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Florian Praetorius
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria.
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
20
|
Cross JA, Dawson WM, Shukla SR, Weijman JF, Mantell J, Dodding MP, Woolfson DN. A de novo designed coiled coil-based switch regulates the microtubule motor kinesin-1. Nat Chem Biol 2024; 20:916-923. [PMID: 38849529 PMCID: PMC11213707 DOI: 10.1038/s41589-024-01640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/09/2024] [Indexed: 06/09/2024]
Abstract
Many enzymes are allosterically regulated via conformational change; however, our ability to manipulate these structural changes and control function is limited. Here we install a conformational switch for allosteric activation into the kinesin-1 microtubule motor in vitro and in cells. Kinesin-1 is a heterotetramer that accesses open active and closed autoinhibited states. The equilibrium between these states centers on a flexible elbow within a complex coiled-coil architecture. We target the elbow to engineer a closed state that can be opened with a de novo designed peptide. The alternative states are modeled computationally and confirmed by biophysical measurements and electron microscopy. In cells, peptide-driven activation increases kinesin transport, demonstrating a primary role for conformational switching in regulating motor activity. The designs are enabled by our understanding of ubiquitous coiled-coil structures, opening possibilities for controlling other protein activities.
Collapse
Affiliation(s)
- Jessica A Cross
- School of Biochemistry, University of Bristol, Bristol, UK.
- School of Chemistry, University of Bristol, Bristol, UK.
| | | | - Shivam R Shukla
- School of Biochemistry, University of Bristol, Bristol, UK
- School of Chemistry, University of Bristol, Bristol, UK
| | | | - Judith Mantell
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Mark P Dodding
- School of Biochemistry, University of Bristol, Bristol, UK.
- Bristol BioDesign Institute, University of Bristol, Bristol, UK.
| | - Derek N Woolfson
- School of Biochemistry, University of Bristol, Bristol, UK.
- School of Chemistry, University of Bristol, Bristol, UK.
- Bristol BioDesign Institute, University of Bristol, Bristol, UK.
| |
Collapse
|
21
|
Shen H, Lynch EM, Akkineni S, Watson JL, Decarreau J, Bethel NP, Benna I, Sheffler W, Farrell D, DiMaio F, Derivery E, De Yoreo JJ, Kollman J, Baker D. De novo design of pH-responsive self-assembling helical protein filaments. NATURE NANOTECHNOLOGY 2024; 19:1016-1021. [PMID: 38570702 PMCID: PMC11286511 DOI: 10.1038/s41565-024-01641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024]
Abstract
Biological evolution has led to precise and dynamic nanostructures that reconfigure in response to pH and other environmental conditions. However, designing micrometre-scale protein nanostructures that are environmentally responsive remains a challenge. Here we describe the de novo design of pH-responsive protein filaments built from subunits containing six or nine buried histidine residues that assemble into micrometre-scale, well-ordered fibres at neutral pH. The cryogenic electron microscopy structure of an optimized design is nearly identical to the computational design model for both the subunit internal geometry and the subunit packing into the fibre. Electron, fluorescent and atomic force microscopy characterization reveal a sharp and reversible transition from assembled to disassembled fibres over 0.3 pH units, and rapid fibre disassembly in less than 1 s following a drop in pH. The midpoint of the transition can be tuned by modulating buried histidine-containing hydrogen bond networks. Computational protein design thus provides a route to creating unbound nanomaterials that rapidly respond to small pH changes.
Collapse
Affiliation(s)
- Hao Shen
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
| | - Eric M Lynch
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Susrut Akkineni
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, USA
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joseph L Watson
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Neville P Bethel
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Issa Benna
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - William Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Daniel Farrell
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - James J De Yoreo
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, USA
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Justin Kollman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - 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, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Wilson CJ, de Groot BL, Gapsys V. Resolving coupled pH titrations using alchemical free energy calculations. J Comput Chem 2024; 45:1444-1455. [PMID: 38471815 DOI: 10.1002/jcc.27318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 03/14/2024]
Abstract
In a protein, nearby titratable sites can be coupled: the (de)protonation of one may affect the other. The degree of this interaction depends on several factors and can influence the measured p K a . Here, we derive a formalism based on double free energy differences ( Δ Δ G ) for quantifying the individual site p K a values of coupled residues. As Δ Δ G values can be obtained by means of alchemical free energy calculations, the presented approach allows for a convenient estimation of coupled residue p K a s in practice. We demonstrate that our approach and a previously proposed microscopic p K a formalism, can be combined with alchemical free energy calculations to resolve pH-dependent protein p K a values. Toy models and both, regular and constant-pH molecular dynamics simulations, alongside experimental data, are used to validate this approach. Our results highlight the insights gleaned when coupling and microstate probabilities are analyzed and suggest extensions to more complex enzymatic contexts. Furthermore, we find that naïvely computed p K a values that ignore coupling, can be significantly improved when coupling is accounted for, in some cases reducing the error by half. In short, alchemical free energy methods can resolve the p K a values of both uncoupled and coupled residues.
Collapse
Affiliation(s)
- Carter J Wilson
- Department of Mathematics, The University of Western Ontario, London, Ontario, Canada
- Centre for Advanced Materials and Biomaterials Research (CAMBR), The University of Western Ontario, London, Ontario, Canada
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bert L de Groot
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vytautas Gapsys
- Computational Biomolecular Dynamics Group, Department of Theoretical and Computational Biophysics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Computational Chemistry, Janssen Research & Development, Beerse, Belgium
| |
Collapse
|
23
|
Zhao Y, Zhang B, Gu H, Xu T, Chen Q, Li J, Zhou P, Guan X, He L, Liang Y, Zhang K, Liu S, Shi K. A mutant GH3 family β-glucosidase from Oenococcus oeni exhibits superior adaptation to wine stresses and potential for improving wine aroma and phenolic profiles. Food Microbiol 2024; 119:104458. [PMID: 38225057 DOI: 10.1016/j.fm.2023.104458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/17/2024]
Abstract
In this study, we conducted a comprehensive investigation into a GH3 family β-glucosidase (BGL) from the wild-type strain of Oenococcus oeni and its mutated counterpart from the acid-tolerant mutant strain. Our analysis revealed the mutant BGL's remarkable capacity to adapt to wine-related stress conditions, including heightened tolerance to low pH, elevated ethanol concentrations, and metal ions. Additionally, the mutant BGL exhibited superior hydrolytic activity towards various substrates. Through de novo modeling, we identified specific amino acid mutations responsible for its resilience to low pH and high ethanol environments. In simulated wine conditions, the mutant BGL outperformed both wild-type and commercial BGLs, efficiently releasing terpene and phenolic aglycones from glycosides in wine grapes. These findings not only expand our understanding of O. oeni BGLs but also highlight their potential in enhancing wine production. The mutant BGL's enhanced adaptation to wine stress conditions opens promising avenue for improving wine quality and flavor.
Collapse
Affiliation(s)
- Yuzhu Zhao
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Biying Zhang
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Huawei Gu
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Tongxin Xu
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Qiling Chen
- College of Food Science and Pharmacy, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| | - Jin Li
- COFCO GreatWall Wine, Penglai, Shandong, China
| | | | - Xueqiang Guan
- Shandong Academy of Grape / Shandong Technology Innovation Center of Wine Grape and Wine, Jinan, Shandong, China
| | - Ling He
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanying Liang
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Kekun Zhang
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuwen Liu
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China.
| | - Kan Shi
- College of Enology, College of Life Sciences, College of Horticulture, Shaanxi Engineering Research Center for Viti-Viniculture, Viti-viniculture Engineering Technology Center of State Forestry and Grassland Administration, Heyang Experimental and Demonstrational Stations for Grape, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Life Science Research Core Services, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
24
|
Wang Z, Zhang J, Wang Y, Zhou J, Jiao X, Han M, Zhang X, Hu H, Su R, Zhang Y, Qi W. Overcoming Endosomal Escape Barriers in Gene Drug Delivery Using De Novo Designed pH-Responsive Peptides. ACS NANO 2024; 18:10324-10340. [PMID: 38547369 DOI: 10.1021/acsnano.4c02400] [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: 04/10/2024]
Abstract
A major challenge in using nanocarriers for intracellular drug delivery is their restricted capacity to escape from endosomes into the cytosol. Here, we significantly enhance the drug delivery efficiency by accurately predicting and regulating the transition pH (pH0) of peptides to modulate their endosomal escape capability. Moreover, by inverting the chirality of the peptide carriers, we could further enhance their ability to deliver nucleic acid drugs as well as antitumor drugs. The resulting peptide carriers exhibit versatility in transfecting various cell types with a high efficiency of up to 90% by using siRNA, pDNA, and mRNA. In vivo antitumor experiments demonstrate a tumor growth inhibition of 83.4% using the peptide. This research offers a potent method for the rapid development of peptide vectors with exceptional transfection efficiencies for diverse pathophysiological indications.
Collapse
Affiliation(s)
- Zixuan Wang
- 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
| | - 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
- Beyonpep Biotechnology Limited, Tianjin 300110, P. R. China
| | - Jialin Zhou
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xinhao Jiao
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Mingshan Han
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xuelin Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Hailiang Hu
- 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
| | - Yumiao Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), 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
|
25
|
Gao F, Yang X, Song W. Bioinspired Supramolecular Hydrogel from Design to Applications. SMALL METHODS 2024; 8:e2300753. [PMID: 37599261 DOI: 10.1002/smtd.202300753] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Indexed: 08/22/2023]
Abstract
Nature offers a wealth of opportunities to solve scientific and technological issues based on its unique structures and function. The dynamic non-covalent interaction is considered to be the main base of living functions of creatures including humans, animals, and plants. Supramolecular hydrogels formed by non-covalent bonding interactions has become a unique platform for constructing promising materials for medicine, energy, electronic, and biological substitute. In this review, the self-assemble principle of supramolecular hydrogels is summarized. Next, the stimulation of external environment that triggers the assembly or disassembly of supramolecular hydrogels are recapitulated, including temperature, mechanics, light, pH, ions, etc. The main applications of bioinspired supramolecular hydrogels in terms of bionic objects including humans, animals, and plants are also described. Although so many efforts are done for revealing the synergized mechanism of the function and non-covalent interactions on the supramolecular hydrogel, the complexity and variability between stimulus and non-covalent bonding in the supramolecular system still require impeccable theories. As an outlook, the bioinspired supramolecular hydrogel is just beginning to exhibit its great potential in human life, offering significant opportunities in drug delivery and screening, implantable devices and substitutions, tissue engineering, micro-fluidic devices, and biosensors.
Collapse
Affiliation(s)
- Feng Gao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Xuhao Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| | - Wenlong Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China
| |
Collapse
|
26
|
Akter L, Flechsig H, Marchesi A, Franz CM. Observing Dynamic Conformational Changes within the Coiled-Coil Domain of Different Laminin Isoforms Using High-Speed Atomic Force Microscopy. Int J Mol Sci 2024; 25:1951. [PMID: 38396630 PMCID: PMC10888245 DOI: 10.3390/ijms25041951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Laminins are trimeric glycoproteins with important roles in cell-matrix adhesion and tissue organization. The laminin α, ß, and γ-chains have short N-terminal arms, while their C-termini are connected via a triple coiled-coil domain, giving the laminin molecule a well-characterized cross-shaped morphology as a result. The C-terminus of laminin alpha chains contains additional globular laminin G-like (LG) domains with important roles in mediating cell adhesion. Dynamic conformational changes of different laminin domains have been implicated in regulating laminin function, but so far have not been analyzed at the single-molecule level. High-speed atomic force microscopy (HS-AFM) is a unique tool for visualizing such dynamic conformational changes under physiological conditions at sub-second temporal resolution. After optimizing surface immobilization and imaging conditions, we characterized the ultrastructure of laminin-111 and laminin-332 using HS-AFM timelapse imaging. While laminin-111 features a stable S-shaped coiled-coil domain displaying little conformational rearrangement, laminin-332 coiled-coil domains undergo rapid switching between straight and bent conformations around a defined central molecular hinge. Complementing the experimental AFM data with AlphaFold-based coiled-coil structure prediction enabled us to pinpoint the position of the hinge region, as well as to identify potential molecular rearrangement processes permitting hinge flexibility. Coarse-grained molecular dynamics simulations provide further support for a spatially defined kinking mechanism in the laminin-332 coiled-coil domain. Finally, we observed the dynamic rearrangement of the C-terminal LG domains of laminin-111 and laminin-332, switching them between compact and open conformations. Thus, HS-AFM can directly visualize molecular rearrangement processes within different laminin isoforms and provide dynamic structural insight not available from other microscopy techniques.
Collapse
Affiliation(s)
- Lucky Akter
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| | - Holger Flechsig
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| | - Arin Marchesi
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Via Tronto, 10/A Torrette di Ancona, 60126 Ancona, Italy
| | - Clemens M. Franz
- WPI Nano Life Science Institute, Kanazawa University, Kanazawa 920-1167, Japan; (L.A.); (H.F.); (A.M.)
| |
Collapse
|
27
|
Chu AE, Lu T, Huang PS. Sparks of function by de novo protein design. Nat Biotechnol 2024; 42:203-215. [PMID: 38361073 PMCID: PMC11366440 DOI: 10.1038/s41587-024-02133-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/09/2024] [Indexed: 02/17/2024]
Abstract
Information in proteins flows from sequence to structure to function, with each step causally driven by the preceding one. Protein design is founded on inverting this process: specify a desired function, design a structure executing this function, and find a sequence that folds into this structure. This 'central dogma' underlies nearly all de novo protein-design efforts. Our ability to accomplish these tasks depends on our understanding of protein folding and function and our ability to capture this understanding in computational methods. In recent years, deep learning-derived approaches for efficient and accurate structure modeling and enrichment of successful designs have enabled progression beyond the design of protein structures and towards the design of functional proteins. We examine these advances in the broader context of classical de novo protein design and consider implications for future challenges to come, including fundamental capabilities such as sequence and structure co-design and conformational control considering flexibility, and functional objectives such as antibody and enzyme design.
Collapse
Affiliation(s)
- Alexander E Chu
- Biophysics Program, Stanford University, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
- Google DeepMind, London, UK
| | - Tianyu Lu
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Po-Ssu Huang
- Biophysics Program, Stanford University, Palo Alto, CA, USA.
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
28
|
Kim K, Kim G, Bae J, Song J, Kim H. A pH-Responsive Virus-Like Particle as a Protein Cage for a Targeted Delivery. Adv Healthc Mater 2024; 13:e2302656. [PMID: 37966427 PMCID: PMC11469083 DOI: 10.1002/adhm.202302656] [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: 08/13/2023] [Revised: 11/05/2023] [Indexed: 11/16/2023]
Abstract
A stimuli-responsive protein self-assembly offers promising utility as a protein nanocage for biotechnological and medical applications. Herein, the development of a virus-like particle (VLP) that undergoes a transition between assembly and disassembly under a neutral and acidic pH, respectively, for a targeted delivery is reported. The structure of the bacteriophage P22 coat protein is used for the computational design of coat subunits that self-assemble into a pH-responsive VLP. Subunit designs are generated through iterative computational cycles of histidine substitutions and evaluation of the interaction energies among the subunits under an acidic and neutral pH. The top subunit designs are tested and one that is assembled into a VLP showing the highest pH-dependent structural transition is selected. The cryo-EM structure of the VLP is determined, and the structural basis of a pH-triggered disassembly is delineated. The utility of the designed VLP is exemplified through the targeted delivery of a cytotoxic protein cargo into tumor cells in a pH-dependent manner. These results provide strategies for the development of self-assembling protein architectures with new functionality for diverse applications.
Collapse
Affiliation(s)
- Kwan‐Jip Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Gijeong Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Jin‐Ho Bae
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Ji‐Joon Song
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| | - Hak‐Sung Kim
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐roDaejon34141South Korea
| |
Collapse
|
29
|
Zhou H, Cai Y, Long M, Zheng N, Zhang Z, You C, Hussain A, Xia X. Computer-Aided Reconstruction and Application of Bacillus halodurans S7 Xylanase with Heat and Alkali Resistance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:1213-1227. [PMID: 38183306 DOI: 10.1021/acs.jafc.3c08221] [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: 01/08/2024]
Abstract
β-1,4-Endoxylanase is the most critical hydrolase for xylan degradation during lignocellulosic biomass utilization. However, its poor stability and activity in hot and alkaline environments hinder its widespread application. In this study, BhS7Xyl from Bacillus halodurans S7 was improved using a computer-aided design through isothermal compressibility (βT) perturbation engineering and by combining three thermostability prediction algorithms (ICPE-TPA). The best variant with remarkable improvement in specific activity, heat resistance (70 °C), and alkaline resistance (both pH 9.0 and 70 °C), R69F/E137M/E145L, exhibited a 4.9-fold increase by wild-type in specific activity (1368.6 U/mg), a 39.4-fold increase in temperature half-life (458.1 min), and a 57.6-fold increase in pH half-life (383.1 min). Furthermore, R69F/E137M/E145L was applied to the hydrolysis of agricultural waste (corncob and hardwood pulp) to efficiently obtain a higher yield of high-value xylooligosaccharides. Overall, the ICPE-TPA strategy has the potential to improve the functional performance of enzymes under extreme conditions for the high-value utilization of lignocellulosic biomass.
Collapse
Affiliation(s)
- Huimin Zhou
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Yongchao Cai
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Mengfei Long
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Nan Zheng
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Zehua Zhang
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Cuiping You
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Asif Hussain
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Xiaole Xia
- Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, Jiangsu, China
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300000, China
| |
Collapse
|
30
|
Erba EB, Pastore A. The Complementarity of Nuclear Magnetic Resonance and Native Mass Spectrometry in Probing Protein-Protein Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 3234:109-123. [PMID: 38507203 DOI: 10.1007/978-3-031-52193-5_8] [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: 03/22/2024]
Abstract
Nuclear magnetic resonance (NMR) and native mass spectrometry (MS) are mature physicochemical techniques with long histories and important applications. NMR spectroscopy provides detailed information about the structure, dynamics, interactions, and chemical environment of biomolecules. MS is an effective approach for determining the mass of biomolecules with high accuracy, sensitivity, and speed. The two techniques offer unique advantages and provide solid tools for structural biology. In the present review, we discuss their individual merits in the context of their applications to structural studies in biology with specific focus on protein interactions and evaluate their limitations. We provide specific examples in which these techniques can complement each other, providing new information on the same scientific case. We discuss how the field may develop and what challenges are expected in the future. Overall, the combination of NMR and MS plays an increasingly important role in integrative structural biology, assisting scientists in deciphering the three-dimensional structure of composite macromolecular assemblies.
Collapse
|
31
|
Li Z, Wang S, Nattermann U, Bera AK, Borst AJ, Yaman MY, Bick MJ, Yang EC, Sheffler W, Lee B, Seifert S, Hura GL, Nguyen H, Kang A, Dalal R, Lubner JM, Hsia Y, Haddox H, Courbet A, Dowling Q, Miranda M, Favor A, Etemadi A, Edman NI, Yang W, Weidle C, Sankaran B, Negahdari B, Ross MB, Ginger DS, Baker D. Accurate computational design of three-dimensional protein crystals. NATURE MATERIALS 2023; 22:1556-1563. [PMID: 37845322 DOI: 10.1038/s41563-023-01683-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 09/07/2023] [Indexed: 10/18/2023]
Abstract
Protein crystallization plays a central role in structural biology. Despite this, the process of crystallization remains poorly understood and highly empirical, with crystal contacts, lattice packing arrangements and space group preferences being largely unpredictable. Programming protein crystallization through precisely engineered side-chain-side-chain interactions across protein-protein interfaces is an outstanding challenge. Here we develop a general computational approach for designing three-dimensional protein crystals with prespecified lattice architectures at atomic accuracy that hierarchically constrains the overall number of degrees of freedom of the system. We design three pairs of oligomers that can be individually purified, and upon mixing, spontaneously self-assemble into >100 µm three-dimensional crystals. The structures of these crystals are nearly identical to the computational design models, closely corresponding in both overall architecture and the specific protein-protein interactions. The dimensions of the crystal unit cell can be systematically redesigned while retaining the space group symmetry and overall architecture, and the crystals are extremely porous and highly stable. Our approach enables the computational design of protein crystals with high accuracy, and the designed protein crystals, which have both structural and assembly information encoded in their primary sequences, provide a powerful platform for biological materials engineering.
Collapse
Affiliation(s)
- Zhe Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Shunzhi Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Una Nattermann
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure & Design, University of Washington, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Muammer Y Yaman
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Matthew J Bick
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Erin C Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure & Design, University of Washington, Seattle, WA, USA
| | - William Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Byeongdu Lee
- X-Ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL, USA
| | - Soenke Seifert
- X-Ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, IL, USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Hannah Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Radhika Dalal
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Joshua M Lubner
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Yang Hsia
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Hugh Haddox
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alexis Courbet
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- HHMI, University of Washington, Seattle, WA, USA
| | - Quinton Dowling
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Marcos Miranda
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Andrew Favor
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Ali Etemadi
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Natasha I Edman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Connor Weidle
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Babak Negahdari
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Michael B Ross
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA, USA
| | - David S Ginger
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- HHMI, University of Washington, Seattle, WA, USA.
| |
Collapse
|
32
|
de Souza AS, de Souza RF, Guzzo CR. Cooperative and structural relationships of the trimeric Spike with infectivity and antibody escape of the strains Delta (B.1.617.2) and Omicron (BA.2, BA.5, and BQ.1). J Comput Aided Mol Des 2023; 37:585-606. [PMID: 37792106 DOI: 10.1007/s10822-023-00534-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/11/2023] [Indexed: 10/05/2023]
Abstract
Herein, we conducted simulations of trimeric Spike from several SARS-CoV-2 variants of concern (Delta and Omicron sub-variants BA.2, BA.5, and BQ.1) and investigated the mechanisms by which specific mutations confer resistance to neutralizing antibodies. We observed that the mutations primarily affect the cooperation between protein domains within and between protomers. The substitutions K417N and L452R expand hydrogen bonding interactions, reducing their interaction with neutralizing antibodies. By interacting with nearby residues, the K444T and N460K mutations in the SpikeBQ.1 variant potentially reduces solvent exposure, thereby promoting resistance to antibodies. We also examined the impact of D614G, P681R, and P681H substitutions on Spike protein structure that may be related to infectivity. The D614G substitution influences communication between a glycine residue and neighboring domains, affecting the transition between up- and -down RBD states. The P681R mutation, found in the Delta variant, enhances correlations between protein subunits, while the P681H mutation in Omicron sub-variants weakens long-range interactions that may be associated with reduced fusogenicity. Using a multiple linear regression model, we established a connection between inter-protomer communication and loss of sensitivity to neutralizing antibodies. Our findings underscore the importance of structural communication between protein domains and provide insights into potential mechanisms of immune evasion by SARS-CoV-2. Overall, this study deepens our understanding of how specific mutations impact SARS-CoV-2 infectivity and shed light on how the virus evades the immune system.
Collapse
Affiliation(s)
- Anacleto Silva de Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, Sao Paulo, SP, 5508-900, Brazil.
| | - Robson Francisco de Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, Sao Paulo, SP, 5508-900, Brazil
| | - Cristiane Rodrigues Guzzo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, Sao Paulo, SP, 5508-900, Brazil.
| |
Collapse
|
33
|
Goldbach N, Benna I, Wicky BIM, Croft JT, Carter L, Bera AK, Nguyen H, Kang A, Sankaran B, Yang EC, Lee KK, Baker D. De novo design of monomeric helical bundles for pH-controlled membrane lysis. Protein Sci 2023; 32:e4769. [PMID: 37632837 PMCID: PMC10578055 DOI: 10.1002/pro.4769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/10/2023] [Accepted: 08/24/2023] [Indexed: 08/28/2023]
Abstract
Targeted intracellular delivery via receptor-mediated endocytosis requires the delivered cargo to escape the endosome to prevent lysosomal degradation. This can in principle be achieved by membrane lysis tightly restricted to endosomal membranes upon internalization to avoid general membrane insertion and lysis. Here, we describe the design of small monomeric proteins with buried histidine containing pH-responsive hydrogen bond networks and membrane permeating amphipathic helices. Of the 30 designs that were experimentally tested, all expressed in Escherichia coli, 13 were monomeric with the expected secondary structure, and 4 designs disrupted artificial liposomes in a pH-dependent manner. Mutational analysis showed that the buried histidine hydrogen bond networks mediate pH-responsiveness and control lysis of model membranes within a very narrow range of pH (6.0-5.5) with almost no lysis occurring at neutral pH. These tightly controlled lytic monomers could help mediate endosomal escape in designed targeted delivery platforms.
Collapse
Affiliation(s)
- Nicolas Goldbach
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Molecular Life SciencesTechnical University of MunichMunichGermany
| | - Issa Benna
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
| | - Basile I. M. Wicky
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Jacob T. Croft
- Department of Medicinal ChemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Lauren Carter
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Asim K. Bera
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Hannah Nguyen
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Alex Kang
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
| | - Banumathi Sankaran
- Molecular Biophysics and Integrated BioimagingLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Erin C. Yang
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
- Biological Physics, Structure and Design Graduate ProgramUniversity of WashingtonSeattleWashingtonUSA
| | - Kelly K. Lee
- Department of Medicinal ChemistryUniversity of WashingtonSeattleWashingtonUSA
- Biological Physics, Structure and Design Graduate ProgramUniversity of WashingtonSeattleWashingtonUSA
- Department of MicrobiologyUniversity of WashingtonSeattleWashingtonUSA
| | - David Baker
- Institute for Protein DesignUniversity of WashingtonSeattleWashingtonUSA
- Department of BiochemistryUniversity of WashingtonSeattleWashingtonUSA
- Howard Hughes Medical InstituteUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
34
|
Zhang Z, Zhao Z, Huang K, Liang Z. Acid-resistant enzymes: the acquisition strategies and applications. Appl Microbiol Biotechnol 2023; 107:6163-6178. [PMID: 37615723 DOI: 10.1007/s00253-023-12702-1] [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: 06/05/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
Enzymes have promising applications in chemicals, food, pharmaceuticals, and other variety products because of their high efficiency, specificity, and environmentally friendly properties. However, due to the complexity of raw materials, pH, temperature, solvents, etc., the application range of enzymes is greatly limited in the industry. Protein engineering and enzyme immobilization are classical strategies to overcome the limitations of industrial applications. Although the pH tendency of enzymes has been extensively researched, the mechanism underlying enzyme acid resistance is unclear, and a less practical strategy for altering the pH propensity of enzymes has been suggested. This review proposes that the optimum pH of enzyme is determined by the pKa values of active center ionizable amino acid residues. Three levels of acquiring acid-resistant enzymes are summarized: mining from extreme environments and enzyme databases, modification with protein engineering and enzyme microenvironment engineering, and de novo synthesis. The industrial applications of acid-resistant enzymes in chemicals, food, and pharmaceuticals are also summarized. KEY POINTS: • The mechanism of enzyme acid resistance is fundamentally determined. • The three aspects of the method for acquiring acid-resistant enzymes are summarized. • Computer-aided strategies and artificial intelligence are used to obtain acid-resistant enzymes.
Collapse
Affiliation(s)
- Zhenzhen Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zitong Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- The Supervision, Inspection and Testing Center of Genetically Modified Organisms, Ministry of Agriculture, Beijing, China
- Beijing Laboratory for Food Quality and Safety, China Agricultural University, Beijing, China
| | - Zhihong Liang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
- The Supervision, Inspection and Testing Center of Genetically Modified Organisms, Ministry of Agriculture, Beijing, China.
- Beijing Laboratory for Food Quality and Safety, China Agricultural University, Beijing, China.
| |
Collapse
|
35
|
Zhao Y, Jiang H, Yu J, Wang L, Du J. Engineered Histidine-Rich Peptides Enhance Endosomal Escape for Antibody-Targeted Intracellular Delivery of Functional Proteins. Angew Chem Int Ed Engl 2023; 62:e202304692. [PMID: 37283024 DOI: 10.1002/anie.202304692] [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: 04/03/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/08/2023]
Abstract
Currently, the clinical application of protein/peptide therapeutics is mainly limited to the modulation of diseases in extracellular spaces. Intracellular targets are hardly accessed, owing largely to the endosomal entrapment of internalized proteins/peptides. Here, we report a strategy to design and construct peptides that enable endosome-to-cytosol delivery based on an extension of the "histidine switch" principle. By substituting the Arg/Lys residues in cationic cell-penetrating peptides (CPPs) with histidine, we obtained peptides with pH-dependent membrane-perturbation activity. These peptides do not randomly penetrate cells like CPPs, but imitate the endosomal escape of CPPs following cellular uptake. Working with one such 16-residue peptide (hsLMWP) with high endosomal escape capacity, we engineered modular fusion proteins and achieved antibody-targeted delivery of diverse protein cargoes-including the pro-apoptotic protein BID (BH3-interacting domain death agonist) and Cre recombinase-into the cytosol of multiple cancer cell types. After extensive in vitro testing, an in vivo analysis with xenograft mice ultimately demonstrated that a trastuzumab-hsLMWP-BID fusion conferred strong anti-tumor efficacy without apparent side effects. Notably, our fusion protein features a modular design, allowing flexible applications for any antibody/cargo combination of choice. Therefore, the potential applications extend throughout life science and biomedicine, including gene editing, cancer treatment, and immunotherapy.
Collapse
Affiliation(s)
- Yan Zhao
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haolin Jiang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Academy for Advanced Interdisciplinary Studies (AAIS), Peking University-Tsinghua University-National Institute Biological Sciences (PTN) Joint Graduate Program, Peking University, Beijing, 100871, China
| | - Jiazhen Yu
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Luyao Wang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Juanjuan Du
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
36
|
de March M, Hickey N, Geremia S. Analysis of the crystal structure of a parallel three-stranded coiled coil. Proteins 2023; 91:1254-1260. [PMID: 37501532 DOI: 10.1002/prot.26557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/26/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Here, we present the crystal structure of the synthetic peptide KE1, which contains four K-coil heptads separated in the middle by the QFLMLMF heptad. The structure determination reveals the presence of a canonical parallel three stranded coiled coil. The geometric characteristics of this structure are compared with other coiled coils with the same topology. Furthermore, for this topology, the analysis of the propensity of the single amino acid to occupy a specific position in the heptad sequence is reported. A number of viral proteins use specialized coiled coil tail needles to inject their genetic material into the host cells. The simplicity and regularity of the coiled coil arrangement made it an attractive system for de novo design of key molecules in drug delivery systems, vaccines, and therapeutics.
Collapse
Affiliation(s)
- Matteo de March
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia
| | - Neal Hickey
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Silvano Geremia
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
37
|
Gassner C, Vongsvivut J, Ng SH, Ryu M, Tobin MJ, Juodkazis S, Morikawa J, Wood BR. Linearly Polarized Infrared Spectroscopy for the Analysis of Biological Materials. APPLIED SPECTROSCOPY 2023; 77:977-1008. [PMID: 37464791 DOI: 10.1177/00037028231180233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The analysis of biological samples with polarized infrared spectroscopy (p-IR) has long been a widely practiced method for the determination of sample orientation and structural properties. In contrast to earlier works, which employed this method to investigate the fundamental chemistry of biological systems, recent interests are moving toward "real-world" applications for the evaluation and diagnosis of pathological states. This focal point review provides an up-to-date synopsis of the knowledge of biological materials garnered through linearly p-IR on biomolecules, cells, and tissues. An overview of the theory with special consideration to biological samples is provided. Different modalities which can be employed along with their capabilities and limitations are outlined. Furthermore, an in-depth discussion of factors regarding sample preparation, sample properties, and instrumentation, which can affect p-IR analysis is provided. Additionally, attention is drawn to the potential impacts of analysis of biological samples with inherently polarized light sources, such as synchrotron light and quantum cascade lasers. The vast applications of p-IR for the determination of the structure and orientation of biological samples are given. In conclusion, with considerations to emerging instrumentation, findings by other techniques, and the shift of focus toward clinical applications, we speculate on the future directions of this methodology.
Collapse
Affiliation(s)
- Callum Gassner
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Australia
| | - Jitraporn Vongsvivut
- Infrared Microspectroscopy (IRM) Beamline, ANSTO-Australian Synchrotron, Clayton, Australia
| | - Soon Hock Ng
- Optical Sciences Centre and ARC Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Science, Swinburne University of Technology, Hawthorn, Australia
| | - Meguya Ryu
- National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Mark J Tobin
- Infrared Microspectroscopy (IRM) Beamline, ANSTO-Australian Synchrotron, Clayton, Australia
| | - Saulius Juodkazis
- Optical Sciences Centre and ARC Training Centre in Surface Engineering for Advanced Materials (SEAM), School of Science, Swinburne University of Technology, Hawthorn, Australia
| | - Junko Morikawa
- School of Materials and Chemical Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Bayden R Wood
- Centre for Biospectroscopy, School of Chemistry, Monash University, Clayton, Australia
| |
Collapse
|
38
|
McElroy C, Ihms E, Yadav DK, Holmquist M, Wadwha V, Wysocki V, Gollnick P, Foster M. Solution structure, dynamics and tetrahedral assembly of Anti-TRAP, a homo-trimeric triskelion-shaped regulator of tryptophan biosynthesis in Bacillus subtilis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547145. [PMID: 37425951 PMCID: PMC10327191 DOI: 10.1101/2023.06.29.547145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Cellular production of tryptophan is metabolically expensive and tightly regulated. The small Bacillus subtilis zinc binding Anti-TRAP protein (AT), which is the product of the yczA/rtpA gene, is upregulated in response to accumulating levels of uncharged tRNATrp through a T-box antitermination mechanism. AT binds to the undecameric ring-shaped protein TRAP (trp RNA Binding Attenuation Protein), thereby preventing it from binding to the trp leader RNA. This reverses the inhibitory effect of TRAP on transcription and translation of the trp operon. AT principally adopts two symmetric oligomeric states, a trimer (AT3) featuring a three-helix bundle, or a dodecamer (AT12) comprising a tetrahedral assembly of trimers, whereas only the trimeric form has been shown to bind and inhibit TRAP. We demonstrate the utility of native mass spectrometry (nMS) and small-angle x-ray scattering (SAXS), together with analytical ultracentrifugation (AUC) for monitoring the pH and concentration-dependent equilibrium between the trimeric and dodecameric structural forms of AT. In addition, we report the use of solution nuclear magnetic resonance (NMR) spectroscopy to determine the solution structure of AT3, while heteronuclear 15N relaxation measurements on both oligomeric forms of AT provide insights into the dynamic properties of binding-active AT3 and binding-inactive AT12, with implications for TRAP inhibition.
Collapse
Affiliation(s)
- Craig McElroy
- Ohio State Biochemistry Program
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
| | - Elihu Ihms
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- Biophysics Program
| | - Deepak Kumar Yadav
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
| | - Melody Holmquist
- Ohio State Biochemistry Program
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
| | - Vibhuti Wadwha
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
| | - Vicki Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- National Resource for Native MS-Guided Structural Biology
| | - Paul Gollnick
- Department of Biological Sciences, State University of New York, Buffalo NY 14260
| | - Mark Foster
- Ohio State Biochemistry Program
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- Biophysics Program
| |
Collapse
|
39
|
Ennist N, Wang S, Kennedy M, Curti M, Sutherland G, Vasilev C, Redler R, Maffeis V, Shareef S, Sica A, Hua A, Deshmukh A, Moyer A, Hicks D, Swartz A, Cacho R, Novy N, Bera A, Kang A, Sankaran B, Johnson M, Reppert M, Ekiert D, Bhabha G, Stewart L, Caram J, Stoddard B, Romero E, Hunter CN, Baker D. De novo design of energy transfer proteins housing excitonically coupled chlorophyll special pairs. RESEARCH SQUARE 2023:rs.3.rs-2736786. [PMID: 37131790 PMCID: PMC10153362 DOI: 10.21203/rs.3.rs-2736786/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Natural photosystems couple light harvesting to charge separation using a "special pair" of chlorophyll molecules that accepts excitation energy from the antenna and initiates an electron-transfer cascade. To investigate the photophysics of special pairs independent of complexities of native photosynthetic proteins, and as a first step towards synthetic photosystems for new energy conversion technologies, we designed C2-symmetric proteins that precisely position chlorophyll dimers. X-ray crystallography shows that one designed protein binds two chlorophylls in a binding orientation matching native special pairs, while a second positions them in a previously unseen geometry. Spectroscopy reveals excitonic coupling, and fluorescence lifetime imaging demonstrates energy transfer. We designed special pair proteins to assemble into 24-chlorophyll octahedral nanocages; the design model and cryo-EM structure are nearly identical. The design accuracy and energy transfer function of these special pair proteins suggest that de novo design of artificial photosynthetic systems is within reach of current computational methods.
Collapse
Affiliation(s)
| | | | | | - Mariano Curti
- Institute of Chemical Research of Catalonia (ICIQ-CERCA)
| | | | | | | | | | - Saeed Shareef
- Institute of Chemical Research of Catalonia (ICIQ-CERCA)
| | | | - Ash Hua
- University of California, Los Angeles
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Yang EC, Divine R, Miranda MC, Borst AJ, Sheffler W, Zhang JZ, Decarreau J, Saragovi A, Abedi M, Goldbach N, Ahlrichs M, Dobbins C, Hand A, Cheng S, Lamb M, Levine PM, Chan S, Skotheim R, Fallas J, Ueda G, Lubner J, Somiya M, Khmelinskaia A, King NP, Baker D. Computational design of non-porous, pH-responsive antibody nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537263. [PMID: 37131615 PMCID: PMC10153164 DOI: 10.1101/2023.04.17.537263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Programming protein nanomaterials to respond to changes in environmental conditions is a current challenge for protein design and important for targeted delivery of biologics. We describe the design of octahedral non-porous nanoparticles with the three symmetry axes (four-fold, three-fold, and two-fold) occupied by three distinct protein homooligomers: a de novo designed tetramer, an antibody of interest, and a designed trimer programmed to disassemble below a tunable pH transition point. The nanoparticles assemble cooperatively from independently purified components, and a cryo-EM density map reveals that the structure is very close to the computational design model. The designed nanoparticles can package a variety of molecular payloads, are endocytosed following antibody-mediated targeting of cell surface receptors, and undergo tunable pH-dependent disassembly at pH values ranging between to 5.9-6.7. To our knowledge, these are the first designed nanoparticles with more than two structural components and with finely tunable environmental sensitivity, and they provide new routes to antibody-directed targeted delivery.
Collapse
Affiliation(s)
- Erin C Yang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure & Design, University of Washington, Seattle, WA, USA
| | - Robby Divine
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biochemistry, University of Washington, Seattle, WA, USA
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Marcos C Miranda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Andrew J Borst
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Will Sheffler
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jason Z Zhang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Justin Decarreau
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Amijai Saragovi
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mohamad Abedi
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Nicolas Goldbach
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Technical University of Munich, Munich, Germany
| | - Maggie Ahlrichs
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexis Hand
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Suna Cheng
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mila Lamb
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Paul M Levine
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rebecca Skotheim
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Jorge Fallas
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Joshua Lubner
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Masaharu Somiya
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- SANKEN, Osaka University, Osaka, Japan
| | - Alena Khmelinskaia
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Transdisciplinary Research Area "Building Blocks of Matter and Fundamental Interactions (TRA Matter)", University of Bonn, Bonn, Germany
- Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
41
|
Yu W, Wei C, Zhang K, Zhang J, Ge Z, Liang X, Guiver MD, Ge X, Wu L, Xu T. Host-Guest Recognition Boosts Biomimetic Mono/Multivalent Cation Separation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:5861-5871. [PMID: 36988386 DOI: 10.1021/acs.est.2c09733] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Biomimetic ion permselective membranes with ultrahigh ion permeability and selectivity represent a research frontier in ion separation, yet the successful fabrication of such membranes remains a formidable challenge. Here, we demonstrate a 4-sulfocalix[4]arene (4-SCA)-modified graphene oxide (GO) membrane that shows extraordinary performance in separating mono-from multivalent cations, as well as having reversible pH-responsiveness. The resulting 4-SCA-modified GO (SCA-GO) membrane preferentially transports potassium ions (K+) over radionuclide cations (Co2+, UO22+, La3+, Eu3+, and Th4+). The ion selectivities are an order of magnitude higher than that of the unmodified GO membrane. Theoretical calculations and experimental investigations demonstrate that the much-improved ion selectivity arises from the specific recognition between 4-SCA and radionuclide cations. The transport of multivalent radionuclides is impeded by a binding-obstructing mechanism from the host-guest interactions. Interestingly, the host-guest interactions are responsive to the protonation/deprotonation transformation of the 4-SCA. Therefore, the SCA-GO membrane mimics pH-regulated ion selective behavior found in biological ion channels. Our strategy of designing a biomimetic permselective GO membrane may allow efficient nuclear wastewater treatment and, more importantly, deepen our understanding of biomimetic ion transport mechanisms.
Collapse
Affiliation(s)
- Weisheng Yu
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Chengpeng Wei
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Kaiyu Zhang
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Jianjun Zhang
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Zijuan Ge
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Xian Liang
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Michael D Guiver
- State Key Laboratory of Engines, School of Mechanical Engineering, Tianjin University, Tianjin 300072, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xiaolin Ge
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Liang Wu
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| | - Tongwen Xu
- Anhui Engineering Laboratory of Functional Membrane Materials and Technology, Collaborative Innovation Centre of Chemistry for Energy Materials, School of Chemistry and Material Science, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
42
|
Bhoyar S, Foster M, Oh YH, Xu X, Traylor SJ, Guo J, Ghose S, Lenhoff AM. Engineering protein A ligands to mitigate antibody loss during high-pH washes in protein A chromatography. J Chromatogr A 2023; 1696:463962. [PMID: 37043977 DOI: 10.1016/j.chroma.2023.463962] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/05/2023]
Abstract
Protein A chromatography is a workhorse in monoclonal antibody (mAb) manufacture since it provides effective separation of mAbs from impurities such as host-cell proteins (HCPs) in a single capture step. HCP clearance can be aided by the inclusion of a wash step prior to low-pH elution. Although high-pH washes can be effective in removing additional HCPs from the loaded column, they may also contribute to a reduced mAb yield. In this work we show that this yield loss is reflected in a pH-dependent variation of the equilibrium binding capacity of the protein A resin, which is also observed for the capacity of the Fc fragments alone and therefore not a result of steric interactions involving the Fab fragments in the intact mAbs. We therefore hypothesized that the high-pH wash loss was due to protonation or deprotonation of ionizable residues on the protein A ligand. To evaluate this, we applied a rational protein engineering approach to the Z domain (the Fc-binding component of most commercial protein A ligands) and expressed engineered mutants in E. coli. Biolayer interferometry and affinity chromatography experiments showed that some of the Z domain mutants were able to mitigate wash loss at high pH while maintaining similar binding characteristics at neutral pH. These experiments enabled elucidation of the roles of specific interactions in the Z domain - Fc complex, but more importantly offer a route to ameliorating the disadvantages of high-pH washes in protein A chromatography.
Collapse
|
43
|
Nadendla K, Simpson GG, Becher J, Journeaux T, Cabeza-Cabrerizo M, Bernardes GJL. Strategies for Conditional Regulation of Proteins. JACS AU 2023; 3:344-357. [PMID: 36873677 PMCID: PMC9975842 DOI: 10.1021/jacsau.2c00654] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 06/18/2023]
Abstract
Design of the next-generation of therapeutics, biosensors, and molecular tools for basic research requires that we bring protein activity under control. Each protein has unique properties, and therefore, it is critical to tailor the current techniques to develop new regulatory methods and regulate new proteins of interest (POIs). This perspective gives an overview of the widely used stimuli and synthetic and natural methods for conditional regulation of proteins.
Collapse
Affiliation(s)
- Karthik Nadendla
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
| | - Grant G. Simpson
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
| | - Julie Becher
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
| | - Toby Journeaux
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
| | - Mar Cabeza-Cabrerizo
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
| | - Gonçalo J. L. Bernardes
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, CB2 1EW, Cambridge, U.K.
- Instituto
de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
44
|
Jin Y, Mandal PK, Wu J, Böcher N, Huc I, Otto S. (Re-)Directing Oligomerization of a Single Building Block into Two Specific Dynamic Covalent Foldamers through pH. J Am Chem Soc 2023; 145:2822-2829. [PMID: 36705469 PMCID: PMC9912251 DOI: 10.1021/jacs.2c09325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Dynamic foldamers are synthetic folded molecules which can change their conformation in response to an external stimulus and are currently at the forefront of foldamer chemistry. However, constitutionally dynamic foldamers, which can change not only their conformation but also their molecular constitution in response to their environment, are without precedent. We now report a size- and shape-switching small dynamic covalent foldamer network which responds to changes in pH. Specifically, acidic conditions direct the oligomerization of a dipeptide-based building block into a 16-subunit macrocycle with well-defined conformation and with high selectivity. At higher pH the same building block yields another cyclic foldamer with a smaller ring size (9mer). The two foldamers readily and repeatedly interconvert upon adjustment of the pH of the solution. We have previously shown that addition of a template can direct oligomerization of the same building block to yet other rings sizes (including a 12mer and a 13mer, accompanied by a minor amount of 14mer). This brings the total number of discrete foldamers that can be accessed from a single building block to five. For a single building block system to exhibit such highly diverse structure space is unique and sets this system of foldamers apart from proteins. Furthermore, the emergence of constitutional dynamicity opens up new avenues to foldamers with adaptive behavior.
Collapse
Affiliation(s)
- Yulong Jin
- Beijing
National Laboratory for Molecular Sciences, CAS Key Laboratory of
Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, 100190 Beijing, China,Centre
for Systems Chemistry, Stratingh Institute, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Pradeep K. Mandal
- Department
of Pharmacy and Center for Integrated Protein Science, Ludwig-Maximilians Universität, 81377 Munich, Germany
| | - Juntian Wu
- Centre
for Systems Chemistry, Stratingh Institute, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Niklas Böcher
- Department
of Pharmacy and Center for Integrated Protein Science, Ludwig-Maximilians Universität, 81377 Munich, Germany
| | - Ivan Huc
- Department
of Pharmacy and Center for Integrated Protein Science, Ludwig-Maximilians Universität, 81377 Munich, Germany,
| | - Sijbren Otto
- Centre
for Systems Chemistry, Stratingh Institute, Nijenborgh 4, 9747 AG Groningen, The Netherlands,
| |
Collapse
|
45
|
A double-switch pHLIP system enables selective enrichment of circulating tumor microenvironment-derived extracellular vesicles. Proc Natl Acad Sci U S A 2023; 120:e2214912120. [PMID: 36595702 PMCID: PMC9926244 DOI: 10.1073/pnas.2214912120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Circulating tumor microenvironment-derived extracellular vesicles (cTME-EVs) are gaining considerable traction in cancer research and liquid biopsy. However, the study of cTME-EVs is largely limited by the dearth of a general isolation technique to selectively enrich cTME-EVs from biological fluids for downstream analysis. In this work, we broke through this dilemma by presenting a double-switch pH-low insertion peptide (D-S pHLIP) system to exclusively harvest cTME-EVs from the blood serum of tumor mouse models. This D-S pHLIP system consists of a highly sensitive pH-driven conformational switch (pKa ≈ 6.8) that allows specific installation of D-S pHLIP on the EV membranes in TME (pH 6.5 to 6.8) and a unique hook-like switch to "lock" the peptide securely on the cTME-EVs during the systemic circulation. The D-S pHLIP-anchored cTME-EVs were magnetically enriched and then analyzed with high-resolution messenger RNA sequencing, by which more than 18 times the number of TME-related differentially expressed genes and 10 times the number of hub genes were identified, compared with those achieved by the gold-standard ultracentrifugation. This work could revolutionize basic TME research as well as clinical liquid biopsy for cancer.
Collapse
|
46
|
Histidine network regulates the structure-stability features of T7 endolysin native and partially folded conformations. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.121118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
47
|
Olshefsky A, Richardson C, Pun SH, King NP. Engineering Self-Assembling Protein Nanoparticles for Therapeutic Delivery. Bioconjug Chem 2022; 33:2018-2034. [PMID: 35487503 PMCID: PMC9673152 DOI: 10.1021/acs.bioconjchem.2c00030] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Despite remarkable advances over the past several decades, many therapeutic nanomaterials fail to overcome major in vivo delivery barriers. Controlling immunogenicity, optimizing biodistribution, and engineering environmental responsiveness are key outstanding delivery problems for most nanotherapeutics. However, notable exceptions exist including some lipid and polymeric nanoparticles, some virus-based nanoparticles, and nanoparticle vaccines where immunogenicity is desired. Self-assembling protein nanoparticles offer a powerful blend of modularity and precise designability to the field, and have the potential to solve many of the major barriers to delivery. In this review, we provide a brief overview of key designable features of protein nanoparticles and their implications for therapeutic delivery applications. We anticipate that protein nanoparticles will rapidly grow in their prevalence and impact as clinically relevant delivery platforms.
Collapse
Affiliation(s)
- Audrey Olshefsky
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
| | - Christian Richardson
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
| | - Suzie H. Pun
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Molecular
Engineering and Sciences Institute, University
of Washington, Seattle, Washington 98195, United States
| | - Neil P. King
- Institute
for Protein Design, University of Washington, Seattle, Washington 98195, United States
- Department
of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
48
|
Abstract
Neurodegenerative diseases are characterized by the pathologic accumulation of aggregated proteins. Known as amyloid, these fibrillar aggregates include proteins such as tau and amyloid-β (Aβ) in Alzheimer's disease (AD) and alpha-synuclein (αSyn) in Parkinson's disease (PD). The development and spread of amyloid fibrils within the brain correlates with disease onset and progression, and inhibiting amyloid formation is a possible route toward therapeutic development. Recent advances have enabled the determination of amyloid fibril structures to atomic-level resolution, improving the possibility of structure-based inhibitor design. In this work, we use these amyloid structures to design inhibitors that bind to the ends of fibrils, "capping" them so as to prevent further growth. Using de novo protein design, we develop a library of miniprotein inhibitors of 35 to 48 residues that target the amyloid structures of tau, Aβ, and αSyn. Biophysical characterization of top in silico designed inhibitors shows they form stable folds, have no sequence similarity to naturally occurring proteins, and specifically prevent the aggregation of their targeted amyloid-prone proteins in vitro. The inhibitors also prevent the seeded aggregation and toxicity of fibrils in cells. In vivo evaluation reveals their ability to reduce aggregation and rescue motor deficits in Caenorhabditis elegans models of PD and AD.
Collapse
|
49
|
Shi WT, Zhang B, Li ML, Liu KH, Jiao J, Tian CF. The convergent xenogeneic silencer MucR predisposes α-proteobacteria to integrate AT-rich symbiosis genes. Nucleic Acids Res 2022; 50:8580-8598. [PMID: 36007892 PMCID: PMC9410896 DOI: 10.1093/nar/gkac664] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/11/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Bacterial adaptation is largely shaped by horizontal gene transfer, xenogeneic silencing mediated by lineage-specific DNA bridgers (H-NS, Lsr2, MvaT and Rok), and various anti-silencing mechanisms. No xenogeneic silencing DNA bridger is known for α-proteobacteria, from which mitochondria evolved. By investigating α-proteobacterium Sinorhizobium fredii, a facultative legume microsymbiont, here we report the conserved zinc-finger bearing MucR as a novel xenogeneic silencing DNA bridger. Self-association mediated by its N-terminal domain (NTD) is required for DNA–MucR–DNA bridging complex formation, maximizing MucR stability, transcriptional silencing, and efficient symbiosis in legume nodules. Essential roles of NTD, CTD (C-terminal DNA-binding domain), or full-length MucR in symbiosis can be replaced by non-homologous NTD, CTD, or full-length protein of H-NS from γ-proteobacterium Escherichia coli, while NTD rather than CTD of Lsr2 from Gram-positive Mycobacterium tuberculosis can replace the corresponding domain of MucR in symbiosis. Chromatin immunoprecipitation sequencing reveals similar recruitment profiles of H-NS, MucR and various functional chimeric xenogeneic silencers across the multipartite genome of S. fredii, i.e. preferring AT-rich genomic islands and symbiosis plasmid with key symbiosis genes as shared targets. Collectively, the convergently evolved DNA bridger MucR predisposed α-proteobacteria to integrate AT-rich foreign DNA including symbiosis genes, horizontal transfer of which is strongly selected in nature.
Collapse
Affiliation(s)
- Wen-Tao Shi
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| | - Biliang Zhang
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| | - Meng-Lin Li
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| | - Ke-Han Liu
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| | - Jian Jiao
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| | - Chang-Fu Tian
- State Key Laboratory of Agrobiotechnology, and College of Biological Sciences, China Agricultural University , Beijing , China
- MOA Key Laboratory of Soil Microbiology, and Rhizobium Research Center, China Agricultural University , Beijing , China
| |
Collapse
|
50
|
Tian JH, Hu XY, Hu ZY, Tian HW, Li JJ, Pan YC, Li HB, Guo DS. A facile way to construct sensor array library via supramolecular chemistry for discriminating complex systems. Nat Commun 2022; 13:4293. [PMID: 35879312 PMCID: PMC9314354 DOI: 10.1038/s41467-022-31986-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022] Open
Abstract
Differential sensing, which discriminates analytes via pattern recognition by sensor arrays, plays an important role in our understanding of many chemical and biological systems. However, it remains challenging to develop new methods to build a sensor unit library without incurring a high workload of synthesis. Herein, we propose a supramolecular approach to construct a sensor unit library by taking full advantage of recognition and assembly. Ten sensor arrays are developed by replacing the building block combinations, adjusting the ratio between system components, and changing the environment. Using proteins as model analytes, we examine the discriminative abilities of these supramolecular sensor arrays. Then the practical applicability for discriminating complex analytes is further demonstrated using honey as an example. This sensor array construction strategy is simple, tunable, and capable of developing many sensor units with as few syntheses as possible.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Zong-Ying Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Han-Wen Tian
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Juan-Juan Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Yu-Chen Pan
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Hua-Bin Li
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, 300071, China.
| |
Collapse
|