1
|
Li Y, Duan Z, Li Z, Xue W. Data and AI-driven synthetic binding protein discovery. Trends Pharmacol Sci 2025; 46:132-144. [PMID: 39755458 DOI: 10.1016/j.tips.2024.12.002] [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: 10/12/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025]
Abstract
Synthetic binding proteins (SBPs) are a class of protein binders that are artificially created and do not exist naturally. Their broad applications in tackling challenges of research, diagnostics, and therapeutics have garnered significant interest. Traditional protein engineering is pivotal to the discovery of SBPs. Recently, this discovery has been significantly accelerated by computational approaches, such as molecular modeling and artificial intelligence (AI). Furthermore, while numerous bioinformatics databases offer a wealth of resources that fuel SBP discovery, the full potential of these data has not yet been fully exploited. In this review, we present a comprehensive overview of SBP data ecosystem and methodologies in SBP discovery, highlighting the critical role of high-quality data and AI technologies in accelerating the discovery of innovative SBPs with promising applications in pharmacological sciences.
Collapse
Affiliation(s)
- Yanlin Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zixin Duan
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Zhenwen Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China; Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Park, Chongqing 401329, China.
| |
Collapse
|
2
|
Wéber E, Ábrányi-Balogh P, Nagymihály B, Menyhárd DK, Péczka N, Gadanecz M, Schlosser G, Orgován Z, Bogár F, Bajusz D, Kecskeméti G, Szabó Z, Bartus É, Tököli A, Tóth GK, Szalai TV, Takács T, de Araujo E, Buday L, Perczel A, Martinek TA, Keserű GM. Target-Templated Construction of Functional Proteomimetics Using Photo-Foldamer Libraries. Angew Chem Int Ed Engl 2025; 64:e202410435. [PMID: 39329252 DOI: 10.1002/anie.202410435] [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: 06/03/2024] [Revised: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 09/28/2024]
Abstract
Current methods for proteomimetic engineering rely on structure-based design. Here we describe a design strategy that allows the construction of proteomimetics against challenging targets without a priori characterization of the target surface. Our approach employs (i) a 100-membered photoreactive foldamer library, the members of which act as local surface mimetics, and (ii) the subsequent affinity maturation of the primary hits using systems chemistry. Two surface-oriented proteinogenic side chains drove the interactions between the short helical foldamer fragments and the proteins. Diazirine-based photo-crosslinking was applied to sensitively detect and localize binding even to shallow and dynamic patches on representatively difficult targets. Photo-foldamers identified functionally relevant protein interfaces, allosteric and previously unexplored targetable regions on the surface of STAT3 and an oncogenic K-Ras variant. Target-templated dynamic linking of foldamer hits resulted in two orders of magnitude affinity improvement in a single step. The dimeric K-Ras ligand mimicked protein-like catalytic functions. The photo-foldamer approach thus enables the highly efficient mapping of protein-protein interaction sites and provides a viable starting point for proteomimetic ligand development without a priori structural hypotheses.
Collapse
Affiliation(s)
- Edit Wéber
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Bence Nagymihály
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Dóra K Menyhárd
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Nikolett Péczka
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Márton Gadanecz
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, Eötvös Loránd University, Egyetem tér 1-3, H-1053, Budapest, Hungary
| | - Zoltán Orgován
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Gábor Kecskeméti
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Zoltán Szabó
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Éva Bartus
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Attila Tököli
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
| | - Gábor K Tóth
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - Tibor V Szalai
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Szt. Gellért tér 4, H-1111, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Tamás Takács
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Egyetem tér 1-3, H-1053, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Elvin de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Ontario, L5 L 1 C6, Mississauga, Canada
| | - László Buday
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - András Perczel
- Laboratory of Structural Chemistry and Biology, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- HUN-REN-ELTE Protein Modeling Research Group, Institute of Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/A, H-1117, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| | - Tamás A Martinek
- Department of Medical Chemistry, University of Szeged, Dóm tér 8, H-6720, Szeged, Hungary
- HUN-REN-SZTE Biomimetic Systems Research Group, Dóm tér 8, H-6720, Szeged, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budafoki út 8, H-1111, Budapest, Hungary
- National Drug Discovery and Development Laboratory, Magyar Tudósok Körútja 2, H-1117, Budapest, Hungary
| |
Collapse
|
3
|
Estevan-Morió E, Ramírez-Larrota JS, Bushi E, Eckhard U. Dissecting Cytophagalysin: Structural and Biochemical Studies of a Bacterial Pappalysin-Family Metallopeptidase. Biomolecules 2024; 14:1604. [PMID: 39766312 PMCID: PMC11674741 DOI: 10.3390/biom14121604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Cytophaga is a genus of Gram-negative bacteria occurring in soil and the gut microbiome. It is closely related to pathogenic Flavobacterium spp. that cause severe diseases in fish. Cytophaga strain L43-1 secretes cytophagalysin (CPL1), a 137 kDa peptidase with reported collagenolytic and gelatinolytic activity. We performed highly-confident structure prediction calculations for CPL1, which identified 11 segments and domains, including a signal peptide for secretion, a prosegment (PS) for latency, a metallopeptidase (MP)-like catalytic domain (CD), and eight immunoglobulin (Ig)-like domains (D3-D10). In addition, two short linkers were found at the D8-D9 and D9-D10 junctions, and the structure would be crosslinked by four disulfide bonds. The CPL1 CD was found closest to ulilysin from Methanosarcina acetivorans, which assigns CPL1 to the lower-pappalysin family within the metzincin clan of MPs. Based on the structure predictions, we aimed to produce constructs spanning the full-length enzyme, as well as PS+CD, PS+CD+D3, and PS+CD+D3+D4. However, we were successful only with the latter three constructs. We could activate recombinant CPL1 by PS removal employing trypsin, and found that both zymogen and mature CPL1 were active in gelatin zymography and against a fluorogenic gelatin variant. This activity was ablated in a mutant, in which the catalytic glutamate described for lower pappalyins and other metzincins was replaced by alanine, and by a broad-spectrum metal chelator. Overall, these results proved that our recombinant CPL1 is a functional active MP, thus supporting the conclusions derived from the structure predictions.
Collapse
Affiliation(s)
- Eva Estevan-Morió
- Synthetic Structural Biology Group, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
- Doctorate in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Juan Sebastián Ramírez-Larrota
- Synthetic Structural Biology Group, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
- Doctorate in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Enkela Bushi
- Synthetic Structural Biology Group, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Ulrich Eckhard
- Synthetic Structural Biology Group, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| |
Collapse
|
4
|
Wang Z, Shi Z, Liao X, Quan G, Dong H, Zhao P, Zhou Y, Shi N, Wang J, Wu Y, Qiao C, Li XY, Zhang R, Wang Z, Wang T, Gao X, Feng J, Luo L. Broad-Spectrum Engineered Multivalent Nanobodies Against SARS-CoV-1/2. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402975. [PMID: 39373693 PMCID: PMC11615778 DOI: 10.1002/advs.202402975] [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: 03/21/2024] [Revised: 06/28/2024] [Indexed: 10/08/2024]
Abstract
SARS-CoV-2 Omicron sublineages escape most preclinical/clinical neutralizing antibodies in development, suggesting that previously employed antibody screening strategies are not well suited to counteract the rapid mutation of SARS-CoV-2. Therefore, there is an urgent need to screen better broad-spectrum neutralizing antibody. In this study, a comprehensive approach to design broad-spectrum inhibitors against both SARS-CoV-1 and SARS-CoV-2 by leveraging the structural diversity of nanobodies is proposed. This includes the de novo design of a fully human nanobody library and the camel immunization-based nanobody library, both targeting conserved epitopes, as well as the development of multivalent nanobodies that bind nonoverlapping epitopes. The results show that trivale B11-E8-F3, three nanobodies joined tandemly in trivalent form, have the broadest spectrum and efficient neutralization activity, which spans from SARS-CoV-1 to SARS-CoV-2 variants. It is also demonstrated that B11-E8-F3 has a very prominent preventive and some therapeutic effect in animal models of three authentic viruses. Therefore, B11-E8-F3 has an outstanding advantage in preventing SARS-CoV-1/SARS-CoV-2 infections, especially in immunocompromised populations or elderly people with high-risk comorbidities.
Collapse
Affiliation(s)
- Zhihong Wang
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Zhuangzhuang Shi
- Key Laboratory of Jilin Province for Zoonosis Prevention and ControlChangchun Veterinary Research InstituteChinese Academy of Agricultural SciencesChangchun130122P. R. China
| | - Xiaochen Liao
- Joint National Laboratory for Antibody Drug Engineeringthe First Affiliated Hospital, Henan UniversityKaifeng CityHenan475004P. R. China
| | - Guiqi Quan
- Hunan Normal University School of medicineChangshaHunan410200P. R. China
| | - Hui Dong
- Joint National Laboratory for Antibody Drug Engineeringthe First Affiliated Hospital, Henan UniversityKaifeng CityHenan475004P. R. China
| | - Pinnan Zhao
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Yangyihua Zhou
- Joint National Laboratory for Antibody Drug Engineeringthe First Affiliated Hospital, Henan UniversityKaifeng CityHenan475004P. R. China
| | - Ning Shi
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Jie Wang
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Yahui Wu
- Hunan Normal University School of medicineChangshaHunan410200P. R. China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Xin ying Li
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Ran Zhang
- Hunan Normal University School of medicineChangshaHunan410200P. R. China
| | - Zekun Wang
- Joint National Laboratory for Antibody Drug Engineeringthe First Affiliated Hospital, Henan UniversityKaifeng CityHenan475004P. R. China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and ControlChangchun Veterinary Research InstituteChinese Academy of Agricultural SciencesChangchun130122P. R. China
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical CountermeasuresBeijing Institute of Pharmacology and ToxicologyBeijing100850P. R. China
| |
Collapse
|
5
|
Harteveld Z, Van Hall-Beauvais A, Morozova I, Southern J, Goverde C, Georgeon S, Rosset S, Defferrard M, Loukas A, Vandergheynst P, Bronstein MM, Correia BE. Exploring "dark-matter" protein folds using deep learning. Cell Syst 2024; 15:898-910.e5. [PMID: 39383860 DOI: 10.1016/j.cels.2024.09.006] [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: 09/17/2023] [Revised: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024]
Abstract
De novo protein design explores uncharted sequence and structure space to generate novel proteins not sampled by evolution. A main challenge in de novo design involves crafting "designable" structural templates to guide the sequence searches toward adopting target structures. We present a convolutional variational autoencoder that learns patterns of protein structure, dubbed Genesis. We coupled Genesis with trRosetta to design sequences for a set of protein folds and found that Genesis is capable of reconstructing native-like distance and angle distributions for five native folds and three novel, the so-called "dark-matter" folds as a demonstration of generalizability. We used a high-throughput assay to characterize the stability of the designs through protease resistance, obtaining encouraging success rates for folded proteins. Genesis enables exploration of the protein fold space within minutes, unrestricted by protein topologies. Our approach addresses the backbone designability problem, showing that small neural networks can efficiently learn structural patterns in proteins. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Zander Harteveld
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Alexandra Van Hall-Beauvais
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Irina Morozova
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Casper Goverde
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | | | - Stéphane Rosset
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Andreas Loukas
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Prescient Design, gRED, Roche, Basel, Switzerland
| | | | | | - Bruno E Correia
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| |
Collapse
|
6
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
7
|
Navratna V, Kumar A, Rana JK, Mosalaganti S. Structure of the human heparan-α-glucosaminide N-acetyltransferase (HGSNAT). eLife 2024; 13:RP93510. [PMID: 39196614 DOI: 10.7554/elife.93510] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
Degradation of heparan sulfate (HS), a glycosaminoglycan (GAG) comprised of repeating units of N-acetylglucosamine and glucuronic acid, begins in the cytosol and is completed in the lysosomes. Acetylation of the terminal non-reducing amino group of α-D-glucosamine of HS is essential for its complete breakdown into monosaccharides and free sulfate. Heparan-α-glucosaminide N-acetyltransferase (HGSNAT), a resident of the lysosomal membrane, catalyzes this essential acetylation reaction by accepting and transferring the acetyl group from cytosolic acetyl-CoA to terminal α-D-glucosamine of HS in the lysosomal lumen. Mutation-induced dysfunction in HGSNAT causes abnormal accumulation of HS within the lysosomes and leads to an autosomal recessive neurodegenerative lysosomal storage disorder called mucopolysaccharidosis IIIC (MPS IIIC). There are no approved drugs or treatment strategies to cure or manage the symptoms of, MPS IIIC. Here, we use cryo-electron microscopy (cryo-EM) to determine a high-resolution structure of the HGSNAT-acetyl-CoA complex, the first step in the HGSNAT-catalyzed acetyltransferase reaction. In addition, we map the known MPS IIIC mutations onto the structure and elucidate the molecular basis for mutation-induced HGSNAT dysfunction.
Collapse
Affiliation(s)
- Vikas Navratna
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Arvind Kumar
- Thermo Fisher Scientific, Waltham, United States
| | - Jaimin K Rana
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Shyamal Mosalaganti
- Life Sciences Institute, University of Michigan, Ann Arbor, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, United States
- Department of Biophysics, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, United States
| |
Collapse
|
8
|
Wang J, Chistov G, Zhang J, Huntington B, Salem I, Sandholu A, Arold ST. P-NADs: PUX-based NAnobody degraders for ubiquitin-independent degradation of target proteins. Heliyon 2024; 10:e34487. [PMID: 39130484 PMCID: PMC11315185 DOI: 10.1016/j.heliyon.2024.e34487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Targeted protein degradation (TPD) allows cells to maintain a functional proteome and to rapidly adapt to changing conditions. Methods that repurpose TPD for the deactivation of specific proteins have demonstrated significant potential in therapeutic and research applications. Most of these methods are based on proteolysis targeting chimaeras (PROTACs) which link the protein target to an E3 ubiquitin ligase, resulting in the ubiquitin-based degradation of the target protein. In this study, we introduce a method for ubiquitin-independent TPD based on nanobody-conjugated plant ubiquitin regulatory X domain-containing (PUX) adaptor proteins. We show that the PUX-based NAnobody Degraders (P-NADs) can unfold a target protein through the Arabidopsis and human orthologues of the CDC48 unfoldase without the need for ubiquitination or initiating motifs. We demonstrate that P-NAD plasmids can be transfected into a human cell line, where the produced P-NADs use the endogenous CDC48 machinery for ubiquitin-independent TPD of a 143 kDa multidomain protein. Thus, P-NADs pave the road for ubiquitin-independent therapeutic TPD approaches. In addition, the modular P-NAD design combined with in vitro and cellular assays provide a versatile platform for elucidating functional aspects of CDC48-based TPD in plants and animals.
Collapse
Affiliation(s)
- Jun Wang
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | | | - Junrui Zhang
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Brandon Huntington
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Israa Salem
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Anandsukeerthi Sandholu
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Stefan T. Arold
- Biological and Environmental Science and Engineering Division, Computational Biology Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
9
|
Goverde CA, Pacesa M, Goldbach N, Dornfeld LJ, Balbi PEM, Georgeon S, Rosset S, Kapoor S, Choudhury J, Dauparas J, Schellhaas C, Kozlov S, Baker D, Ovchinnikov S, Vecchio AJ, Correia BE. Computational design of soluble and functional membrane protein analogues. Nature 2024; 631:449-458. [PMID: 38898281 PMCID: PMC11236705 DOI: 10.1038/s41586-024-07601-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
De novo design of complex protein folds using solely computational means remains a substantial challenge1. Here we use a robust deep learning pipeline to design complex folds and soluble analogues of integral membrane proteins. Unique membrane topologies, such as those from G-protein-coupled receptors2, are not found in the soluble proteome, and we demonstrate that their structural features can be recapitulated in solution. Biophysical analyses demonstrate the high thermal stability of the designs, and experimental structures show remarkable design accuracy. The soluble analogues were functionalized with native structural motifs, as a proof of concept for bringing membrane protein functions to the soluble proteome, potentially enabling new approaches in drug discovery. In summary, we have designed complex protein topologies and enriched them with functionalities from membrane proteins, with high experimental success rates, leading to a de facto expansion of the functional soluble fold space.
Collapse
Affiliation(s)
- Casper A Goverde
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Martin Pacesa
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nicolas Goldbach
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Lars J Dornfeld
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Petra E M Balbi
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sandrine Georgeon
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Stéphane Rosset
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Srajan Kapoor
- Department of Structural Biology, University at Buffalo, Buffalo, NY, USA
| | - Jagrity Choudhury
- Department of Structural Biology, University at Buffalo, Buffalo, NY, USA
| | - Justas Dauparas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Christian Schellhaas
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Simon Kozlov
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 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
| | - Sergey Ovchinnikov
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alex J Vecchio
- Department of Structural Biology, University at Buffalo, Buffalo, NY, USA
| | - Bruno E Correia
- Laboratory of Protein Design and Immunoengineering, École Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
10
|
Hermosilla AM, Berner C, Ovchinnikov S, Vorobieva AA. Validation of de novo designed water-soluble and transmembrane β-barrels by in silico folding and melting. Protein Sci 2024; 33:e5033. [PMID: 38864690 PMCID: PMC11168064 DOI: 10.1002/pro.5033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/14/2024] [Accepted: 05/08/2024] [Indexed: 06/13/2024]
Abstract
In silico validation of de novo designed proteins with deep learning (DL)-based structure prediction algorithms has become mainstream. However, formal evidence of the relationship between a high-quality predicted model and the chance of experimental success is lacking. We used experimentally characterized de novo water-soluble and transmembrane β-barrel designs to show that AlphaFold2 and ESMFold excel at different tasks. ESMFold can efficiently identify designs generated based on high-quality (designable) backbones. However, only AlphaFold2 can predict which sequences have the best chance of experimentally folding among similar designs. We show that ESMFold can generate high-quality structures from just a few predicted contacts and introduce a new approach based on incremental perturbation of the prediction ("in silico melting"), which can reveal differences in the presence of favorable contacts between designs. This study provides a new insight on DL-based structure prediction models explainability and on how they could be leveraged for the design of increasingly complex proteins; in particular membrane proteins which have historically lacked basic in silico validation tools.
Collapse
Affiliation(s)
- Alvaro Martin Hermosilla
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
| | - Carolin Berner
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
| | - Sergey Ovchinnikov
- John Harvard Distinguished Science Fellowship ProgramHarvard UniversityCambridgeMassachusettsUSA
- Present address:
Department of BiologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Anastassia A. Vorobieva
- Structural Biology BrusselsVrije Universiteit BrusselBrusselsBelgium
- VIB‐VUB Center for Structural BiologyBrusselsBelgium
- VIB Center for AI and Computational BiologyBelgium
| |
Collapse
|
11
|
Ma B, Liu D, Wang Z, Zhang D, Jian Y, Zhang K, Zhou T, Gao Y, Fan Y, Ma J, Gao Y, Chen Y, Chen S, Liu J, Li X, Li L. A Top-Down Design Approach for Generating a Peptide PROTAC Drug Targeting Androgen Receptor for Androgenetic Alopecia Therapy. J Med Chem 2024; 67:10336-10349. [PMID: 38836467 DOI: 10.1021/acs.jmedchem.4c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
While large-scale artificial intelligence (AI) models for protein structure prediction and design are advancing rapidly, the translation of deep learning models for practical macromolecular drug development remains limited. This investigation aims to bridge this gap by combining cutting-edge methodologies to create a novel peptide-based PROTAC drug development paradigm. Using ProteinMPNN and RFdiffusion, we identified binding peptides for androgen receptor (AR) and Von Hippel-Lindau (VHL), followed by computational modeling with Alphafold2-multimer and ZDOCK to predict spatial interrelationships. Experimental validation confirmed the designed peptide's binding ability to AR and VHL. Transdermal microneedle patching technology was seamlessly integrated for the peptide PROTAC drug delivery in androgenic alopecia treatment. In summary, our approach provides a generic method for generating peptide PROTACs and offers a practical application for designing potential therapeutic drugs for androgenetic alopecia. This showcases the potential of interdisciplinary approaches in advancing drug development and personalized medicine.
Collapse
Affiliation(s)
- Bohan Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Donghua Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhe Wang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Dize Zhang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanlin Jian
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Kun Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tianyang Zhou
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yibo Gao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yizeng Fan
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jian Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yang Gao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Si Chen
- School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Jing Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
12
|
Feng Z, Xia F, Jiang Z. The Effect of β-Sheet Secondary Structure on All-β Proteins by Molecular Dynamics Simulations. Molecules 2024; 29:2967. [PMID: 38998919 PMCID: PMC11243317 DOI: 10.3390/molecules29132967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 07/14/2024] Open
Abstract
The effect of β-sheet ratio and chain length on all-β proteins was investigated by MD simulations. Protein samples composed of different repeating units with various β-sheet ratios or a different number of repeating units were simulated under a broad temperature range. The simulation results show that the smaller radius of gyration was achieved by the protein with the higher proportion of β-sheet secondary structure, which had the lower nonbonded energy with more HBs within the protein. The root mean square deviation (RMSD) and the root mean square fluctuation (RMSF) both increased with temperature, especially in the case of a longer chain. The visible period was also shown according to the repeated secondary structure. Several minimum values of RMSF were located on the skeleton of Cα atoms participating in the β-sheet, indicating that it is a kind of stable secondary structure. We also concluded that proteins with a short chain or a lower ratio of β-sheet could easily transform their oriented and compact structures to other ones, such as random coils, turns, and even α-helices. These results clarified the relationship from the primary level to the 3D structure of proteins and potentially predicted protein folding.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Applied Physics, China Jiliang University, Hangzhou 310018, China
| | - Fang Xia
- Department of Applied Physics, China Jiliang University, Hangzhou 310018, China
| | - Zhouting Jiang
- Department of Applied Physics, China Jiliang University, Hangzhou 310018, China
| |
Collapse
|
13
|
Navratna V, Kumar A, Rana JK, Mosalaganti S. Structure of the human heparan-α-glucosaminide N-acetyltransferase (HGSNAT). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.23.563672. [PMID: 37961489 PMCID: PMC10634761 DOI: 10.1101/2023.10.23.563672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Degradation of heparan sulfate (HS), a glycosaminoglycan (GAG) comprised of repeating units of N-acetylglucosamine and glucuronic acid, begins in the cytosol and is completed in the lysosomes. Acetylation of the terminal non-reducing amino group of a-D-glucosamine of HS is essential for its complete breakdown into monosaccharides and free sulfate. Heparan-a-glucosaminide N-acetyltransferase (HGSNAT), a resident of the lysosomal membrane, catalyzes this essential acetylation reaction by accepting and transferring the acetyl group from cytosolic acetyl-CoA to terminal a-D-glucosamine of HS in the lysosomal lumen. Mutation-induced dysfunction in HGSNAT causes abnormal accumulation of HS within the lysosomes and leads to an autosomal recessive neurodegenerative lysosomal storage disorder called mucopolysaccharidosis IIIC (MPS IIIC). There are no approved drugs or treatment strategies to cure or manage the symptoms of, MPS IIIC. Here, we use cryo-electron microscopy (cryo-EM) to determine a high-resolution structure of the HGSNAT-acetyl-CoA complex, the first step in HGSNAT catalyzed acetyltransferase reaction. In addition, we map the known MPS IIIC mutations onto the structure and elucidate the molecular basis for mutation-induced HGSNAT dysfunction.
Collapse
Affiliation(s)
- Vikas Navratna
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Arvind Kumar
- Thermo Fisher Scientific, Waltham, Massachusetts, 02451, United States
| | - Jaimin K. Rana
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Shyamal Mosalaganti
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, 48109, United States
- Department of Biophysics, College of Literature, Science and the Arts, University of Michigan, Ann Arbor, Michigan, 48109, United States
| |
Collapse
|
14
|
Roel‐Touris J, Carcelén L, Marcos E. The structural landscape of the immunoglobulin fold by large-scale de novo design. Protein Sci 2024; 33:e4936. [PMID: 38501461 PMCID: PMC10949314 DOI: 10.1002/pro.4936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 03/20/2024]
Abstract
De novo designing immunoglobulin-like frameworks that allow for functional loop diversification shows great potential for crafting antibody-like scaffolds with fully customizable structures and functions. In this work, we combined de novo parametric design with deep-learning methods for protein structure prediction and design to explore the structural landscape of 7-stranded immunoglobulin domains. After screening folding of nearly 4 million designs, we have assembled a structurally diverse library of ~50,000 immunoglobulin domains with high-confidence AlphaFold2 predictions and structures diverging from naturally occurring ones. The designed dataset enabled us to identify structural requirements for the correct folding of immunoglobulin domains, shed light on β-sheet-β-sheet rotational preferences and how these are linked to functional properties. Our approach eliminates the need for preset loop conformations and opens the route to large-scale de novo design of immunoglobulin-like frameworks.
Collapse
Affiliation(s)
- Jorge Roel‐Touris
- Protein Design and Modeling Lab, Department of Structural and Molecular BiologyMolecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| | - Lourdes Carcelén
- Protein Design and Modeling Lab, Department of Structural and Molecular BiologyMolecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| | - Enrique Marcos
- Protein Design and Modeling Lab, Department of Structural and Molecular BiologyMolecular Biology Institute of Barcelona (IBMB), CSICBarcelonaSpain
| |
Collapse
|
15
|
Townsend DR, Towers DM, Lavinder JJ, Ippolito GC. Innovations and trends in antibody repertoire analysis. Curr Opin Biotechnol 2024; 86:103082. [PMID: 38428225 DOI: 10.1016/j.copbio.2024.103082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/07/2023] [Accepted: 01/28/2024] [Indexed: 03/03/2024]
Abstract
Monoclonal antibodies have revolutionized the treatment of human diseases, which has made them the fastest-growing class of therapeutics, with global sales expected to reach $346.6 billion USD by 2028. Advances in antibody engineering and development have led to the creation of increasingly sophisticated antibody-based therapeutics (e.g. bispecific antibodies and chimeric antigen receptor T cells). However, approaches for antibody discovery have remained comparatively grounded in conventional yet reliable in vitro assays. Breakthrough developments in high-throughput single B-cell sequencing and immunoglobulin proteomic serology, however, have enabled the identification of high-affinity antibodies directly from endogenous B cells or circulating immunoglobulin produced in vivo. Moreover, advances in artificial intelligence offer vast potential for antibody discovery and design with large-scale repertoire datasets positioned as the optimal source of training data for such applications. We highlight advances and recent trends in how these technologies are being applied to antibody repertoire analysis.
Collapse
Affiliation(s)
- Douglas R Townsend
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Dalton M Towers
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Jason J Lavinder
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Gregory C Ippolito
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
16
|
Goverde CA, Pacesa M, Goldbach N, Dornfeld LJ, Balbi PEM, Georgeon S, Rosset S, Kapoor S, Choudhury J, Dauparas J, Schellhaas C, Kozlov S, Baker D, Ovchinnikov S, Vecchio AJ, Correia BE. Computational design of soluble functional analogues of integral membrane proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.09.540044. [PMID: 38496615 PMCID: PMC10942269 DOI: 10.1101/2023.05.09.540044] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
De novo design of complex protein folds using solely computational means remains a significant challenge. Here, we use a robust deep learning pipeline to design complex folds and soluble analogues of integral membrane proteins. Unique membrane topologies, such as those from GPCRs, are not found in the soluble proteome and we demonstrate that their structural features can be recapitulated in solution. Biophysical analyses reveal high thermal stability of the designs and experimental structures show remarkable design accuracy. The soluble analogues were functionalized with native structural motifs, standing as a proof-of-concept for bringing membrane protein functions to the soluble proteome, potentially enabling new approaches in drug discovery. In summary, we designed complex protein topologies and enriched them with functionalities from membrane proteins, with high experimental success rates, leading to a de facto expansion of the functional soluble fold space.
Collapse
|
17
|
Rakesh S, Aravind L, Krishnan A. Reappraisal of the DNA phosphorothioate modification machinery: uncovering neglected functional modalities and identification of new counter-invader defense systems. Nucleic Acids Res 2024; 52:1005-1026. [PMID: 38163645 PMCID: PMC10853773 DOI: 10.1093/nar/gkad1213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/03/2023] [Accepted: 12/10/2023] [Indexed: 01/03/2024] Open
Abstract
The DndABCDE systems catalysing the unusual phosphorothioate (PT) DNA backbone modification, and the DndFGH systems, which restrict invasive DNA, have enigmatic and paradoxical features. Using comparative genomics and sequence-structure analyses, we show that the DndABCDE module is commonly functionally decoupled from the DndFGH module. However, the modification gene-neighborhoods encode other nucleases, potentially acting as the actual restriction components or suicide effectors limiting propagation of the selfish elements. The modification module's core consists of a coevolving gene-pair encoding the DNA-scanning apparatus - a DndD/CxC-clade ABC ATPase and DndE with two ribbon-helix-helix (MetJ/Arc) DNA-binding domains. Diversification of DndE's DNA-binding interface suggests a multiplicity of target specificities. Additionally, many systems feature DNA cytosine methylase genes instead of PT modification, indicating the DndDE core can recruit other nucleobase modifications. We show that DndFGH is a distinct counter-invader system with several previously uncharacterized domains, including a nucleotide kinase. These likely trigger its restriction endonuclease domain in response to multiple stimuli, like nucleotides, while blocking protective modifications by invader methylases. Remarkably, different DndH variants contain a HerA/FtsK ATPase domain acquired from multiple sources, including cellular genome-segregation systems and mobile elements. Thus, we uncovered novel HerA/FtsK-dependent defense systems that might intercept invasive DNA during replication, conjugation, or packaging.
Collapse
Affiliation(s)
- Siuli Rakesh
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur 760010, India
| | - L Aravind
- National Center for Biotechnology Information (NCBI), National Library of Medicine (NLM), National Institutes of Health (NIH), Bethesda, MD 20894, USA
| | - Arunkumar Krishnan
- Department of Biological Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Berhampur 760010, India
| |
Collapse
|
18
|
Kortemme T. De novo protein design-From new structures to programmable functions. Cell 2024; 187:526-544. [PMID: 38306980 PMCID: PMC10990048 DOI: 10.1016/j.cell.2023.12.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 02/04/2024]
Abstract
Methods from artificial intelligence (AI) trained on large datasets of sequences and structures can now "write" proteins with new shapes and molecular functions de novo, without starting from proteins found in nature. In this Perspective, I will discuss the state of the field of de novo protein design at the juncture of physics-based modeling approaches and AI. New protein folds and higher-order assemblies can be designed with considerable experimental success rates, and difficult problems requiring tunable control over protein conformations and precise shape complementarity for molecular recognition are coming into reach. Emerging approaches incorporate engineering principles-tunability, controllability, and modularity-into the design process from the beginning. Exciting frontiers lie in deconstructing cellular functions with de novo proteins and, conversely, constructing synthetic cellular signaling from the ground up. As methods improve, many more challenges are unsolved.
Collapse
Affiliation(s)
- Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
19
|
Tawfeeq C, Song J, Khaniya U, Madej T, Wang J, Youkharibache P, Abrol R. Towards a structural and functional analysis of the immunoglobulin-fold proteome. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 138:135-178. [PMID: 38220423 DOI: 10.1016/bs.apcsb.2023.11.002] [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: 01/16/2024]
Abstract
The immunoglobulin fold (Ig fold) domain is a super-secondary structural motif consisting of a sandwich with two layers of β-sheets that is present in many proteins with very diverse biological functions covering a wide range of physiological processes. This domain presents a modular architecture built with β strands connected by variable length loops that has a highly conserved structural core of four β-strands and quite variable β-sheet extensions in the two sandwich layers that enable both divergent and convergent evolutionary mechanisms in the known Ig fold proteome. The central role of this Ig fold's structural plasticity in the evolutionary success of antibodies in our immune system is well established. Nature has also utilized this Ig fold in all domains of life in many different physiological contexts that go way beyond the immune system. Here we will present a structural and functional overview of the utilization of the Ig fold in different biological processes and in different cellular contexts to highlight some of the innumerable ways that this structural motif can interact in multidomain proteins to enable their diversity of functions. This includes shareable specific protein structure visualizations behind those functions that serve as starting points for further explorations of the biomolecular interactions spanning the Ig fold proteome. This overview also highlights how this Ig fold is being utilized through natural adaptation, engineering, and even building from scratch for a range of biotechnological applications.
Collapse
Affiliation(s)
- Caesar Tawfeeq
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, United States
| | - James Song
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Umesh Khaniya
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, United States
| | - Thomas Madej
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Jiyao Wang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Philippe Youkharibache
- Cancer Data Science Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, United States.
| | - Ravinder Abrol
- Department of Chemistry and Biochemistry, California State University Northridge, Northridge, United States.
| |
Collapse
|
20
|
Wang J, Chen C, Yao G, Ding J, Wang L, Jiang H. Intelligent Protein Design and Molecular Characterization Techniques: A Comprehensive Review. Molecules 2023; 28:7865. [PMID: 38067593 PMCID: PMC10707872 DOI: 10.3390/molecules28237865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
In recent years, the widespread application of artificial intelligence algorithms in protein structure, function prediction, and de novo protein design has significantly accelerated the process of intelligent protein design and led to many noteworthy achievements. This advancement in protein intelligent design holds great potential to accelerate the development of new drugs, enhance the efficiency of biocatalysts, and even create entirely new biomaterials. Protein characterization is the key to the performance of intelligent protein design. However, there is no consensus on the most suitable characterization method for intelligent protein design tasks. This review describes the methods, characteristics, and representative applications of traditional descriptors, sequence-based and structure-based protein characterization. It discusses their advantages, disadvantages, and scope of application. It is hoped that this could help researchers to better understand the limitations and application scenarios of these methods, and provide valuable references for choosing appropriate protein characterization techniques for related research in the field, so as to better carry out protein research.
Collapse
Affiliation(s)
| | | | | | - Junjie Ding
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (J.W.); (C.C.); (G.Y.)
| | - Liangliang Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (J.W.); (C.C.); (G.Y.)
| | - Hui Jiang
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China; (J.W.); (C.C.); (G.Y.)
| |
Collapse
|
21
|
Roel-Touris J, Nadal M, Marcos E. Single-chain dimers from de novo immunoglobulins as robust scaffolds for multiple binding loops. Nat Commun 2023; 14:5939. [PMID: 37741853 PMCID: PMC10517939 DOI: 10.1038/s41467-023-41717-5] [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: 03/03/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
Antibody derivatives have sought to recapitulate the antigen binding properties of antibodies, but with improved biophysical attributes convenient for therapeutic, diagnostic and research applications. However, their success has been limited by the naturally occurring structure of the immunoglobulin dimer displaying hypervariable binding loops, which is hard to modify by traditional engineering approaches. Here, we devise geometrical principles for de novo designing single-chain immunoglobulin dimers, as a tunable two-domain architecture that optimizes biophysical properties through more favorable dimer interfaces. Guided by these principles, we computationally designed protein scaffolds that were hyperstable, structurally accurate and robust for accommodating multiple functional loops, both individually and in combination, as confirmed through biochemical assays and X-ray crystallography. We showcase the modularity of this architecture by deep-learning-based diversification, opening up the possibility for tailoring the number, positioning, and relative orientation of ligand-binding loops targeting one or two distal epitopes. Our results provide a route to custom-design robust protein scaffolds for harboring multiple functional loops.
Collapse
Affiliation(s)
- Jorge Roel-Touris
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Marta Nadal
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Enrique Marcos
- Protein Design and Modeling Lab, Department of Structural and Molecular Biology, Molecular Biology Institute of Barcelona (IBMB), CSIC, Baldiri Reixac 10, 08028, Barcelona, Spain.
| |
Collapse
|
22
|
Qiu Y, Wei GW. Artificial intelligence-aided protein engineering: from topological data analysis to deep protein language models. Brief Bioinform 2023; 24:bbad289. [PMID: 37580175 PMCID: PMC10516362 DOI: 10.1093/bib/bbad289] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/16/2023] Open
Abstract
Protein engineering is an emerging field in biotechnology that has the potential to revolutionize various areas, such as antibody design, drug discovery, food security, ecology, and more. However, the mutational space involved is too vast to be handled through experimental means alone. Leveraging accumulative protein databases, machine learning (ML) models, particularly those based on natural language processing (NLP), have considerably expedited protein engineering. Moreover, advances in topological data analysis (TDA) and artificial intelligence-based protein structure prediction, such as AlphaFold2, have made more powerful structure-based ML-assisted protein engineering strategies possible. This review aims to offer a comprehensive, systematic, and indispensable set of methodological components, including TDA and NLP, for protein engineering and to facilitate their future development.
Collapse
Affiliation(s)
- Yuchi Qiu
- Department of Mathematics, Michigan State University, East Lansing, 48824 MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, 48824 MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, 48824 MI, USA
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, 48824 MI, USA
| |
Collapse
|
23
|
Jiang J, Boughter CT, Ahmad J, Natarajan K, Boyd LF, Meier-Schellersheim M, Margulies DH. SARS-CoV-2 antibodies recognize 23 distinct epitopic sites on the receptor binding domain. Commun Biol 2023; 6:953. [PMID: 37726484 PMCID: PMC10509263 DOI: 10.1038/s42003-023-05332-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023] Open
Abstract
The COVID-19 pandemic and SARS-CoV-2 variants have dramatically illustrated the need for a better understanding of antigen (epitope)-antibody (paratope) interactions. To gain insight into the immunogenic characteristics of epitopic sites (ES), we systematically investigated the structures of 340 Abs and 83 nanobodies (Nbs) complexed with the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike protein. We identified 23 distinct ES on the RBD surface and determined the frequencies of amino acid usage in the corresponding CDR paratopes. We describe a clustering method for analysis of ES similarities that reveals binding motifs of the paratopes and that provides insights for vaccine design and therapies for SARS-CoV-2, as well as a broader understanding of the structural basis of Ab-protein antigen (Ag) interactions.
Collapse
Affiliation(s)
- Jiansheng Jiang
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA.
| | - Christopher T Boughter
- Computational Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Javeed Ahmad
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Kannan Natarajan
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Lisa F Boyd
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Martin Meier-Schellersheim
- Computational Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - David H Margulies
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Qiu Y, Wei GW. Artificial intelligence-aided protein engineering: from topological data analysis to deep protein language models. ARXIV 2023:arXiv:2307.14587v1. [PMID: 37547662 PMCID: PMC10402185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Protein engineering is an emerging field in biotechnology that has the potential to revolutionize various areas, such as antibody design, drug discovery, food security, ecology, and more. However, the mutational space involved is too vast to be handled through experimental means alone. Leveraging accumulative protein databases, machine learning (ML) models, particularly those based on natural language processing (NLP), have considerably expedited protein engineering. Moreover, advances in topological data analysis (TDA) and artificial intelligence-based protein structure prediction, such as AlphaFold2, have made more powerful structure-based ML-assisted protein engineering strategies possible. This review aims to offer a comprehensive, systematic, and indispensable set of methodological components, including TDA and NLP, for protein engineering and to facilitate their future development.
Collapse
Affiliation(s)
- Yuchi Qiu
- Department of Mathematics, Michigan State University, East Lansing, 48824, MI, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, 48824, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, 48824, MI, USA
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, 48824, MI, USA
| |
Collapse
|
25
|
Jiang J, Boughter CT, Ahmad J, Natarajan K, Boyd LF, Meier-Schellersheim M, Margulies DH. SARS-CoV-2 antibodies recognize 23 distinct epitopic sites on the receptor binding domain. RESEARCH SQUARE 2023:rs.3.rs-2800118. [PMID: 37333174 PMCID: PMC10275037 DOI: 10.21203/rs.3.rs-2800118/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The COVID-19 pandemic and SARS-CoV-2 variants have dramatically illustrated the need for a better understanding of antigen (epitope)-antibody (paratope) interactions. To gain insight into the immunogenic characteristics of epitopic sites (ES), we systematically investigated the structures of 340 Abs and 83 nanobodies (Nbs) complexed with the Receptor Binding Domain (RBD) of the SARS-CoV-2 spike protein. We identified 23 distinct ES on the RBD surface and determined the frequencies of amino acid usage in the corresponding CDR paratopes. We describe a clustering method for analysis of ES similarities that reveals binding motifs of the paratopes and that provides insights for vaccine design and therapies for SARS-CoV-2, as well as a broader understanding of the structural basis of Ab-protein antigen (Ag) interactions.
Collapse
Affiliation(s)
- Jiansheng Jiang
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - Christopher T. Boughter
- Computational Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - Javeed Ahmad
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - Kannan Natarajan
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - Lisa F. Boyd
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - Martin Meier-Schellersheim
- Computational Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| | - David H. Margulies
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 10892, USA
| |
Collapse
|
26
|
Arai N, Yamamoto E, Koishi T, Hirano Y, Yasuoka K, Ebisuzaki T. Wetting hysteresis induces effective unidirectional water transport through a fluctuating nanochannel. NANOSCALE HORIZONS 2023; 8:652-661. [PMID: 36883765 DOI: 10.1039/d2nh00563h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We propose a water pump that actively transports water molecules through nanochannels. Spatially asymmetric noise fluctuations imposed on the channel radius cause unidirectional water flow without osmotic pressure, which can be attributed to hysteresis in the cyclic transition between the wetting/drying states. We show that the water transport depends on fluctuations, such as white, Brownian, and pink noises. Because of the high-frequency components in white noise, fast switching of open and closed states inhibits channel wetting. Conversely, pink and Brownian noises generate high-pass filtered net flow. Brownian fluctuation leads to a faster water transport rate, whereas pink noise has a higher capability to overcome pressure differences in the opposite direction. A trade-off relationship exists between the resonant frequency of the fluctuation and the flow amplification. The proposed pump can be considered as an analogy for the reversed Carnot cycle, which is the upper limit of the energy conversion efficiency.
Collapse
Affiliation(s)
- Noriyoshi Arai
- Department of Mechanical Engineering, Keio University, Yokohama 223-8522, Japan.
- Computational Astrophysics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Eiji Yamamoto
- Department of System Design Engineering, Keio University, Yokohama, 223-8522, Japan
| | - Takahiro Koishi
- Department of Applied Physics, University of Fukui, Bunkyo, Fukui 910-8507, Japan
| | - Yoshinori Hirano
- Department of Mechanical Engineering, Keio University, Yokohama 223-8522, Japan.
| | - Kenji Yasuoka
- Department of Mechanical Engineering, Keio University, Yokohama 223-8522, Japan.
| | | |
Collapse
|
27
|
Hederman AP, Ackerman ME. Leveraging deep learning to improve vaccine design. Trends Immunol 2023; 44:333-344. [PMID: 37003949 PMCID: PMC10485910 DOI: 10.1016/j.it.2023.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/05/2023] [Accepted: 03/05/2023] [Indexed: 04/03/2023]
Abstract
Deep learning has led to incredible breakthroughs in areas of research, from self-driving vehicles to solutions, to formal mathematical proofs. In the biomedical sciences, however, the revolutionary results seen in other fields are only now beginning to be realized. Vaccine research and development efforts represent an application with high public health significance. Protein structure prediction, immune repertoire analysis, and phylogenetics are three principal areas in which deep learning is poised to provide key advances. Here, we opine on some of the current challenges with deep learning and how they are being addressed. Despite the nascent stage of deep learning applications in immunological studies, there is ample opportunity to utilize this new technology to address the most challenging and burdensome infectious diseases confronting global populations.
Collapse
Affiliation(s)
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA; Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
28
|
Troisi M, Marini E, Abbiento V, Stazzoni S, Andreano E, Rappuoli R. A new dawn for monoclonal antibodies against antimicrobial resistant bacteria. Front Microbiol 2022; 13:1080059. [PMID: 36590399 PMCID: PMC9795047 DOI: 10.3389/fmicb.2022.1080059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial resistance (AMR) is a quickly advancing threat for human health worldwide and almost 5 million deaths are already attributable to this phenomenon every year. Since antibiotics are failing to treat AMR-bacteria, new tools are needed, and human monoclonal antibodies (mAbs) can fill this role. In almost 50 years since the introduction of the first technology that led to mAb discovery, enormous leaps forward have been made to identify and develop extremely potent human mAbs. While their usefulness has been extensively proved against viral pathogens, human mAbs have yet to find their space in treating and preventing infections from AMR-bacteria and fully conquer the field of infectious diseases. The novel and most innovative technologies herein reviewed can support this goal and add powerful tools in the arsenal of weapons against AMR.
Collapse
Affiliation(s)
- Marco Troisi
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Eleonora Marini
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Valentina Abbiento
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Samuele Stazzoni
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Emanuele Andreano
- Monoclonal Antibody Discovery (MAD) Laboratory, Fondazione Toscana Life Sciences, Siena, Italy
| | - Rino Rappuoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Fondazione Biotecnopolo di Siena, Siena, Italy
| |
Collapse
|