201
|
Šolinc G, Srnko M, Merzel F, Crnković A, Kozorog M, Podobnik M, Anderluh G. Cryo-EM structures of a protein pore reveal a cluster of cholesterol molecules and diverse roles of membrane lipids. Nat Commun 2025; 16:2972. [PMID: 40140423 PMCID: PMC11947440 DOI: 10.1038/s41467-025-58334-z] [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: 07/04/2024] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The structure and function of membrane proteins depend on their interactions with lipids that constitute membranes. Actinoporins are α-pore-forming proteins that bind preferentially to sphingomyelin-containing membranes, where they oligomerize and form transmembrane pores. Through a comprehensive cryo-electron microscopic analysis of a pore formed by an actinoporin Fav from the coral Orbicella faveolata, we show that the octameric pore interacts with 112 lipids in the upper leaflet of the membrane, reveal the roles of lipids, and demonstrate that the actinoporin surface is suited for binding multiple receptor sphingomyelin molecules. When cholesterol is present in the membrane, it forms a cluster of four molecules associated with each protomer. Atomistic simulations support the structural data and reveal additional effects of the pore on the lipid membrane. These data reveal a complex network of protein-lipid and lipid-lipid interactions and an underrated role of lipids in the structure and function of transmembrane protein complexes.
Collapse
Affiliation(s)
- Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marija Srnko
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Franci Merzel
- Theory Department, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ana Crnković
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Mirijam Kozorog
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, Slovenia.
| |
Collapse
|
202
|
Hoffpauir ZA, Lamb AL. Identification of the phosphatase essential for riboflavin biosynthesis in Aquifex aeolicus. J Biol Chem 2025; 301:108443. [PMID: 40147773 PMCID: PMC12051542 DOI: 10.1016/j.jbc.2025.108443] [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: 01/21/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
The riboflavin biosynthetic pathway uses dedicated enzymes that function exclusively for riboflavin production. Indeed, the pathway is fully annotated, with the exception of an unknown phosphatase that catalyzes the dephosphorylation of 5-amino-6-ribitylamino-pyrimidinedione 5'-phosphate (ARAPDP) to generate 5-amino-6-ribitylamino-pyrimidinedione (ARAPD), which is the substrate for the penultimate enzyme of the pathway, lumazine synthase. Whereas nonspecific phosphatases from the haloacid dehalogenase superfamily capable of catalyzing the dephosphorylation of ARAPDP have been reported for Bacillus subtilis, Escherichia coli, and Arabadopsis thaliana, we hypothesized that a specific phosphatase may carry out this reaction. Using an anaerobic activity-based screen, two phosphatases from Aquifex aeolicus were identified that dephosphorylate ARAPDP, but only one reconstitutes riboflavin production in a one-pot experiment with the other four enzymes of riboflavin biosynthesis. The first enzyme, annotated as an IMP, is nonspecific, and indiscriminately dephosphorylates ARAPDP along with ribulose 5-phosphate and NADPH, two required substrates of riboflavin biosynthesis. The second enzyme, a histidine family phosphatase, only dephosphorylates ARAPDP in the one-pot experiment thus facilitating riboflavin formation. The structures of both enzymes were determined by X-ray crystallography to reveal the vastly different folds capable of performing the ARAPDP dephosphorylation chemistry. This work has impact both for the production of riboflavin by microbial fermentation and for antimicrobial drug design.
Collapse
Affiliation(s)
- Zoe A Hoffpauir
- Department of Chemistry, 1 UTSA Circle, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Audrey L Lamb
- Department of Chemistry, 1 UTSA Circle, University of Texas at San Antonio, San Antonio, Texas, USA.
| |
Collapse
|
203
|
Yang X, Guo L, Duan H, Fan M, Xu F, Chi X, Pan S, Liu X, Zhang X, Gao P, Zhang F, Wang X, Guo F, Ge J, Ren L, Yang W. Identification of neutralizing nanobodies protecting against poxvirus infection. Cell Discov 2025; 11:31. [PMID: 40133273 PMCID: PMC11937253 DOI: 10.1038/s41421-025-00771-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/05/2025] [Indexed: 03/27/2025] Open
Abstract
An outbreak of mpox has triggered concerns regarding the adequacy of intervention strategies. Passive immunity conferred by neutralizing antibodies exhibits potential in the prophylaxis and treatment of orthopoxvirus infections. Despite this, the investigations of effective antibody therapeutics have been hindered by the varied nature of orthopoxvirus envelope proteins and the intricate mechanisms underpinning viral invasion. Our study involves the production of six mpox virus (MPXV) envelope proteins, which are relatively conservative and considered to play a role in the neutralization process. We employed a synthetic nanobody (Nb) library to derive a broad array of specific Nbs against these viral proteins. We identified a cross-reactive Nb, termed M1R-01, which targets the M1R protein and effectively neutralizes both vaccinia virus (VACV) and MPXV. Notably, the M1R-01-based antibody strategy provided optimal protection against a lethal VACV challenge in mice. Additionally, we determined the crystal structure of the M1R-Nb complex, uncovering novel binding attributes of M1R-01 and detailed conformational epitope information. This work provides a promising candidate for the therapy and prophylaxis of orthopoxvirus infections.
Collapse
Affiliation(s)
- Xuehua Yang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Guo
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huarui Duan
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Miao Fan
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fengwen Xu
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojing Chi
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shengnan Pan
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuying Liu
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinhui Zhang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Peixiang Gao
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fangyuan Zhang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinyi Wang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fei Guo
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiwan Ge
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Lili Ren
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Wei Yang
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
204
|
Wang W, Zhou X, Li W, Zeng P, Guo L, Wang Q, Li J. Inhibitory efficacy and structural insights of Bofutrelvir against SARS-CoV-2 M pro mutants and MERS-CoV M pro. Commun Biol 2025; 8:493. [PMID: 40133408 PMCID: PMC11937426 DOI: 10.1038/s42003-025-07929-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
The COVID-19 pandemic has caused significant global health and economic disruption. Mutations E166N, E166R, E166N, S144A and His163A in the SARS-CoV-2 main protease (Mpro) have been implicated in reducing the efficacy of certain antiviral treatments. Bofutrelvir, a promising inhibitor, has shown effectiveness against SARS-CoV-2 Mpro. This study aims to evaluate the inhibitory effects of Bofutrelvir on the E166N, E166R, His163A, E166V and S144A mutants of SARS-CoV-2 Mpro, as well as on MERS-CoV Mpro. Our findings indicate a substantial reduction in the inhibitory potency of Bofutrelvir against these mutants and MERS-CoV, with IC50 values significantly higher than those for the wild-type SARS-CoV-2 Mpro. Specifically, the E166N, E166R, E166V, S144A, and H163A mutations significantly reduce the binding affinity and inhibitory effectiveness of Bofutrelvir due to disrupted hydrogen bonds, altered binding site stability, and reduced enzyme activity. Structural analysis of the crystal complexes showed that changes in interactions at the S1 subsite in the mutants and the loss of hydrogen bonds at the S4 subsite in MERS-CoV Mpro are critical factors contributing to the diminished inhibitory activity. These insights reveal the necessity of ongoing structural analysis to adapt therapeutic strategies.
Collapse
Affiliation(s)
- Weiwei Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Xuelan Zhou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Wenwen Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Pei Zeng
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen, China
| | - Li Guo
- Jiangxi Jmerry Biopharmaceutical Co, Ltd, Ganzhou, China
| | - Qisheng Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China.
| | - Jian Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
205
|
Tröster V, Wong RP, Börgel A, Cakilkaya B, Renz C, Möckel MM, Eifler-Olivi K, Marinho J, Reinberg T, Furler S, Schaefer JV, Plückthun A, Wolf E, Ulrich HD. Custom affinity probes reveal DNA-damage-induced, ssDNA-independent chromatin SUMOylation in budding yeast. Cell Rep 2025; 44:115353. [PMID: 40019834 DOI: 10.1016/j.celrep.2025.115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/17/2024] [Accepted: 02/05/2025] [Indexed: 03/29/2025] Open
Abstract
The small ubiquitin-related modifier SUMO regulates cellular processes in eukaryotes either by modulating individual protein-protein interactions or with relaxed substrate selectivity by group modification. Here, we report the isolation and characterization of designed ankyrin repeat protein (DARPin)-based affinity probes directed against budding yeast SUMO (Smt3). We validate selected DARPins as compartment-specific inhibitors or neutral detection agents. Structural characterization reveals a recognition mode distinct from that of natural SUMO interactors. In vivo application pinpoints Smt3's essential function to the nucleus and demonstrates DARPin-mediated sensitization toward various stress conditions. A subset of selected clones is validated as SUMOylation reporters in cells. In this manner, we identify a DNA-damage-induced nuclear SUMOylation response that-in contrast to previously reported chromatin group SUMOylation-is independent of single-stranded DNA and the SUMO-E3 Siz2 but depends on Mms21 and likely reflects late intermediates of homologous recombination. Thus, Smt3-specific DARPins can provide insight into the dynamics of SUMOylation in defined subcellular structures.
Collapse
Affiliation(s)
- Vera Tröster
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Ronald P Wong
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Arne Börgel
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Baris Cakilkaya
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Christian Renz
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Martin M Möckel
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | | | - Joana Marinho
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Thomas Reinberg
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Sven Furler
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Eva Wolf
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Helle D Ulrich
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany.
| |
Collapse
|
206
|
Seager BA, Lim PS, Lai KH, Feufack-Donfack LB, Dass S, Xiao X, Jung NC, Abraham A, Grigg MJ, Anstey NM, William T, Sattabongkot J, Leis A, Longley RJ, Duraisingh MT, Popovici J, Wilson DW, Scally SW, Cowman AF. PTRAMP, CSS and Ripr form a conserved complex required for merozoite invasion of Plasmodium species into erythrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.644866. [PMID: 40196582 PMCID: PMC11974866 DOI: 10.1101/2025.03.25.644866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Invasion of erythrocytes by members of the Plasmodium genus is an essential step of the parasite lifecycle, orchestrated by numerous host-parasite interactions. In P. falciparum Rh5, with PfCyRPA, PfRipr, PfCSS, and PfPTRAMP, forms the essential PCRCR complex which binds basigin on the erythrocyte surface. Rh5 is restricted to P. falciparum and its close relatives; however, PTRAMP, CSS and Ripr orthologs are present across the Plasmodium genus. We investigated PTRAMP, CSS and Ripr orthologs from three species to elucidate common features of the complex. Like P. falciparum, PTRAMP and CSS form a disulfide-linked heterodimer in both P. vivax and P. knowlesi with all three species forming a complex (PCR) with Ripr by binding its C-terminal region. Cross-reactive antibodies targeting the PCR complex differentially inhibit merozoite invasion. Cryo-EM visualization of the P. knowlesi PCR complex confirmed predicted models and revealed a core invasion scaffold in Plasmodium spp. with implications for vaccines targeting multiple species of malaria-causing parasites.
Collapse
Affiliation(s)
- Benjamin A. Seager
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Pailene S. Lim
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Keng Heng Lai
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Australia 5005
| | | | - Sheena Dass
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Xiao Xiao
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Nicolai C. Jung
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Anju Abraham
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
| | - Matthew J. Grigg
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
| | - Nicholas M. Anstey
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Northern Territory, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
| | - Timothy William
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Clinical Research Unit, Kota Kinabalu, Sabah, Malaysia
- Clinical Research Centre-Queen Elizabeth Hospital, Ministry of Health, Kota Kinabalu, Sabah, Malaysia
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Andrew Leis
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Rhea J. Longley
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Manoj T. Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- Infectious Disease Epidemiology and Analytics G5 Unit, Institut Pasteur, Université Paris Cité, Paris, France
| | - Danny W. Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Australia 5005
- Burnet Institute, 85 Commercial Road, Melbourne 3004, Victoria, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, 5005, SA, Australia
| | - Stephen W. Scally
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| | - Alan F. Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- University of Melbourne, Melbourne 3010, Australia
| |
Collapse
|
207
|
Kalita P, Khatavkar O, Uwase G, Korshunova Y, Hu Y, Wagner ND, Xu J, Pan J, Nix JC, Gross ML, Brody SL, Borek D, Amarasinghe GK, Payton JE, Leung DW. Molecular basis for human respiratory syncytial virus transcriptional regulator NS1 interactions with MED25. Nat Commun 2025; 16:2883. [PMID: 40128225 PMCID: PMC11933697 DOI: 10.1038/s41467-025-58216-4] [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: 07/11/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
The Mediator complex facilitates interactions between transcription factors and RNA polymerase II, a process that is required for host gene transcription, including in response to viral infections. Among the many subunits in the Mediator complex, the MED25 subunit has been shown to be a target for viral activators during infection. Here we provide the molecular basis for the interaction between human respiratory syncytial virus (hRSV) nonstructural 1 protein (NS1) and the activator interaction domain (ACID) of MED25. The X-ray crystal structure of the complex revealed that NS1 straddles and binds two faces of MED25 ACID. This interaction is distinct from previously known viral activators. Importantly, our data support the conformational flexibility of viral transcriptional regulators. Furthermore, ChIP-seq and RNA-seq analysis identified the ATF3 transcription factor and a role for NS1/Mediator/ATF3 interaction in host gene regulation in hRSV infections. Our findings provide a molecular basis for hRSV NS1-based regulation of host gene transcription and reveal how viruses exploit the conformational heterogeneity at fuzzy transcription activator interfaces.
Collapse
Affiliation(s)
- Parismita Kalita
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Oam Khatavkar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Uwase
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yulia Korshunova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yuying Hu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicole D Wagner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Jian Xu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jiehong Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jay C Nix
- Molecular Biology Consortium, Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO, USA
| | - Steven L Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dominika Borek
- Departments of Biophysics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daisy W Leung
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
208
|
Moussawi MA, de Azambuja F, Parac-Vogt TN. Discrete Hybrid Vanadium-oxo Cluster as a Targeted Tool for Selective Protein Oxidative Modifications and Cleavage. Angew Chem Int Ed Engl 2025; 64:e202423078. [PMID: 39792069 DOI: 10.1002/anie.202423078] [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: 11/26/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/12/2025]
Abstract
Understanding the impact of oxidative modification on protein structure and functions is essential for developing therapeutic strategies to combat macromolecular damage and cell death. However, selectively inducing oxidative modifications in proteins under physiological conditions remains challenging. Herein we demonstrate that [V6O13{(OCH2)3CCH2OH}2]2- (V6-OH) hybrid metal-oxo cluster can be used for selective protein oxidative cleavage and modifications. We present the first example of a protein-bound hybrid vanadate cluster, where its interactions with protein surfaces and the redox activity of vanadium enable selective oxidative modifications. Single Crystal X-ray Diffraction (SC-XRD) of the V6-OH and hen egg white lysozyme (HEWL) complex revealed that the binding is dictated both by the inorganic core and the organic ligands attached to it. Selective oxidation or cleavage of HEWL occurs under physiological conditions by producing reactive oxygen species (ROS) in presence of ascorbate (Asc) as a reducing agent. The outcome of the oxidative reaction can be tuned by varying the concentration of V6-OH to result either in selective oxidation of the amino acid side chains or peptide bond cleavage. LC-MS and crystallography revealed that oxidative modifications were mainly concentrated near the cluster binding sites, providing spatial control of ROS production. This study advances our understanding of vanadium's role in biological systems and demonstrates the potential of hybrid metal-oxo clusters in protein modification.
Collapse
Affiliation(s)
- Mhamad Aly Moussawi
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001, Leuven, Belgium
| | | | | |
Collapse
|
209
|
Marminon C, Werner C, Gast A, Herfindal L, Charles J, Lindenblatt D, Aichele D, Mularoni A, Døskeland SO, Jose J, Niefind K, Le Borgne M. Exploring the biological potential of the brominated indenoindole MC11 and its interaction with protein kinase CK2. Biol Chem 2025:hsz-2024-0160. [PMID: 40116007 DOI: 10.1515/hsz-2024-0160] [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: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 03/23/2025]
Abstract
Protein kinase CK2 is a promising therapeutic target, especially in oncology. Over the years, various inhibitors have been developed, with polyhalogenated scaffolds emerging as a particularly effective class. Halogens like bromine and chlorine enhance inhibitor stability by forming additional interactions within the ATP pocket. Among halogenated scaffolds, benzotriazole and benzimidazole have led to potent molecules such as 4,5,6,7-tetrabromo-1H-benzotriazole (IC50 = 300 nM) and 4,5,6,7-tetrabromo-2-(dimethylamino)benzimidazole (IC50 = 140 nM). Modifications, including 4,5,6-tribromo-7-ethyl-1H-benzotriazole (IC50 = 160 nM), further improved activity. Changing scaffolds while retaining halogens has enabled design of new inhibitors. Flavonols, dibenzofuranones, and the indeno[1,2-b]indole scaffold are key examples. Halogenation of the reference molecule 5-isopropyl-5,6,7,8-tetrahydroindeno[1,2-b]indole-9,10-dione (4b, IC50 = 360 nM) significantly boosted potency. The study focused on introducing four halogens, yielding to the compound 1,2,3,4-tetrabromo-5-isopropyl-5,6,7,8-tetrahydroindeno[1,2-b]indole-9,10-dione (MC11), with an IC50 of 16 nM. Co-crystallography revealed how bromine atoms enhance binding, and MC11 demonstrated strong in cellulo activity, particularly against leukemic cell lines like IPC-Bcl2.
Collapse
Affiliation(s)
- Christelle Marminon
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | - Christian Werner
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Alexander Gast
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Lars Herfindal
- Department of Clinical Science, University of Bergen, N-5009 Bergen, Norway
| | - Johana Charles
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | - Dirk Lindenblatt
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Dagmar Aichele
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Angélique Mularoni
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| | | | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, PharmaCampus, Corrensstraße 48, D-48149 Münster, Germany
| | - Karsten Niefind
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zülpicher Str. 47, D-50674 Köln, Germany
| | - Marc Le Borgne
- Small Molecules for Biological Targets Team, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, University of Lyon, F-69373 Lyon, France
| |
Collapse
|
210
|
Meng C, Guo W, Xiao C, Wen Y, Zhu X, Zhang Q, Liang Y, Li H, Xu S, Qiu Y, Chen H, Lin WJ, Wu B. Structural basis for psilocybin biosynthesis. Nat Commun 2025; 16:2827. [PMID: 40121242 PMCID: PMC11929908 DOI: 10.1038/s41467-025-58239-x] [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: 07/17/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Psilocybin shows significant therapeutic potential for psilocybin-assisted psychotherapy in addressing various psychiatric conditions. The biosynthetic approach promises rapid and efficient production of psilocybin. Understanding the enzymes that contribute to the biosynthesis of psilocybin can enhance its production process. In this study, we elucidate the crystal structures of L-tryptophan-specific decarboxylase PsiD in both its apo and tryptamine-bound states, the 4-hydroxytryptamine kinase PsiK bound to its substrate, and several forms of the methyltransferase PsiM in either apo or substrate-bound forms derived from the psychedelic mushroom. Structure-based evaluations reveal the mechanisms of self-cleavage and self-inhibition in PsiD, along with the sequential catalytic steps from 4-hydroxytryptamine to the final compound, psilocybin. Additionally, we showcase the antidepressant properties of biosynthetic intermediates of psilocybin on female mice experiencing depression-like behaviors induced by sub-chronic variable stress. Our studies establish a structural basis for the future biosynthetic production of psilocybin using these enzymes and emphasize the clinical potential of norbaeocystin.
Collapse
Affiliation(s)
- Chunyan Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
| | - Wenting Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chuan Xiao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yan Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xudong Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qingrong Zhang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Yuxuan Liang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Hongwei Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yuntan Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China.
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.
| | - Baixing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China.
| |
Collapse
|
211
|
Walla B, Maslakova A, Bischoff D, Janowski R, Niessing D, Weuster-Botz D. Rational Introduction of Electrostatic Interactions at Crystal Contacts to Enhance Protein Crystallization of an Ene Reductase. Biomolecules 2025; 15:467. [PMID: 40305164 PMCID: PMC12024682 DOI: 10.3390/biom15040467] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Protein crystallization is an alternative to well-established but cost-intensive and time-consuming chromatography in biotechnological processes, with protein crystallization defined as an essential unit operation for isolating proteins, e.g., active pharmaceutical ingredients. Crystalline therapeutic proteins attract interest in formulation and delivery processes of biopharmaceuticals due to the high purity, concentration, and stability of the crystalline state. Although improving protein crystallization is mainly achieved by high-throughput screening of crystallization conditions, recent studies have established a rational protein engineering approach to enhance crystallization for two homologous alcohol dehydrogenases from Lactobacillus brevis (LbADH) and Lactobacillus kefiri (LkADH). As generalizing crystallization processes across a wide range of target proteins remains challenging, this study takes a further step by applying the successful crystal contact engineering strategies for LbADH/LkADH to a non-homologous protein, an NADH-binding derivative of the Nostoc sp. PCC 1720 ene reductase (NspER1-L1,5). Here, the focus lies on introducing electrostatic interactions at crystal contacts, specifically between lysine and glutamic acid. Out of the nine tested NspER1-L1,5 mutants produced in E. coli, six crystallized, while four mutants revealed an increased propensity to crystallize in static µL-batch crystallization compared to the wild type: Q204K, Q350K, D352K, and T354K. The best-performing mutant Q204K was selected for upscaling, crystallizing faster than the wild type in a stirred batch crystallizer. Even when spiked with E. coli cell lysate, the mutant maintained increased crystallizability compared to the wild type. The results of this study highlight the potential of crystal contact engineering as a reliable tool for improving protein crystallization as an alternative to chromatography, paving the way for more efficient biotechnological downstream processing.
Collapse
Affiliation(s)
- Brigitte Walla
- Biochemical Engineering, Department of Energy and Process Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany; (B.W.); (D.B.)
| | - Anna Maslakova
- Biochemical Engineering, Department of Energy and Process Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany; (B.W.); (D.B.)
| | - Daniel Bischoff
- Biochemical Engineering, Department of Energy and Process Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany; (B.W.); (D.B.)
| | - Robert Janowski
- Molecular Targets and Therapeutics Center, Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany (D.N.)
| | - Dierk Niessing
- Molecular Targets and Therapeutics Center, Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany (D.N.)
- Institute of Pharmaceutical Biotechnology, Ulm University, James-Franck-Ring N27, 89081 Ulm, Germany
| | - Dirk Weuster-Botz
- Biochemical Engineering, Department of Energy and Process Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany; (B.W.); (D.B.)
| |
Collapse
|
212
|
Ye JS, Majumdar A, Park BC, Black MH, Hsieh TS, Osinski A, Servage KA, Kulkarni K, Naidoo J, Alto NM, Stratton MM, Alfandari D, Ready JM, Pawłowski K, Tomchick DR, Tagliabracci VS. Bacterial ubiquitin ligase engineered for small molecule and protein target identification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644192. [PMID: 40166235 PMCID: PMC11957136 DOI: 10.1101/2025.03.20.644192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The Legionella SidE effectors ubiquitinate host proteins independently of the canonical E1-E2 cascade. Here we engineer the SidE ligases to develop a modular proximity ligation approach for the identification of targets of small molecules and proteins, which we call SidBait. We validate the method with known small molecule-protein interactions and use it to identify CaMKII as an off-target interactor of the breast cancer drug ribociclib. Structural analysis and activity assays confirm that ribociclib binds the CaMKII active site and inhibits its activity. We further customize SidBait to identify protein-protein interactions, including substrates for enzymes, and discover the F-actin capping protein (CapZ) as a target of the Legionella effector RavB during infection. Structural and biochemical studies indicate that RavB allosterically binds CapZ and decaps actin, thus functionally mimicking eukaryotic CapZ interacting proteins. Collectively, our results establish SidBait as a reliable tool for identifying targets of small molecules and proteins.
Collapse
Affiliation(s)
- James S. Ye
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Abir Majumdar
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Brenden C. Park
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Miles H. Black
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ting-Sung Hsieh
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam Osinski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kelly A. Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kartik Kulkarni
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jacinth Naidoo
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Neal M. Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Margaret M. Stratton
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Joseph M. Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Krzysztof Pawłowski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Diana R. Tomchick
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vincent S. Tagliabracci
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
213
|
Jia M, Chen Y, Wang J, Wang J, Ma Y, Wang Y, Ma Q, Zhang Y, Liu W, Liu K. His 70 of Acetivibrio alkalicellulosi Cel5A is important for efficient hydrolysis of short cellodextrins. AMB Express 2025; 15:53. [PMID: 40111668 PMCID: PMC11926323 DOI: 10.1186/s13568-025-01858-w] [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: 02/03/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
Cellulose, a linear glucan linked by β-1,4 glycosidic bonds, is the most abundant renewable polysaccharide on earth. Complete enzymatic hydrolysis of cellulose liberates the readily metabolizable glucose that could be further converted to valuable biocommodities, and essential to this process are cellulases that hydrolyze the β-1,4 glycosidic bonds. Cellulases are among the most intensively studied and best understood enzymes, and many key residues have been uncovered and interrogated with respect to their functions in catalysis and/or substrate binding. However, it remains to be explored whether additional residues, especially in many poorly characterized cellulases such as processive endoglucanases, might also be functionally important. Here, we investigated a processive endoglucanase from an alkaliphilic bacterium Acetivibrio alkalicellulosi AaCel5A that consists of a glycohydrolase family 5 (GH5) domain and two tandem carbohydrate-binding module family 6 (CBM6) domains. Via structure-guided engineering, we uncovered the functional importance of a previously underexplored but relatively conserved histidine (histidine70 or His70). His70 itself appears to be largely dispensable for hydrolyzing β-1,4 glycosidic bonds, but it is important for efficient hydrolysis of short cellodextrins such as cellotriose, cellotetraose, and cellopentaose, likely through its ability to coordinate substrate binding. Our work thus provides important mechanistic insights into how processive endoglucanases may act on short cellodextrins.
Collapse
Affiliation(s)
- Mengxiang Jia
- Department of Bioengineering, Tianjin University of Science and Technology, Tianjin, China
| | - Yangyang Chen
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Jingting Wang
- in vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Jiahan Wang
- in vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Yihua Ma
- in vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Yujiao Wang
- Cosychem Technology (Tianjin) Co., Ltd., Tianjin, China
| | - Qian Ma
- Department of Bioengineering, Tianjin University of Science and Technology, Tianjin, China
| | - Yiheng Zhang
- in vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China.
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin, China.
| | - Weidong Liu
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
| | - Kuanqing Liu
- in vitro Synthetic Biology Center, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China.
- National Center of Technology Innovation for Synthetic Biology, Tianjin, China.
| |
Collapse
|
214
|
Hermanns T, Kolek S, Uthoff M, de Heiden RA, Mulder MPC, Baumann U, Hofmann K. A family of bacterial Josephin-like deubiquitinases with an irreversible cleavage mode. Mol Cell 2025; 85:1202-1215.e5. [PMID: 40037356 DOI: 10.1016/j.molcel.2025.02.002] [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: 07/24/2024] [Revised: 12/05/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025]
Abstract
Many intracellular bacteria secrete deubiquitinase (DUB) effectors into eukaryotic host cells to keep the bacterial surface or the enclosing vesicle membrane free of ubiquitin marks. This study describes a family of DUBs from several bacterial genera, including Simkania, Parachlamydia, Burkholderia, and Pigmentiphaga, which is structurally related to eukaryotic Josephin-type DUBs but contains members that catalyze a unique destructive substrate deubiquitination. These ubiquitin C-terminal clippases (UCCs) cleave ubiquitin before the C-terminal diGly motif, thereby truncating the modifier and leaving a remnant on the substrate. By comparing the crystal structures of substrate-bound clippases and a closely related conventional DUB, we identified the factors causing this shift and found them to be conserved in other clippases, including one highly specific for M1-linked ubiquitin chains. This enzyme class has great potential to serve as tools for studying the ubiquitin system, particularly aspects involving branched chains.
Collapse
Affiliation(s)
- Thomas Hermanns
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany
| | - Susanne Kolek
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany
| | - Matthias Uthoff
- Institute of Biochemistry, University of Cologne, Zülpicher Straße 47, 50674 Cologne, Germany
| | - Richard A de Heiden
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333ZC Leiden, the Netherlands
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333ZC Leiden, the Netherlands
| | - Ulrich Baumann
- Institute of Biochemistry, University of Cologne, Zülpicher Straße 47, 50674 Cologne, Germany
| | - Kay Hofmann
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany.
| |
Collapse
|
215
|
Fukunaga K, Teramoto T, Nakashima M, Ohtani T, Katsuki R, Matsuura T, Yokobayashi Y, Kakuta Y. Structural insights into lab-coevolved RNA-RBP pairs and applications of synthetic riboswitches in cell-free system. Nucleic Acids Res 2025; 53:gkaf212. [PMID: 40119732 PMCID: PMC11928940 DOI: 10.1093/nar/gkaf212] [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: 01/16/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
CS1-LS4 and CS2-LS12 are ultra-high affinity and orthogonal RNA-protein pairs that were identified by PD-SELEX (Phage Display coupled with Systematic Evolution of Ligands by EXponential enrichment). To investigate the molecular basis of the lab-coevolved RNA-RBP pairs, we determined the structures of the CS1-LS4 and CS2-LS12 complexes and the LS12 homodimer in an RNA-free state by X-ray crystallography. The structural analyses revealed that the lab-coevolved RNA-RBPs have acquired unique molecular recognition mechanisms, whereas the overall structures of the RNP complexes were similar to the typical kink-turn RNA-L7Ae complex. The orthogonal RNA-RBP pairs were applied to construct high-performance cell-free riboswitches that regulate translation in response to LS4 or LS12. In addition, by using the orthogonal protein-responsive switches, we generated an AND logic gate that outputs staphylococcal γ-hemolysin in cell-free system and carried out hemolysis assay and calcein leakage assay using rabbit red blood cells and artificial cells, respectively.
Collapse
Affiliation(s)
- Keisuke Fukunaga
- Earth-Life Science Institute (ELSI), Institute of Science Tokyo, Tokyo 152 8550, Japan
- Nucleic Acid Chemistry and Engineering Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904 0495, Japan
- Institute for Tenure Track Promotion, University of Miyazaki, Miyazaki 889 2192, Japan
| | - Takamasa Teramoto
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 819 0395, Japan
| | - Momoka Nakashima
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 819 0395, Japan
| | - Toshitaka Ohtani
- Earth-Life Science Institute (ELSI), Institute of Science Tokyo, Tokyo 152 8550, Japan
| | - Riku Katsuki
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications (UEC), Tokyo 182 8585, Japan
| | - Tomoaki Matsuura
- Earth-Life Science Institute (ELSI), Institute of Science Tokyo, Tokyo 152 8550, Japan
| | - Yohei Yokobayashi
- Nucleic Acid Chemistry and Engineering Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa 904 0495, Japan
| | - Yoshimitsu Kakuta
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 819 0395, Japan
| |
Collapse
|
216
|
Bano F, Banerji S, Ni T, Green DE, Cook KR, Manfield IW, DeAngelis PL, Paci E, Lepšík M, Gilbert RJC, Richter RP, Jackson DG. Structure and unusual binding mechanism of the hyaluronan receptor LYVE-1 mediating leucocyte entry to lymphatics. Nat Commun 2025; 16:2754. [PMID: 40113779 PMCID: PMC11926218 DOI: 10.1038/s41467-025-57866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Immune surveillance involves the continual migration of antigen-scavenging immune cells from the tissues to downstream lymph nodes via lymphatic vessels. To enable such passage, cells first dock with the lymphatic entry receptor LYVE-1 on the outer surface of endothelium, using their endogenous hyaluronan glycocalyx, anchored by a second hyaluronan receptor, CD44. Why the process should require two different hyaluronan receptors and by which specific mechanism the LYVE-1•hyaluronan interaction enables lymphatic entry is however unknown. Here we describe the crystal structures and binding mechanics of murine and human LYVE-1•hyaluronan complexes. These reveal a highly unusual, sliding mode of ligand interaction, quite unlike the conventional sticking mode of CD44, in which the receptor grabs free hyaluronan chain-ends and winds them in through conformational re-arrangements in a deep binding cleft, lubricated by a layer of structured waters. Our findings explain the mode of action of a dedicated lymphatic entry receptor and define a distinct, low tack adhesive interaction that enables migrating immune cells to slide through endothelial junctions with minimal resistance, while clinging onto their hyaluronan glycocalyx for essential downstream functions.
Collapse
Affiliation(s)
- Fouzia Bano
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Bragg Centre for Materials Research, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
- CIC biomaGUNE, Paseo Miramon 182, 20014, Donostia-San Sebastián, Spain
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
| | - Suneale Banerji
- Medical Research Council Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Tao Ni
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
- L1-60, Laboratory Block, University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Dixy E Green
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73126, USA
| | - Kalila R Cook
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Bragg Centre for Materials Research, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Iain W Manfield
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Paul L DeAngelis
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73126, USA
| | - Emanuele Paci
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
- Dipartimento di Fisica e Astronomia, Università di Bologna, 40127, Bologna, Italy
| | - Martin Lepšík
- CERMAV, Université Grenoble Alpes, CNRS, 38000, Grenoble, France.
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 16610, Prague 6, Czech Republic.
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| | - Ralf P Richter
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK.
- Bragg Centre for Materials Research, University of Leeds, Leeds, LS2 9JT, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
- CIC biomaGUNE, Paseo Miramon 182, 20014, Donostia-San Sebastián, Spain.
| | - David G Jackson
- Medical Research Council Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
217
|
Yirmiya E, Hobbs SJ, Leavitt A, Osterman I, Avraham C, Hochhauser D, Madhala B, Skovorodka M, Tan JMJ, Toyoda HC, Chebotar I, Itkin M, Malitsky S, Amitai G, Kranzusch PJ, Sorek R. Structure-guided discovery of viral proteins that inhibit host immunity. Cell 2025; 188:1681-1692.e17. [PMID: 39855193 DOI: 10.1016/j.cell.2024.12.035] [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: 04/05/2024] [Revised: 10/29/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Viruses encode proteins that inhibit host defenses, but sifting through the millions of available viral sequences for immune-modulatory proteins has been so far impractical. Here, we develop a process to systematically screen virus-encoded proteins for inhibitors that physically bind host immune proteins. Focusing on Thoeris and CBASS, bacterial defense systems that are the ancestors of eukaryotic Toll/interleukin-1 receptor (TIR) and cyclic GMP-AMP synthase (cGAS) immunity, we discover seven families of Thoeris and CBASS inhibitors, encompassing thousands of genes widespread in phages. Verified inhibitors exhibit extensive physical interactions with the respective immune protein counterpart, with all inhibitors blocking the active site of the immune protein. Remarkably, a phage-encoded inhibitor of bacterial TIR proteins can bind and inhibit distantly related human and plant immune TIRs, and a phage-derived inhibitor of bacterial cGAS-like enzymes can inhibit the human cGAS. Our results demonstrate that phages are a reservoir for immune-modulatory proteins capable of inhibiting bacterial, animal, and plant immunity.
Collapse
Affiliation(s)
- Erez Yirmiya
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Samuel J Hobbs
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Azita Leavitt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ilya Osterman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Carmel Avraham
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Dina Hochhauser
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Barak Madhala
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Marharyta Skovorodka
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Joel M J Tan
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Hunter C Toyoda
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Igor Chebotar
- High Performance Computing Section, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maxim Itkin
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sergey Malitsky
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gil Amitai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Philip J Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
218
|
Urdiciain A, Madej T, Wang J, Song J, Erausquin E, Youkharibache P, López-Sagaseta J. Unusual traits shape the architecture of the Ig ancestor molecule. Commun Biol 2025; 8:463. [PMID: 40113983 PMCID: PMC11926128 DOI: 10.1038/s42003-025-07830-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Understanding the ancestral Ig domain's molecular structure and tracing the evolution of Ig-like proteins are fundamental components missing from our comprehension of their evolutionary trajectory and function. We have determined high-resolution structures of two Ig-like proteins from the evolutionary most ancestral phylum, Porifera. The structures reveal N-terminal Ig-like domains with an unconventional configuration of features that set them apart from canonical Ig domains. These findings prompted us to call this novel domain as Ig "Early Variable" (EV)-set. Remarkably, the EV-sets are linked to C1-set domains. To the best of our knowledge, the C1-set has not been previously reported in non-vertebrates. The IgV and IgC1 tandems and their combination into functional Ig-like receptors are part of the adaptive immune system in higher vertebrates, which allows for highly specific immune responses. By unveiling important clues into the molecular configuration of ancestral Ig domains, these findings challenge and expand our understanding of how immunity has evolved within its current landscape.
Collapse
Affiliation(s)
- Alejandro Urdiciain
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed, 31008, Navarra, Spain
- Public University of Navarra (UPNA), Pamplona, 31008, Navarra, Spain
- Navarra University Hospital, Pamplona, 31008, Navarra, Spain
| | - Thomas Madej
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jiyao Wang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - James Song
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Elena Erausquin
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed, 31008, Navarra, Spain
- Public University of Navarra (UPNA), Pamplona, 31008, Navarra, Spain
- Navarra University Hospital, Pamplona, 31008, Navarra, Spain
| | - Philippe Youkharibache
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jacinto López-Sagaseta
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed, 31008, Navarra, Spain.
- Public University of Navarra (UPNA), Pamplona, 31008, Navarra, Spain.
- Navarra University Hospital, Pamplona, 31008, Navarra, Spain.
| |
Collapse
|
219
|
Zhu Z, Li M, Xu Q, Huang L, Zhou H, Wang W, Wang Q, Yu F. Mechanistic insights into cyclodextrins as substrates and inhibitors of GH57 family amylopullulanase from Aquifex aeolicus. J Struct Biol 2025; 217:108199. [PMID: 40120836 DOI: 10.1016/j.jsb.2025.108199] [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: 12/19/2024] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Maltooligosaccharides (MOs) have gained significant attention in the food and pharmaceutical industries owing to their valuable functional properties, including controlled sweetness, digestibility, and enhanced bioavailability. However, conventional MOs is production involves complex processing steps and significant production costs. A potential high-efficiency synthesis of specific MOs can be achieved through the ring-opening reaction of cyclodextrins (CDs) catalyzed by amylolytic enzymes. In this study, we analyze the catalytic conversion of α-, β-, and γ-CDs by a GH57 family amylopullulanase from Aquifex aeolicus (AaApu) using thin-layer chromatography (TLC). Our findings demonstrate that AaApu has a substrate specificity for γ-CD, while all three CDs exert competitive inhibition on pullulan hydrolysis. To elucidate the molecular mechanism of CDs as inhibitor and substrate of amylopullulanase, we determined high-resolution crystal structures of AaApu (wild-type and D352N) in complex with α-, β-, and γ-CD through co-crystallization. These findings establish a structure-function framework for understanding the bifunctional nature of CDs as both substrates and inhibitors in GH57 amylopullulanases.
Collapse
Affiliation(s)
- Zhimin Zhu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Qin Xu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Liqing Huang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huan Zhou
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Weiwei Wang
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Qisheng Wang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Feng Yu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China; Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| |
Collapse
|
220
|
Hou C, Tsodikov OV. Structure and cooperative formation of a FLI1 filament on contiguous GGAA DNA sites. Nucleic Acids Res 2025; 53:gkaf205. [PMID: 40131773 PMCID: PMC11934925 DOI: 10.1093/nar/gkaf205] [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: 07/18/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Ewing sarcoma, a pediatric cancer of bone and soft tissue, is driven in most cases by an abnormal oncogenic fusion of the N-terminal region of EWS with the C-terminal region of FLI1 (EWS-FLI1). The FLI1 region contains a conserved DNA-binding domain (DBD) essential for the oncogenesis. Binding of EWS-FLI1 to microsatellites composed of contiguous GGAA sites, shown previously to be critical for the oncogenic program of this fusion, is not well understood. In this study, we demonstrate that the FLI1 DBD binds cooperatively to contiguous GGAA sites, thereby forming a nucleoprotein filament. A series of crystal structures of two, three, and four FLI1 DBD proteins in complexes with DNA oligomers containing two, three, and four contiguous GGAA sites, respectively, reveal the structure of this filament and the basis for its cooperative formation. We expect this mechanistic insight to be an important milestone in our understanding of the oncogenic function of EWS-FLI1 and exploiting it as a drug target.
Collapse
Affiliation(s)
- Caixia Hou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, United States
| | - Oleg V Tsodikov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, United States
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, United States
| |
Collapse
|
221
|
Warren GM, Shuman S. In vivo nucleotide excision repair by mycobacterial UvrD1 requires ATP hydrolysis but does not depend on cysteine disulfide-mediated dimerization and DNA unwinding. Nucleic Acids Res 2025; 53:gkaf269. [PMID: 40193706 PMCID: PMC11975281 DOI: 10.1093/nar/gkaf269] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Mycobacterial UvrD1 is an SF1-type ATPase that participates in nucleotide excision repair (NER). UvrD1 consists of N-terminal ATPase and C-terminal Tudor domains. The monomeric UvrD1 characterized originally displays vigorous DNA-dependent ATPase activity but only feeble helicase activity. A recent study demonstrated that: (i) cysteine disulfide-mediated homodimerization of UvrD1 generates a highly active helicase; and (ii) an obligate monomeric UvrD1 (by virtue of mutating the domain 2B cysteine) is active as an ATP-dependent 3'-to-5' single-stranded DNA translocase but not as a double-stranded DNA-unwinding helicase. Here we test genetically which physical and functional states of UvrD1 are relevant for its functions in DNA repair, by complementation of an NER-defective Mycobacterium smegmatis ΔuvrD1 strain with a series of biochemically-defined UvrD1 mutants. By assaying complemented strains for sensitivity to UVC, MMC, cisplatin, and psoralen-UVA, we conclude that monomeric UvrD1 ATPase activity suffices for the NER functions of UvrD1 in vivo. Decoupling ATP hydrolysis from duplex unwinding does not affect the repair activity of UvrD1, nor does interdiction of domain 2B cysteine disulfide-mediated dimerization or deletion of the Tudor domain. Our results militate against a proposed model in which UvrD1's repair function is governed by the redox state of the bacterium via its impact on UvrD1 dimerization and helicase activity.
Collapse
Affiliation(s)
- Garrett M Warren
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, NY, NY 10065, United States
| | - Stewart Shuman
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, NY, NY 10065, United States
| |
Collapse
|
222
|
Chiang CC, Lu YJ, Liu JW, Lin SW, Chou CC, Lin CH, Chien IW, Hsu CH. Structural Insights into 4,5-DOPA Extradiol Dioxygenase from Beta vulgaris: Unraveling the Key Step in Versatile Betalain Biosynthesis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6785-6794. [PMID: 40055856 PMCID: PMC11926856 DOI: 10.1021/acs.jafc.4c09501] [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] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Betalains, a group of pigments widely distributed in various plants, are extensively applied in the food, beverage, and medicinal industries. The biosynthesis of betalains involves the enzymatic action of 4,5-DOPA-dioxygenase, which catalyzes the key ring-opening reaction of DOPA to produce betalamic acid, a crucial intermediate in the pathway. The crystal structure of a 4,5-DOPA-dioxygenase from Beta vulgaris (BvDOD) was determined in this study. The structural analysis revealed that BvDOD exhibited a structural fold similar to that of other members of the extradiol dioxygenase family. Moreover, the Fe-ligand residues His15, His53, and His229 indicated the enzyme's reliance on nonheme iron for catalyzing the ring-opening reaction. Molecular docking and mutational analysis identified two conserved residues, His119 and His175, in the active site essential for the catalytic reaction. In addition, Thr17, Asp254, and Tyr260 contributed to properly positioning the substrate in the active site. This study has provided structural insights into substrate recognition and catalytic mechanisms of BvDOD, which can be applied to develop enzymes for improved betalain production.
Collapse
Affiliation(s)
- Chih-Chia Chiang
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
| | - Yen-Ju Lu
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Jia-Wei Liu
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
| | - Sheng-Wei Lin
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Chun-Chi Chou
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
| | - Chia-Hsin Lin
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - I-Weh Chien
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
| | - Chun-Hua Hsu
- Department
of Agricultural Chemistry, National Taiwan
University, Taipei 10617, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
- Genome
and
Systems Biology Degree Program, National
Taiwan University and Academia Sinica, Taipei 10617, Taiwan
- Center for
Computational and Systems Biology, National
Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
223
|
Chen Y, Bhattacharya S, Bergmann L, Correy GJ, Tan S, Hou K, Biel J, Lu L, Bakanas I, Polizzi NF, Fraser JS, DeGrado WF. Emergence of specific binding and catalysis from a designed generalist binding protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635804. [PMID: 39975260 PMCID: PMC11838529 DOI: 10.1101/2025.01.30.635804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The evolution of binding and catalysis played a central role in the emergence of life. While natural proteins have finely tuned affinities for their primary ligands, they also bind weakly and promiscuously to other molecules, which serve as starting points for stepwise, incremental evolution of entirely new specificities. Thus, modern proteins emerged from the joint exploration of sequence and structural space. The ability of natural proteins to bind promiscuously to small molecule fragments has been widely evaluated using methods including crystallographic fragment screening. However, this approach had not been applied to de novo proteins. Here, we apply this method to explore the promiscuity of a de novo small molecule-binding protein ABLE. As in Nature, we found ABLE was capable of forming weak complexes, which were found to be excellent starting points for evolving entirely new functions, including a binder of a turn-on fluorophore and a highly efficient and specific Kemp eliminase enzyme. This work shows how Nature and protein designers can take advantage of promiscuous binding interactions to evolve new proteins with specialized functions.
Collapse
Affiliation(s)
- Yuda Chen
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Sagar Bhattacharya
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Lena Bergmann
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Sophia Tan
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Kaipeng Hou
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Justin Biel
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Lei Lu
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Ian Bakanas
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Nicholas F. Polizzi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
224
|
Nagano S, Song C, Rohr V, Mackintosh MJ, Hoang OT, Kraskov A, Yang Y, Hughes J, Heyne K, Mroginski MA, Schapiro I, Hildebrandt P. Integrated Study of Fluorescence Enhancement in the Y176H Variant of Cyanobacterial Phytochrome Cph1. Biochemistry 2025; 64:1348-1358. [PMID: 40015976 PMCID: PMC11924222 DOI: 10.1021/acs.biochem.4c00687] [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: 10/14/2024] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 03/01/2025]
Abstract
Phytochromes are red-light-sensitive biliprotein photoreceptors that control a variety of physiological processes in plants, fungi, and bacteria. Lately, greater attention has been paid to these photoreceptors due to their potential as fluorescent probes for deep-tissue microscopy. Such fluorescing phytochromes have been generated by multiple amino acid substitutions in weakly fluorescent wild-type (WT) proteins. Remarkably, the single substitution of conserved Tyr176 by His in cyanobacterial phytochrome Cph1 increases the fluorescence quantum yield from 2.4 to 14.5%. In this work, we studied this Y176H variant by crystallography, MAS NMR, resonance Raman spectroscopy, and ultrafast absorption spectroscopy complemented by theoretical methods. Two factors were identified to account for the strong fluorescence increase. First, the equilibrium between the photoactive and fluorescent substates of WT Cph1 was shown to shift entirely to the fluorescent substate in Y176H. Second, structural flexibility of the chromophore is drastically reduced and the photoisomerization barrier is raised, thereby increasing the excited-state lifetime. The most striking finding, however, is that Y176H includes the structural properties of both the dark-adapted Pr and the light-activated Pfr state. While the chromophore adopts the Pr-typical ZZZssa configuration, the tongue segment of the protein adopts a Pfr-typical α-helical structure. This implies that Tyr176 plays a key role in coupling chromophore photoisomerization to the sheet-to-helix transition of the tongue and the final Pfr structure. This conclusion extends to plant phytochromes, where the homologous substitution causes light-independent signaling activity akin to that of Pfr.
Collapse
Affiliation(s)
- Soshichiro Nagano
- Institute
for Plant Physiology, Justus Liebig University, Senckenbergstr. 3, Giessen D-35390, Germany
| | - Chen Song
- Institute
for Analytical Chemistry, University of
Leipzig, Johannisallee 29, Leipzig D-04103, Germany
| | - Valentin Rohr
- Institute
for Analytical Chemistry, University of
Leipzig, Johannisallee 29, Leipzig D-04103, Germany
| | - Megan J. Mackintosh
- Fritz Haber
Center for Molecular Dynamics, Institute of Chemistry, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Oanh Tu Hoang
- Institute
for Chemistry, Technical University of Berlin, Str. des 17. Juni 135, Berlin D-10623, Germany
| | - Anastasia Kraskov
- Institute
for Chemistry, Technical University of Berlin, Str. des 17. Juni 135, Berlin D-10623, Germany
| | - Yang Yang
- Department
of Physics, Free University of Berlin, Arnimallee 14, Berlin D-14195, Germany
| | - Jon Hughes
- Institute
for Plant Physiology, Justus Liebig University, Senckenbergstr. 3, Giessen D-35390, Germany
- Department
of Physics, Free University of Berlin, Arnimallee 14, Berlin D-14195, Germany
| | - Karsten Heyne
- Department
of Physics, Free University of Berlin, Arnimallee 14, Berlin D-14195, Germany
| | - Maria-Andrea Mroginski
- Institute
for Chemistry, Technical University of Berlin, Str. des 17. Juni 135, Berlin D-10623, Germany
| | - Igor Schapiro
- Fritz Haber
Center for Molecular Dynamics, Institute of Chemistry, Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Peter Hildebrandt
- Institute
for Chemistry, Technical University of Berlin, Str. des 17. Juni 135, Berlin D-10623, Germany
| |
Collapse
|
225
|
Patel PN, Diouf A, Dickey TH, Tang WK, Hopp CS, Traore B, Long CA, Miura K, Crompton PD, Tolia NH. A strain-transcending anti-AMA1 human monoclonal antibody neutralizes malaria parasites independent of direct RON2L receptor blockade. Cell Rep Med 2025; 6:101985. [PMID: 40020675 PMCID: PMC11970402 DOI: 10.1016/j.xcrm.2025.101985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Plasmodium falciparum apical membrane antigen 1 (AMA1) binds a loop in rhoptry neck protein 2 (RON2L) during red cell invasion and is a target for vaccines and therapeutic antibodies against malaria. Here, we report a panel of AMA1-specific naturally acquired human monoclonal antibodies (hmAbs) derived from individuals living in malaria-endemic regions. Two neutralizing hmAbs engage AMA1 independent of the RON2L-binding site. The hmAb 75B10 demonstrates potent strain-transcending neutralization that is independent of RON2L blockade, emphasizing that epitopes outside the RON2L-binding site elicit broad protection against variant parasite strains. The combination of these hmAbs synergistically enhances parasite neutralization. Vaccination with a structure-based design (SBD1) that mimics the AMA1-RON2L complex elicited antibodies similar to the two neutralizing hmAbs connecting vaccination to naturally acquired immunity in humans. The structural definition of a strain-transcending epitope on AMA1 targeted by naturally acquired hmAb establishes paradigms for developing AMA1-based vaccines and therapeutic antibodies.
Collapse
Affiliation(s)
- Palak N Patel
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Thayne H Dickey
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wai Kwan Tang
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christine S Hopp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Boubacar Traore
- Malaria Research and Training Centre, Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Point G, Bamako 1805, Mali
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Niraj H Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
226
|
Rivas-Fernández JP, Vuillemin M, Pilgaard B, Klau LJ, Fredslund F, Lund-Hanssen C, Welner DH, Meyer AS, Morth JP, Meilleur F, Aachmann FL, Rovira C, Wilkens C. Unraveling the molecular mechanism of polysaccharide lyases for efficient alginate degradation. Nat Commun 2025; 16:2670. [PMID: 40102416 PMCID: PMC11920209 DOI: 10.1038/s41467-025-56754-5] [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: 01/15/2024] [Accepted: 01/29/2025] [Indexed: 03/20/2025] Open
Abstract
Alginate lyases (ALs) catalyze the depolymerization of brown macroalgae alginates, widely used naturally occurring polysaccharides. Their molecular reaction mechanism remains elusive due to the lack of catalytically competent Michaelis-Menten-like complex structures. Here, we provide structural snapshots and dissect the mechanism of mannuronan-specific ALs from family 7 polysaccharide lyases (PL7), employing time-resolved NMR, X-ray, neutron crystallography, and QM/MM simulations. We reveal the protonation state of critical active site residues, enabling atomic-level analysis of the reaction coordinate. Our approach reveals an endolytic and asynchronous syn β-elimination reaction, with Tyr serving as both Brønsted base and acid, involving a carbanion-type transition state. This study not only reconciles previous structural and kinetic discrepancies, but also establishes a comprehensive PL reaction mechanism which is most likely applicable across all enzymes of the PL7 family as well as other PL families.
Collapse
Affiliation(s)
- José Pablo Rivas-Fernández
- Departament de Química Inorgànica i Orgànica (secció de Química Orgànica) & IQTCUB, Universitat de Barcelona, Barcelona, Spain
| | - Marlene Vuillemin
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Bo Pilgaard
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Leesa J Klau
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- SINTEF Industry, Oslo, Norway
| | - Folmer Fredslund
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
- Novo Nordisk A/S, Gentofte, Denmark
| | - Charlotte Lund-Hanssen
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Ditte H Welner
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Anne S Meyer
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - J Preben Morth
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Flora Meilleur
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, USA
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Finn L Aachmann
- Norwegian Biopolymer Laboratory (NOBIPOL), Department of Biotechnology and Food Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Carme Rovira
- Departament de Química Inorgànica i Orgànica (secció de Química Orgànica) & IQTCUB, Universitat de Barcelona, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluís Companys, Barcelona, Spain.
| | - Casper Wilkens
- Section for Protein Chemistry and Enzyme Technology, Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
- Novo Nordisk Pharmatech A/S, Køge, Denmark.
| |
Collapse
|
227
|
Nix MN, Gourisankar S, Sarott RC, Dwyer BG, Nettles SA, Martinez MM, Abuzaid H, Yang H, Wang Y, Simanauskaite JM, Romero BA, Jones HM, Krokhotin A, Lowensohn TN, Chen L, Low C, Davis MM, Fernandez D, Zhang T, Green MR, Hinshaw SM, Gray NS, Crabtree GR. A Bivalent Molecular Glue Linking Lysine Acetyltransferases to Oncogene-induced Cell Death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643404. [PMID: 40166243 PMCID: PMC11956963 DOI: 10.1101/2025.03.14.643404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Developing cancer therapies that induce robust death of the malignant cell is critical to prevent relapse. Highly effective strategies, such as immunotherapy, exemplify this observation. Here we provide the structural and molecular underpinnings for an approach that leverages chemical induced proximity to produce specific cell killing of diffuse large B cell lymphoma, the most common non-Hodgkin's lymphoma. We develop KAT-TCIPs (lysine acetyltransferase transcriptional/epigenetic chemical inducers of proximity) that redirect p300 and CBP to activate programmed cell death genes normally repressed by the oncogenic driver, BCL6. Acute treatment rapidly reprograms the epigenome to initiate apoptosis and repress c-MYC. The crystal structure of the chemically induced p300-BCL6 complex reveals how chance interactions between the two proteins can be systematically exploited to produce the exquisite potency and selectivity of KAT-TCIPs. Thus, the malignant function of an oncogenic driver can be co-opted to activate robust cell death, with implications for precision epigenetic therapies.
Collapse
Affiliation(s)
- Meredith N. Nix
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sai Gourisankar
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Roman C. Sarott
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Brendan G. Dwyer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | - Michael M. Martinez
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Hind Abuzaid
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Haopeng Yang
- Department of Lymphoma- & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanlan Wang
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Bryan A. Romero
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Hannah M. Jones
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | | | | | - Lei Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Cara Low
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Mark M. Davis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Daniel Fernandez
- Macromolecular Structure, Nucleus at Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Michael R. Green
- Department of Lymphoma- & Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen M. Hinshaw
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Gerald R. Crabtree
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
| |
Collapse
|
228
|
Bosco J, Gagliano E, Boshae KL, Statz JP, Wheeler TB, Cuello D, Sliter A, Newby C, Lin B, Demeler A, Pierpont CL, Yates-Hansen C, Sydor MJ, Ferrini ME, Kuch KC, Cooper BS, Piggott BJ, Certel SJ, Hansen KB, Sprang SR, Bowler B, McClelland L, Berkmen M, Voronina E. A galactose-based auto-expression system improves T7-inducible protein production in Escherichia coli. Sci Rep 2025; 15:8936. [PMID: 40089537 PMCID: PMC11910606 DOI: 10.1038/s41598-025-91954-5] [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: 10/10/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
Protein production using Escherichia coli is a cornerstone of modern biotechnology. In this study, we developed a novel auto-expression medium to maximize protein production. Each E. coli strain tested was capable of auto-expression in response to galactose, including strains in which the endogenous lacZ had been disrupted. This provides key evidence that galactose can regulate the lac operon independent of known lac operon-regulated metabolism. The enhanced capabilities of the novel auto-expression medium were documented across protein production systems including (1) increased yields for routinely expressed proteins (e.g. eGFP), (2) improved expression of human cytochrome c within a dual expression system, (3) robust auto-expression in lacZ-deficient strains producing proteins with challenging disulfide bonds, and (4) reproducible 8-fold increase in SpCas9 yields, at ≥ 95% purity. This novel medium can streamline production and improve yields for routine as well as challenging proteins, accelerating recombinant protein production and creating new opportunities in biotechnology and structural biology.
Collapse
Affiliation(s)
- James Bosco
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA.
| | - Emily Gagliano
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kassandra L Boshae
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - John P Statz
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Timothy B Wheeler
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - DeAnna Cuello
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Ashlyn Sliter
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Christian Newby
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Bernice Lin
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Aysha Demeler
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - C Logan Pierpont
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Cindee Yates-Hansen
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Matthew J Sydor
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Maria E Ferrini
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kellie C Kuch
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Brandon S Cooper
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Beverly J Piggott
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Sarah J Certel
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Kasper B Hansen
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Stephen R Sprang
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Bruce Bowler
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Levi McClelland
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Mehmet Berkmen
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Ekaterina Voronina
- Division of Biological Sciences, Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Center for Environmental Health Sciences, University of Montana, Missoula, MT, 59812, USA.
| |
Collapse
|
229
|
Williford E, Xue YP, Tang WK, Li R, Jones KV, Blake KS, Blaine HC, Lian X, Stallings CL, Tolia NH, Dantas G, Wencewicz TA. C10-Benzoate Esters of Anhydrotetracycline Inhibit Tetracycline Destructases and Recover Tetracycline Antibacterial Activity. ACS Infect Dis 2025; 11:738-749. [PMID: 39912785 PMCID: PMC11915366 DOI: 10.1021/acsinfecdis.4c00912] [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: 11/15/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/07/2025]
Abstract
Tetracyclines (TCs) are an important class of antibiotics threatened by enzymatic inactivation. These tetracycline-inactivating enzymes, also known as tetracycline destructases (TDases), are a subfamily of class A flavin monooxygenases (FMOs) that catalyze hydroxyl group transfer and oxygen insertion (Baeyer-Villiger type) reactions on TC substrate scaffolds. Semisynthetic modification of TCs (e.g., tigecycline, omadacycline, eravacycline, and sarecycline) has proven effective in evading certain resistance mechanisms, such as ribosomal protection and efflux, but does not protect against TDase-mediated resistance. Here, we report the design, synthesis, and evaluation of a new series of 22 semisynthetic TDase inhibitors that explore D-ring substitution of anhydrotetracycline (aTC) including 14 C10-benzoate ester and eight C9-benzamides. Overall, the C10-benzoate esters displayed enhanced bioactivity and water solubility compared to the corresponding C9-benzamides featuring the same heterocyclic aryl side chains. The C10-benzoate ester derivatives of aTC were prepared in a high-yield one-step synthesis without the need for protecting groups. The C10-esters are water-soluble, stable toward hydrolysis, and display dose-dependent rescue of tetracycline antibiotic activity in E. coli expressing two types of tetracycline destructases, represented by TetX7 (Type 1) and Tet50 (Type 2). The best inhibitors recovered tetracycline antibiotic activity at concentrations as low as 2 μM, producing synergistic scores <0.5 in the fractional inhibitory concentration index (FICI) against TDase-expressing strains of E. coli and clinical P. aeruginosa. The C10-benzoate ester derivatives of aTC reported here are promising new leads for the development of tetracycline drug combination therapies to overcome TDase-mediated antibiotic resistance.
Collapse
Affiliation(s)
- Emily
E. Williford
- Department
of Chemistry, Washington University in St.
Louis, St. Louis, Missouri 63130, United States
| | - Yao-Peng Xue
- The
Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
| | - Wai Kwan Tang
- Host−Pathogen
Interactions and Structural Vaccinology Section, Laboratory of Malaria
Immunology and Vaccinology, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20814, United States
| | - Ruihao Li
- Department
of Chemistry, Washington University in St.
Louis, St. Louis, Missouri 63130, United States
| | - Katherine V. Jones
- Department
of Chemistry, Washington University in St.
Louis, St. Louis, Missouri 63130, United States
| | - Kevin S. Blake
- The
Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
- Department
of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Helen C. Blaine
- Department
of Molecular Microbiology, Washington University
School of Medicine, St. Louis, Missouri 63110, United States
| | - Xiang Lian
- Department
of Chemistry, Washington University in St.
Louis, St. Louis, Missouri 63130, United States
| | - Christina L. Stallings
- Department
of Molecular Microbiology, Washington University
School of Medicine, St. Louis, Missouri 63110, United States
| | - Niraj H. Tolia
- Host−Pathogen
Interactions and Structural Vaccinology Section, Laboratory of Malaria
Immunology and Vaccinology, National Institute of Allergy and Infectious
Diseases, National Institutes of Health, Bethesda, Maryland 20814, United States
| | - Gautam Dantas
- The
Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, Missouri 63108, United States
- Department
of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Molecular Microbiology, Washington University
School of Medicine, St. Louis, Missouri 63110, United States
- Department
of Biomedical Engineering, Washington University
in St. Louis, St. Louis, Missouri 63130, United States
- Department
of Pediatrics, Washington University School
of Medicine, St. Louis, Missouri 63110, United States
| | - Timothy A. Wencewicz
- Department
of Chemistry, Washington University in St.
Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
230
|
Ashley B, Mathew S, Sajjad M, Zhu Y, Novikovs N, Baslé A, Marles-Wright J, Campopiano DJ. Rational engineering of a thermostable α-oxoamine synthase biocatalyst expands the substrate scope and synthetic applicability. Commun Chem 2025; 8:78. [PMID: 40082705 PMCID: PMC11906848 DOI: 10.1038/s42004-025-01448-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Carbon-carbon bond formation is one of the key pillars of organic synthesis. Green, selective and efficient biocatalytic methods for such are therefore highly desirable. The α-oxoamine synthases (AOSs) are a class of pyridoxal 5'-phosphate (PLP)-dependent, irreversible, carbon-carbon bond-forming enzymes, which have been limited previously by their narrow substrate specificity and requirement of acyl-CoA thioester substrates. We recently characterized a thermophilic enzyme from Thermus thermophilus (ThAOS) with a much broader substrate scope and described its use in a chemo-biocatalytic cascade process to generate pyrroles in good yields and timescales. Herein, we report the structure-guided engineering of ThAOS to arrive at variants able to use a greatly expanded range of amino acid and simplified N-acetylcysteamine (SNAc) acyl-thioester substrates. The crystal structure of the improved ThAOS V79A variant with a bound PLP:L-penicillamine external aldimine ligand, provides insight into the properties of the engineered biocatalyst.
Collapse
Affiliation(s)
- Ben Ashley
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK
| | - Sam Mathew
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK
| | - Mariyah Sajjad
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK
| | - Yaoyi Zhu
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK
| | - Nikita Novikovs
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK
| | - Arnaud Baslé
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jon Marles-Wright
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dominic J Campopiano
- School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster Road, Edinburgh, UK.
| |
Collapse
|
231
|
Juarez MG, O'Rourke SM, Dzimianski JV, Gagnon D, Penunuri G, Serrão VHB, Corbett-Detig RB, Kauvar LM, DuBois RM. Structures of respiratory syncytial virus G bound to broadly reactive antibodies provide insights into vaccine design. Sci Rep 2025; 15:8666. [PMID: 40082629 PMCID: PMC11906780 DOI: 10.1038/s41598-025-92886-w] [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: 11/03/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease in infants and older adults. The attachment glycoprotein (RSV G) binds to the chemokine receptor CX3CR1 to promote viral entry and modulate host immunity. Antibodies against RSV G are a known correlate of protection. Previously, several broadly reactive, high-affinity anti-RSV G human monoclonal antibodies were isolated from RSV-exposed individuals and were shown to be protective in vitro and in vivo. Here, we determined the structures of three of these antibodies in complex with RSV G and defined distinct conformational epitopes comprised of highly conserved RSV G residues. Binding competition and structural studies demonstrated that this highly conserved region displays two non-overlapping antigenic sites. Analyses of anti-RSV G antibody sequences reveal that antigenic site flexibility may promote the elicitation of diverse antibody germlines. Together, these findings provide a foundation for next-generation RSV prophylactics, and they expand concepts in vaccine design for the elicitation of germline lineage-diverse, broadly reactive, high-affinity antibodies.
Collapse
Affiliation(s)
- Maria G Juarez
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Sara M O'Rourke
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - John V Dzimianski
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Delia Gagnon
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Gabriel Penunuri
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Vitor H B Serrão
- Department of Chemistry & Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
- Biomolecular Cryo-Electron Microscopy Facility, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Russell B Corbett-Detig
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | | | - Rebecca M DuBois
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
232
|
Kaneshige A, Yang Y, Bai L, Wang M, Xu R, Mallik L, Chinnaswamy K, Metwally H, Wang Y, McEachern D, Tošović J, Yang CY, Kirchhoff PD, Meagher JL, Stuckey JA, Wang S. Discovery of AK-1690: A Potent and Highly Selective STAT6 PROTAC Degrader. J Med Chem 2025; 68:5125-5151. [PMID: 39311434 DOI: 10.1021/acs.jmedchem.4c01009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
STAT6 is an attractive therapeutic target for human cancers and other human diseases. Starting from a STAT6 ligand with Ki = 3.5 μM binding affinity, we obtained AK-068 with Ki = 6 nM to STAT6 and at least >85-fold binding selectivity over STAT5. Using AK-068 and cereblon ligands, we discovered AK-1690 as the first, potent and selective PROTAC STAT6 degrader. AK-1690 effectively induces degradation of STAT6 protein in cells with DC50 values of as low as 1 nM while showing minimal effect on other STAT members up to 10 μM. A single dose of AK-1690 effectively depletes STAT6 in mouse tissues. Determination of the first cocrystal structure of STAT6 in complex with AK-1690 provides a structural basis for their interactions. AK-1690 is a powerful tool with which to investigate the roles of STAT6 in human diseases and biological processes and a promising lead compound for further optimization.
Collapse
Affiliation(s)
- Atsunori Kaneshige
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yiqing Yang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mi Wang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Renqi Xu
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Leena Mallik
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Hoda Metwally
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jelena Tošović
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chao-Yie Yang
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul D Kirchhoff
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jennifer L Meagher
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne A Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
233
|
Pérez Carrillo V, Whittaker JJ, Wiedemann C, Harder JM, Lohr T, Jamithireddy AK, Dajka M, Goretzki B, Joseph B, Guskov A, Harmer NJ, Holzgrabe U, Hellmich UA. Structure and Dynamics of Macrophage Infectivity Potentiator Proteins from Pathogenic Bacteria and Protozoans Bound to Fluorinated Pipecolic Acid Inhibitors. J Med Chem 2025; 68:5926-5941. [PMID: 39976355 PMCID: PMC11912469 DOI: 10.1021/acs.jmedchem.5c00134] [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: 01/15/2025] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 02/21/2025]
Abstract
Macrophage infectivity potentiator (MIP) proteins, found in pro- and eukaryotic pathogens, influence microbial virulence, host cell infection, pathogen replication, and dissemination. MIPs share an FKBP (FK506 binding protein)-like prolyl-cis/trans-isomerase domain, making them attractive targets for inhibitor development. We determined high-resolution crystal structures of Burkholderia pseudomallei and Trypanosoma cruzi MIPs in complex with fluorinated pipecolic acid inhibitors. The inhibitor binding profiles in solution were compared across B. pseudomallei, T. cruzi, and Legionella pneumophila MIPs using 1H, 15N, and 19F NMR spectroscopy. Demonstrating the versatility of fluorinated ligands for characterizing inhibitor complexes, 19F NMR spectroscopy identified differences in ligand binding dynamics across MIPs. EPR spectroscopy and SAXS further revealed inhibitor-induced global structural changes in homodimeric L. pneumophila MIP. This study demonstrates the importance of integrating diverse methods to probe protein dynamics and provides a foundation for optimizing MIP-targeted inhibitors in this structurally conserved yet dynamically variable protein family.
Collapse
Affiliation(s)
- Victor
Hugo Pérez Carrillo
- Faculty
of Chemistry and Earth Sciences, Institute of Organic Chemistry and
Macromolecular Chemistry, Friedrich Schiller
University Jena, 07743 Jena, Germany
| | - Jacob J. Whittaker
- Groningen
Institute for Biomolecular Sciences and Biotechnology, University of Groningen, 9747AG Groningen, The Netherlands
| | - Christoph Wiedemann
- Faculty
of Chemistry and Earth Sciences, Institute of Organic Chemistry and
Macromolecular Chemistry, Friedrich Schiller
University Jena, 07743 Jena, Germany
| | - Jean-Martin Harder
- Faculty
of Chemistry and Earth Sciences, Institute of Organic Chemistry and
Macromolecular Chemistry, Friedrich Schiller
University Jena, 07743 Jena, Germany
| | - Theresa Lohr
- Institute
of Pharmacy and Food Chemistry, University
of Würzburg, Am
Hubland, 97074 Würzburg, Germany
| | - Anil K. Jamithireddy
- Living
Systems Institute, University of Exeter, Stocker Road, EX4 4QD Exeter, U.K.
| | - Marina Dajka
- Department
of Physics, Free University of Berlin, 14195 Berlin, Germany
| | - Benedikt Goretzki
- Faculty
of Chemistry and Earth Sciences, Institute of Organic Chemistry and
Macromolecular Chemistry, Friedrich Schiller
University Jena, 07743 Jena, Germany
- Center for
Biomolecular Magnetic Resonance, Goethe-University, 60438 Frankfurt/Main, Germany
| | - Benesh Joseph
- Department
of Physics, Free University of Berlin, 14195 Berlin, Germany
| | - Albert Guskov
- Groningen
Institute for Biomolecular Sciences and Biotechnology, University of Groningen, 9747AG Groningen, The Netherlands
| | - Nicholas J. Harmer
- Living
Systems Institute, University of Exeter, Stocker Road, EX4 4QD Exeter, U.K.
| | - Ulrike Holzgrabe
- Institute
of Pharmacy and Food Chemistry, University
of Würzburg, Am
Hubland, 97074 Würzburg, Germany
| | - Ute A. Hellmich
- Faculty
of Chemistry and Earth Sciences, Institute of Organic Chemistry and
Macromolecular Chemistry, Friedrich Schiller
University Jena, 07743 Jena, Germany
- Center for
Biomolecular Magnetic Resonance, Goethe-University, 60438 Frankfurt/Main, Germany
- Cluster
of Excellence “Balance of the Microverse”, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
234
|
Ewert W, Bartens C, Ongouta J, Holmes M, Heutling A, Kishore A, Urbansky T, Zeilinger C, Preller M, Kirschning A. Structure and function of the geldanamycin amide synthase from Streptomyces hygroscopicus. Nat Commun 2025; 16:2464. [PMID: 40075103 PMCID: PMC11903869 DOI: 10.1038/s41467-025-57013-3] [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: 06/19/2023] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Amide synthases catalyze the formation of macrolactam rings from aniline-containing polyketide-derived seco-acids as found in the important class of ansamycin antibiotics. One of these amide synthases is the geldanamycin amide synthase GdmF, which we recombinantly expressed, purified and studied in detail both functionally as well as structurally. Here we show that purified GdmF catalyzes the amide formation using synthetically derived substrates. The atomic structures of the ligand-free enzyme and in complex with simplified substrates reveal distinct structural features of the substrate binding site and a putative role of the flexible interdomain region for the catalysis reaction.
Collapse
Affiliation(s)
- Wiebke Ewert
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Christian Bartens
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Jekaterina Ongouta
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Monika Holmes
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anja Heutling
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Anusha Kishore
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany
| | - Tim Urbansky
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany
| | - Carsten Zeilinger
- Center of Biomolecular Drug Research (BMWZ) Leibniz University Hannover, Hannover, Germany.
| | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany.
- Institute for Functional Gene Analytics (IFGA), University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach, Germany.
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University Hannover, Hannover, Germany.
- Uppsala Biomedical Center (BMC), University Uppsala, Uppsala, Sweden.
| |
Collapse
|
235
|
Nangle LA, Xu Z, Siefker D, Burkart C, Chong YE, Zhai L, Geng Y, Polizzi C, Guy L, Eide L, Tong Y, Klopp-Savino S, Ferrer M, Rauch K, Wang A, Hamel K, Crampton S, Paz S, Chiang KP, Do MH, Burman L, Lee D, Zhang M, Ogilvie K, King D, Adams RA, Schimmel P. A human histidyl-tRNA synthetase splice variant therapeutic targets NRP2 to resolve lung inflammation and fibrosis. Sci Transl Med 2025; 17:eadp4754. [PMID: 40073151 DOI: 10.1126/scitranslmed.adp4754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/24/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Interstitial lung disease (ILD) consists of a group of immune-mediated disorders that can cause inflammation and progressive fibrosis of the lungs, representing an area of unmet medical need given the lack of disease-modifying therapies and toxicities associated with current treatment options. Tissue-specific splice variants (SVs) of human aminoacyl-tRNA synthetases (aaRSs) are catalytic nulls thought to confer regulatory functions. One example from human histidyl-tRNA synthetase (HARS), termed HARSWHEP because the splicing event resulted in a protein encompassing the WHEP-TRS domain of HARS (a structurally conserved domain found in multiple aaRSs), is enriched in human lung and up-regulated by inflammatory cytokines in lung and immune cells. Structural analysis of HARSWHEP confirmed a well-organized helix-turn-helix motif. This motif bound specifically and selectively to neuropilin-2 (NRP2), a receptor expressed by myeloid cells in active sites of inflammation, to inhibit expression of proinflammatory receptors and cytokines and to down-regulate inflammatory pathways in primary human macrophages. In animal models of lung injury and ILD, including bleomycin treatment, silicosis, sarcoidosis, chronic hypersensitivity pneumonitis, systemic sclerosis, and rheumatoid arthritis-ILD, HARSWHEP reduced lung inflammation, immune cell infiltration, and fibrosis. In patients with sarcoidosis, efzofitimod treatment resulted in down-regulation of gene expression for inflammatory pathways in peripheral immune cells and stabilization of inflammatory biomarkers in serum after steroid tapering. We demonstrate the immunomodulatory activity of HARSWHEP and present preclinical data supporting ongoing clinical development of the biologic efzofitimod based on HARSWHEP in ILD.
Collapse
Affiliation(s)
| | - Zhiwen Xu
- aTyr Pharma, San Diego, CA 92121, USA
| | | | | | | | - Liting Zhai
- IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Pangu Biopharma, Hong Kong, China
| | - Yanyan Geng
- IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Pangu Biopharma, Hong Kong, China
| | | | | | - Lisa Eide
- aTyr Pharma, San Diego, CA 92121, USA
| | - Yao Tong
- IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Pangu Biopharma, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | | - Darin Lee
- aTyr Pharma, San Diego, CA 92121, USA
| | - Mingjie Zhang
- IAS HKUST-Scripps R&D Laboratory, Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | | | | - Paul Schimmel
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
236
|
Fadini A, Li M, McCoy AJ, Terwilliger TC, Read RJ, Hekstra D, AlQuraishi M. AlphaFold as a Prior: Experimental Structure Determination Conditioned on a Pretrained Neural Network. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638828. [PMID: 40027838 PMCID: PMC11870471 DOI: 10.1101/2025.02.18.638828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Advances in machine learning have transformed structural biology, enabling swift and accurate prediction of protein structure from sequence. However, challenges persist in capturing sidechain packing, condition-dependent conformational dynamics, and biomolecular interactions, primarily due to scarcity of high-quality training data. Emerging techniques, including cryo-electron tomography (cryo-ET) and high-throughput crystallography, promise vast new sources of structural data, but translating experimental observations into mechanistically interpretable atomic models remains a key bottleneck. Here, we address these challenges by improving the efficiency of structural analysis through combining experimental measurements with a landmark protein structure prediction method - AlphaFold2. We present an augmentation of AlphaFold2, ROCKET, that refines its predictions using cryo-EM, cryo-ET, and X-ray crystallography data, and demonstrate that this approach captures biologically important structural variation that AlphaFold2 does not. By performing structure optimization in the space of coevolutionary embeddings, rather than Cartesian coordinates, ROCKET automates difficult modeling tasks, such as flips of functional loops and domain rearrangements, that are beyond the scope of current state-of-the-art methods and, in some instances, even manual human modeling. The ability to efficiently sample these barrier-crossing rearrangements unlocks a new horizon for scalable and automated model building. Crucially, ROCKET does not require retraining of AlphaFold2 and is readily adaptable to multimers, ligand-cofolding, and other data modalities. Conversely, our differentiable crystal-lographic and cryo-EM target functions are capable of augmenting other structure prediction methods. ROCKET thus provides an extensible framework for the integration of experimental observables with biomolecular machine learning.
Collapse
Affiliation(s)
- Alisia Fadini
- Cambridge Institute for Medical Research, University of Cambridge
| | - Minhuan Li
- John A. Paulson School of Engineering & Applied Sciences, Harvard University
| | - Airlie J. McCoy
- Cambridge Institute for Medical Research, University of Cambridge
| | | | - Randy J. Read
- Cambridge Institute for Medical Research, University of Cambridge
| | - Doeke Hekstra
- John A. Paulson School of Engineering & Applied Sciences, Harvard University
- Department of Molecular and Cellular Biology, Harvard University
| | | |
Collapse
|
237
|
Masuyer G, Taverner A, MacKay J, Lima Marques AR, Wang Y, Hunter T, Liu K, Mrsny RJ. Discovery of mono-ADP ribosylating toxins with high structural homology to Pseudomonas exotoxin A. Commun Biol 2025; 8:413. [PMID: 40069285 PMCID: PMC11897225 DOI: 10.1038/s42003-025-07845-y] [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: 09/16/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
Mono-ADP-ribosyl transferase (mART) proteins are secreted virulence factors produced by several human pathogens, the founding member being diphtheria toxin (DT). Pseudomonas aeruginosa can also secrete a mART toxin, known as exotoxin A (PE), but with an organization of its three functional domains (receptor, translocation, and enzymatic elements) that is opposite to DT. Two additional PE-like toxins (PLTs) have been identified from Vibrio cholerae and Aeromonas hydrophila, suggesting more PLT family members may exist. Database mining discovered six additional putative homologues, considerably extending this group of PLTs across a wide range of bacterial species. Here, we examine sequence and structural information for these new family members with respect to previously identified PLTs. The X-ray crystal structures of four new homologues show the conservation of critical features responsible for structure and function. This study shows the potential of these newly described toxins for the development of novel drug delivery platforms. Additionally, genomic analysis suggests horizontal gene transfer to account for the wide distribution of PLTs across a range of eubacteria species, highlighting the need to monitor emerging pathogens and their virulence factors.
Collapse
Affiliation(s)
- Geoffrey Masuyer
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
- Department of Life Sciences, University of Bath, Bath, UK.
- Centre for Therapeutic Innovation, University of Bath, Bath, UK.
| | | | - Julia MacKay
- Department of Life Sciences, University of Bath, Bath, UK
| | | | - Yuye Wang
- Department of Life Sciences, University of Bath, Bath, UK
| | - Tom Hunter
- Applied Molecular Transport Inc., South San Francisco, CA, USA
| | - Keyi Liu
- Applied Molecular Transport Inc., South San Francisco, CA, USA
| | - Randall J Mrsny
- Department of Life Sciences, University of Bath, Bath, UK.
- Centre for Therapeutic Innovation, University of Bath, Bath, UK.
| |
Collapse
|
238
|
Rosen PC, Horwitz SM, Brooks DJ, Kim E, Ambarian JA, Waidmann L, Davis KM, Yellen G. State-dependent motion of a genetically encoded fluorescent biosensor. Proc Natl Acad Sci U S A 2025; 122:e2426324122. [PMID: 40048274 PMCID: PMC11912384 DOI: 10.1073/pnas.2426324122] [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: 12/16/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
Genetically encoded biosensors can measure biochemical properties such as small-molecule concentrations with single-cell resolution, even in vivo. Despite their utility, these sensors are "black boxes": Very little is known about the structures of their low- and high-fluorescence states or what features are required to transition between them. We used LiLac, a lactate biosensor with a quantitative fluorescence-lifetime readout, as a model system to address these questions. X-ray crystal structures and engineered high-affinity metal bridges demonstrate that LiLac exhibits a large interdomain twist motion that pulls the fluorescent protein away from a "sealed," high-lifetime state in the absence of lactate to a "cracked," low-lifetime state in its presence. Understanding the structures and dynamics of LiLac will help to think about and engineer other fluorescent biosensors.
Collapse
Affiliation(s)
- Paul C. Rosen
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | | | - Daniel J. Brooks
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Erica Kim
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | - Lidia Waidmann
- Department of Chemistry, Emory University, Atlanta, GA30322
| | | | - Gary Yellen
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| |
Collapse
|
239
|
Maurer DP, Vu M, Schmidt AG. Antigenic drift expands influenza viral escape pathways from recalled humoral immunity. Immunity 2025; 58:716-727.e6. [PMID: 40023162 PMCID: PMC11906258 DOI: 10.1016/j.immuni.2025.02.006] [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: 04/23/2024] [Revised: 11/16/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Initial exposure to a rapidly evolving virus establishes B cell memory that biases later responses to antigenically drifted strains. This "immune imprinting" implies that subsequent exposure to a drifted strain can induce affinity maturation of memory B cells toward cross-reactivity with the drifted strain and hence toward greater overall breadth. Here, we used deep mutational scanning of H1 influenza hemagglutinins (HAs) to investigate how viruses evolve in response to these broad antibody response. We identified escape mutations from clonal antibody lineages that targeted the receptor binding site and lateral patch. By adjusting the antigen-antibody contacts, antibody affinity maturation restricted the potential escape routes for the eliciting strain. However, escape occurred readily in drifted strains. We attribute this escape-prone property of the drifted strains to epistatic networks within HA. Our data explain how the influenza virus continues to evolve in the human population by escaping even broad antibody responses.
Collapse
Affiliation(s)
- Daniel P Maurer
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mya Vu
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aaron G Schmidt
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
240
|
Iversen R, Heggelund JE, Das S, Høydahl LS, Sollid LM. Enzyme-activating B-cell receptors boost antigen presentation to pathogenic T cells in gluten-sensitive autoimmunity. Nat Commun 2025; 16:2387. [PMID: 40064932 PMCID: PMC11894174 DOI: 10.1038/s41467-025-57564-5] [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: 09/23/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Autoantibodies against the enzyme transglutaminase 3 (TG3) are characteristic to the gluten-sensitive skin disorder dermatitis herpetiformis (DH), which is an extraintestinal manifestation of celiac disease. We here demonstrate that TG3-specific B cells can activate gluten-specific CD4+ T cells through B-cell receptor (BCR)-mediated internalization of TG3-gluten enzyme-substrate complexes. Stereotypic anti-TG3 antibodies using IGHV2-5/IGKV4-1 gene segments enhance the catalytic activity of TG3, and this effect translates into increased gluten presentation to T cells when such antibodies are expressed as BCRs. The crystal structure of TG3 bound to an IGHV2-5/IGKV4-1 Fab shows that antibody binding to a β-sheet in the catalytic core domain causes the enzyme to adopt the active conformation. This mechanism explains the production of stereotypic anti-TG3 autoantibodies in DH and highlights a role for TG3-specific B cells as antigen-presenting cells for gluten-specific T cells. Similar boosting effects of autoreactive BCRs could be relevant for other autoimmune diseases, including rheumatoid arthritis.
Collapse
Affiliation(s)
- Rasmus Iversen
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Julie Elisabeth Heggelund
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Saykat Das
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Lene S Høydahl
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Nextera AS, Oslo, Norway
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
241
|
Kim DK, Cho HY, Cho HJ, Kang BS. Allosteric inhibition of cytosolic NADP +-dependent isocitrate dehydrogenase by oxaloacetate. J Struct Biol 2025; 217:108183. [PMID: 40064325 DOI: 10.1016/j.jsb.2025.108183] [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: 12/14/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
NADP+-dependent cytosolic isocitrate dehydrogenase (IDH1) plays a crucial role in providing reducing energy in response to oxidative stress through the oxidative decarboxylation of isocitrate. NADPH generated by IDH1 serves as an essential cofactor for fatty acid synthesis. The regulation of IDH1 activity is vital for the biological functions of NADPH within cells, and mutations in IDH1 have been implicated in various cancers. In an effort to identify small regulatory molecules for IDH1, we determined the crystal structures of mouse IDH1 complexed with isocitrate and with oxaloacetate. Each IDH1 comprises large and small domains that form an active site, along with a clasp domain that connects two IDH1 molecules for dimerization. Isocitrate was located at the active site in the presence of a magnesium ion, while oxaloacetate was found at a novel site formed by the two clasp domains, in addition to the active site. The activity of IDH1 was diminished in the presence of oxaloacetate and could not be restored by the addition of isocitrate, indicating the presence of allosteric regulation. The activity of the IDH1 H170A mutant, which is unable to bind oxaloacetate in the clasp domain, was unaffected by oxaloacetate. This allosteric regulatory site may serve as a potential target for novel IDH1 inhibitors.
Collapse
Affiliation(s)
- Dong Kyu Kim
- School of Life Science and Biotechnology, KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ha Yeon Cho
- School of Life Science and Biotechnology, KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyo Je Cho
- Department of Biochemistry, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - Beom Sik Kang
- School of Life Science and Biotechnology, KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
242
|
Wang M, Medarametla P, Kronenberger T, Deingruber T, Brear P, Figueroa W, Ho PM, Krueger T, Pearce JC, Poso A, Wakefield JG, Spring DR, Welch M. Pseudomonas aeruginosa acyl-CoA dehydrogenases and structure-guided inversion of their substrate specificity. Nat Commun 2025; 16:2334. [PMID: 40057486 PMCID: PMC11890623 DOI: 10.1038/s41467-025-57532-z] [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: 04/18/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Fatty acids are a primary source of carbon for Pseudomonas aeruginosa (PA) in the airways of people with cystic fibrosis (CF). Here, we use tandem mass-tag proteomics to analyse the protein expression profile of a CF clinical isolate grown on different fatty acids. Two fatty acyl-CoA dehydrogenases (designated FadE1 and FadE2) are strongly induced during growth on fatty acids. FadE1 displays a strong preference for long-chain acyl-CoAs, whereas FadE2 exclusively utilizes medium-chain acyl-CoAs. Structural analysis of the enzymes enables us to identify residues comprising the substrate selectivity filter in each. Engineering these residues enables us to invert the substrate specificity of each enzyme. Mutants in fadE1 displayed impaired virulence in an infection model, and decreased growth on long chain fatty acids. The unique features of the substrate binding pocket enable us to identify an inhibitor that is differentially active against FadE1 and FadE2.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
| | - Prasanthi Medarametla
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Thales Kronenberger
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany
- Partner-site Tübingen, German Center for Infection Research (DZIF), Tübingen, Germany
| | - Tomas Deingruber
- Yusuf Hamied Department of Chemistry, Lensfield Road, Cambridge, UK
| | - Paul Brear
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
| | - Wendy Figueroa
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
| | - Pok-Man Ho
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
| | - Thomas Krueger
- Department of Biochemistry, Tennis Court Road, Cambridge, UK
| | - James C Pearce
- Living System Institute, Department of Biosciences, University of Exeter, Exeter, UK
| | - Antti Poso
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - James G Wakefield
- Living System Institute, Department of Biosciences, University of Exeter, Exeter, UK
| | - David R Spring
- Yusuf Hamied Department of Chemistry, Lensfield Road, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Tennis Court Road, Cambridge, UK.
| |
Collapse
|
243
|
Funke FJ, Schlee S, Bento I, Bourenkov G, Sterner R, Wilmanns M. Activity Regulation of a Glutamine Amidotransferase Bienzyme Complex by Substrate-Induced Subunit Interface Expansion. ACS Catal 2025; 15:4359-4373. [PMID: 40365074 PMCID: PMC7617670 DOI: 10.1021/acscatal.4c07438] [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] [Indexed: 05/15/2025]
Abstract
Glutamine amidotransferases are multienzyme machineries in which reactive ammonia is generated by a glutaminase and then transferred through a sequestered protein tunnel to a synthase active site for incorporation into diverse metabolites. To avoid wasteful metabolite consumption, there is a requirement for synchronized catalysis, but any generally applicable mechanistic insight is still lacking. As synthase activity depends on glutamine turnover, we investigated possible mechanisms controlling glutaminase catalysis using aminodeoxychorismate synthase involved in folate biosynthesis as a model. By analyzing this system in distinct states of catalysis, we found that incubation with glutamine leads to a subunit interface expansion by one-third of its original area. These changes completely enclose the glutaminase active site for sequestered catalysis and the subsequent transport of volatile ammonia to the synthase active site. In view of similar rearrangements in other glutamine amidotransferases, our observations may provide a general mechanism for the catalysis synchronization of this multienzyme family.
Collapse
Affiliation(s)
- Franziska Jasmin Funke
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg 93040, Germany
| | - Sandra Schlee
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg 93040, Germany
| | - Isabel Bento
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg 22607, Germany
| | - Gleb Bourenkov
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg 22607, Germany
| | - Reinhard Sterner
- Institute of Biophysics and Physical Biochemistry, Regensburg Center for Biochemistry, University of Regensburg, Regensburg 93040, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg 22607, Germany; University Hamburg Clinical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| |
Collapse
|
244
|
Sieskind R, Missoury S, Madru C, Commenge I, Niogret G, Hollenstein M, Rondelez Y, Sauguet L, Haouz A, Legrand P, Delarue M. Crystal structures of monomeric BsmI restriction endonuclease reveal coordinated sequential cleavage of two DNA strands. Commun Biol 2025; 8:387. [PMID: 40055548 PMCID: PMC11889245 DOI: 10.1038/s42003-025-07612-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/28/2025] [Indexed: 05/13/2025] Open
Abstract
BsmI, a thermophilic Type IIS restriction endonuclease from Bacillus stearothermophilus, presents a unique structural composition, housing two distinct active sites within a single monomer. Recognition of the non-symmetrical 5'-GAATGC-3' sequence enables precise cleavage of the top and bottom DNA strands. Synthetic biology interventions have led to the transformation of BsmI into Nb.BsmI, a nicking endonuclease. Here we introduce Nt*.BsmI, tailored for top-strand cleavage, which is inactive on standard double-stranded DNA, but active on bottom-strand nicked DNA, suggesting a sequential cleavage mechanism. Crystallographic structures of pre- and post-reactive complexes with cognate DNA show one major conformational change, a retractable loop possibly governing sequential active site accessibility. The x-ray structures reveal the position of the divalent metal ions in the active sites and the DNA:protein interactions, while the models predicted by Alphafold3 are incorrect. This comprehensive structural and functional study lays a foundation for rational enzyme redesign and potential applications in biotechnology.
Collapse
Affiliation(s)
- Rémi Sieskind
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France
| | - Sophia Missoury
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France
| | - Clément Madru
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France
| | - Isciane Commenge
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France
| | - Germain Niogret
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Unit of Bioorganic Chemistry of Nucleic Acids, 75724, Paris, France
| | - Marcel Hollenstein
- Institut Pasteur, Université Paris Cité, CNRS UMR 3523, Unit of Bioorganic Chemistry of Nucleic Acids, 75724, Paris, France
| | - Yannick Rondelez
- Laboratoire Gulliver, UMR7083 CNRS/ESPCI Paris-PSL Research University, 10 rue Vauquelin, 75005, Paris, France
| | - Ludovic Sauguet
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Plateforme de cristallographie-C2RT, 75724, Paris, France
| | | | - Marc Delarue
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unit of Architecture and Dynamics of Biological Macromolecules, 75724, Paris, France.
| |
Collapse
|
245
|
DiDonato M, Simpson CT, Vo T, Knuth M, Geierstanger B, Jamontt J, Jones DH, Fathman JW, DeLarosa D, Junt T, Picard D, Sommer U, Bagger M, Peters E, Meeusen S, Spraggon G. A novel interleukin-10 antibody graft to treat inflammatory bowel disease. Structure 2025; 33:475-488.e7. [PMID: 39798572 DOI: 10.1016/j.str.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/19/2024] [Accepted: 12/12/2024] [Indexed: 01/15/2025]
Abstract
Inflammatory bowel disease (IBD) consists of chronic conditions that severely impact a patient's health and quality of life. Interleukin-10 (IL-10), a potent anti-inflammatory cytokine has strong genetic links to IBD susceptibility and has shown strong efficacy in IBD rodent models, suggesting it has great therapeutic potential. However, when tested in clinical trials for IBD, recombinant human IL-10 (rhIL-10) showed weak and inconsistent efficacy due to its short half-life and pro-inflammatory properties that counteract the anti-inflammatory efficacy. Here we present an engineered, IL-10, antibody-graft therapeutic (GFT-IL10M) designed to rectify these issues. GFT-IL10M combines the half-life extension properties of a monoclonal IgG antibody with altered IL-10 cell-type selective signaling, retaining desirable signaling on monocytes while reducing unwanted signaling on T, natural killer (NK), and B cells. Our structural and biochemical results indicate that the altered IL-10 topology in GFT-IL10M leads to a predominantly anti-inflammatory profile, potentially altering cell-type specific signaling patterns and extending half-life.
Collapse
Affiliation(s)
- Michael DiDonato
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Carolina Turk Simpson
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Todd Vo
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Mark Knuth
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Bernhard Geierstanger
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | | | - David H Jones
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - John W Fathman
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Donnie DeLarosa
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Tobias Junt
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Damien Picard
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Ulrike Sommer
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Morten Bagger
- Novartis Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Eric Peters
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Shelly Meeusen
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Glen Spraggon
- Novartis Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| |
Collapse
|
246
|
Bou-Nader C, Link KA, Suddala KC, Knutson JR, Zhang J. Structures of complete HIV-1 TAR RNA portray a dynamic platform poised for protein binding and structural remodeling. Nat Commun 2025; 16:2252. [PMID: 40050622 PMCID: PMC11885821 DOI: 10.1038/s41467-025-57519-w] [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: 10/01/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
The HIV-1 TAR RNA plays key roles in viral genome architecture, transcription and replication. Previous structural analyses focused on its upper stem loop, which has served as a paradigm to study RNA structural dynamics. However, an imperfectly paired lower stem immediately abuts and stacks with the upper half, both of which are required for efficient HIV replication. Here, we report crystal structures of the full-length HIV-1 TAR which reveal substantial conformational mobility in its three conserved bulges and in its lower stem, which coordinately maintain the structural fluidity of the entire RNA. We find that TAR RNA is a robust inhibitor of PKR, and primarily uses its lower stem to capture and sequester PKR monomers, preventing their dimerization and activation. The lower stem exhibits transient conformational excursions detected by a ligation assay. Time-resolved fluorescence spectroscopy reveals local and global TAR structural remodeling by HIV-1 nucleocapsid, Tat, and PKR. This study portrays the structure, dynamics, and interactions of a complete TAR RNA, uncovers a convergent RNA-based viral strategy to evade innate immunity, and provides avenues to develop antivirals that target a dynamic, multifunctional viral RNA.
Collapse
Affiliation(s)
- Charles Bou-Nader
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Katie A Link
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Krishna C Suddala
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Jay R Knutson
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jinwei Zhang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA.
| |
Collapse
|
247
|
Clabbers MTB, Hattne J, Martynowycz MW, Gonen T. Energy filtering enables macromolecular MicroED data at sub-atomic resolution. Nat Commun 2025; 16:2247. [PMID: 40050283 PMCID: PMC11885823 DOI: 10.1038/s41467-025-57425-1] [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: 09/10/2024] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
High-resolution information is important for accurate structure modeling but is challenging to attain in macromolecular crystallography due to the rapid fading of diffracted intensities at increasing resolution. While direct electron detection essentially eliminates the read-out noise during MicroED data collection, other sources of noise remain and limit the measurement of faint high-resolution reflections. Inelastic scattering significantly contributes to noise, raising background levels and broadening diffraction peaks. We demonstrate a substantial improvement in signal-to-noise ratio by using energy filtering to remove inelastically scattered electrons. This strategy results in sub-atomic resolution MicroED data from proteinase K crystals, enabling the visualization of detailed structural features. Interestingly, reducing the noise further reveals diffuse scattering that may hold additional structural information. Our findings suggest that combining energy filtering and direct detection provides more accurate measurements at higher resolution, facilitating precise model refinement and improved insights into protein structure and function.
Collapse
Affiliation(s)
- Max T B Clabbers
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Johan Hattne
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Michael W Martynowycz
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA
| | - Tamir Gonen
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, University of California, Los Angeles, CA, 90095, USA.
- Department of Physiology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
248
|
Schlosser A, Pilecki B, Allen C, Benest AV, Lynch AP, Hua J, Ved N, Blackley Z, Andersen TL, Hennig D, Graversen JH, Möller S, Skallerup S, Ormhøj M, Lange C, Agostini HT, Grauslund J, Heegaard S, Dacheva I, Koss M, Hu W, Iglesias B, Lawrence MS, Beck HC, Steffensen LB, Laursen NS, Andersen GR, Holmskov U, Bates DO, Sorensen GL. Pharmacological blocking of microfibrillar-associated protein 4 reduces retinal neoangiogenesis and vascular leakage. Mol Ther 2025; 33:1048-1072. [PMID: 39863929 PMCID: PMC11897753 DOI: 10.1016/j.ymthe.2025.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025] Open
Abstract
Neovascular age-related macular degeneration and diabetic macular edema are leading causes of vision loss evoked by retinal neovascularization and vascular leakage. The glycoprotein microfibrillar-associated protein 4 (MFAP4) is an integrin αVβ3/5/6 ligand present in the extracellular matrix. Single-cell transcriptomics reveal MFAP4 expression in cell types in close proximity to vascular endothelial cells, including choroidal vascular mural cells, retinal astrocytes, and Müller cells. Binding of the anti-MFAP4 antibody, hAS0326, makes MFAP4 inaccessible for integrin receptor interaction, and thereby hAS0326 blocked endothelial cell motility in vitro. Intravitreal hAS0326 inhibited retinal vascular lesion area and neovessel volume in a laser-induced choroidal neovascularization mouse model, vascular permeability in streptozotocin-induced retinopathy, and vascular leakage area in a chronic non-human primate model of DL-2-aminoadipic acid-induced retinopathy. One dose of hAS0326 showed duration of efficacy of at least 12 weeks in the latter model. Moreover, hAS0326 treatment significantly enriched Gene Ontology terms involving reduction of integrin binding. Our data suggest that hAS0326 constitutes a promising treatment of neovascularization and vascular leakage in retinal diseases.
Collapse
Affiliation(s)
- Anders Schlosser
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Bartosz Pilecki
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Claire Allen
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Andrew V Benest
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Amy P Lynch
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Jing Hua
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Nikita Ved
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Zoe Blackley
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Thomas L Andersen
- Molecular Bone Histology Laboratory, Department of Pathology, Odense University Hospital, 5000 Odense, Denmark; Molecular Bone Histology Laboratory, Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark; Danish Spatial Imaging Consortium (DanSIC), University of Southern Denmark, 5230 Odense, Denmark
| | - Dorle Hennig
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Jonas H Graversen
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Sören Möller
- OPEN - Open Patient Data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Sofie Skallerup
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Maria Ormhøj
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Clemens Lange
- Ophtha-Lab, Department of Ophthalmology, St. Franziskus Hospital, 48145 Münster, Germany; Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Hansjürgen T Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jakob Grauslund
- Department of Ophthalmology, Odense University Hospital, 5000 Odense, Denmark
| | - Steffen Heegaard
- Department of Pathology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark; Department of Ophthalmology, Rigshospitalet-Glostrup, University of Copenhagen, 2600 Glostrup, Denmark
| | - Ivanka Dacheva
- Department of Ophthalmology, University of Heidelberg, 69047 Heidelberg, Germany
| | - Michael Koss
- Department of Ophthalmology, University of Heidelberg, 69047 Heidelberg, Germany
| | - Wenzheng Hu
- Virscio, Inc., 5 Science Park, New Haven, CT 06511, USA
| | | | | | - Hans Christian Beck
- Department of Clinical Biochemistry, Odense University Hospital, 5000 Odense, Denmark
| | - Lasse Bach Steffensen
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Nick S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Gregers R Andersen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| | - Uffe Holmskov
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2UH, UK
| | - Grith L Sorensen
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark.
| |
Collapse
|
249
|
Majocchi S, Lloveras P, Nouveau L, Legrand M, Viandier A, Malinge P, Charreton M, Raymond C, Pace EA, Millard BL, Svensson LA, Kelpšas V, Anceriz N, Salgado-Pires S, Daubeuf B, Magistrelli G, Gueneau F, Moine V, Masternak K, Shang L, Fischer N, Ferlin WG. NI-3201 Is a Bispecific Antibody Mediating PD-L1-Dependent CD28 Co-stimulation on T Cells for Enhanced Tumor Control. Cancer Immunol Res 2025; 13:365-383. [PMID: 39760515 PMCID: PMC11876958 DOI: 10.1158/2326-6066.cir-24-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025]
Abstract
Despite advances in cancer immunotherapy, such as targeting the PD-1/PD-L1 axis, a substantial number of patients harbor tumors that are resistant or relapse. Selective engagement of T-cell co-stimulatory molecules with bispecific antibodies may offer novel therapeutic options by enhancing signal 1-driven activation occurring via T-cell receptor engagement. In this study, we report the development and preclinical characterization of NI-3201, a PD-L1×CD28 bispecific antibody generated on the κλ-body platform that was designed to promote T-cell activity and antitumor function through a dual mechanism of action. We confirmed that NI-3201 blocks the PD-L1/PD-1 immune checkpoint pathway and conditionally provides T-cell co-stimulation via CD28 (signal 2) when engaging PD-L1+ tumors or immune cells. In systems with signal 1-primed T cells, NI-3201 enhanced potent effector functionality: in vitro through antigen-specific recall assays with cytomegalovirus-specific T cells and in vivo by inducing tumor regression and immunologic memory in tumor-associated antigen-expressing MC38 syngeneic mouse models. When T-cell engagers were used to provide synthetic signal 1, the combination with NI-3201 resulted in synergistic T cell-dependent cytotoxicity and potent antitumor activity in two humanized mouse tumor models. Nonhuman primate safety assessments showed favorable tolerability and pharmacokinetics at pharmacologically active doses. Quantitative systems pharmacology modeling predicted that NI-3201 exposure results in antitumor activity in patients, but this remains to be investigated. Overall, this study suggests that by combining PD-L1 blockade with safe and effective CD28 co-stimulation, NI-3201 has the potential to improve cancer immunotherapy outcomes, and the clinical development of NI-3201 for PD-L1+ solid tumors is planned.
Collapse
Affiliation(s)
- Sara Majocchi
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Lise Nouveau
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | | | | - Maud Charreton
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | - Cecile Raymond
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | | | | | | - Nadia Anceriz
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Bruno Daubeuf
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Franck Gueneau
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | - Valéry Moine
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Limin Shang
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | |
Collapse
|
250
|
Mockler N, Ramberg KO, Flood RJ, Crowley PB. N-Terminal Protein Binding and Disorder-to-Order Transition by a Synthetic Receptor. Biochemistry 2025; 64:1092-1098. [PMID: 39977527 PMCID: PMC11883740 DOI: 10.1021/acs.biochem.4c00729] [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: 10/29/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
We describe the capture and structuring of disordered N-terminal regions by the macrocycle sulfonato-calix[4]arene (sclx4). Using the trimeric β-propeller Ralstonia solanacearum lectin (RSL) as a scaffold, we generated a series of mutants with extended and dynamic N-termini. Three of the mutants feature an N-terminal methionine-lysine motif. The fourth mutant contains the disordered 8-residue N-terminus of Histone 3, a component of the nucleosome. X-ray crystallography and NMR spectroscopy provide evidence for sclx4 binding to the flexible N-terminal regions. Three crystal structures reveal that the calixarene recognizes the N-terminal Met-Lys motif, capturing either residue. We provide crystallographic proof for sclx4 encapsulation of N-terminal methionine. Calixarene capture of intrinsically disordered regions may have applications in regulating protein secondary (and tertiary) structure.
Collapse
Affiliation(s)
- Niamh
M. Mockler
- School of Biological and
Chemical Sciences, University of Galway, Galway H91 TK33, Ireland
| | | | - Ronan J. Flood
- School of Biological and
Chemical Sciences, University of Galway, Galway H91 TK33, Ireland
| | - Peter B. Crowley
- School of Biological and
Chemical Sciences, University of Galway, Galway H91 TK33, Ireland
| |
Collapse
|