1
|
Zhu H, Terashi G, Farheen F, Nakamura T, Kihara D. AI-based quality assessment methods for protein structure models from cryo-EM. Curr Res Struct Biol 2025; 9:100164. [PMID: 39996138 PMCID: PMC11848767 DOI: 10.1016/j.crstbi.2025.100164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Cryogenic electron microscopy (cryo-EM) has revolutionized structural biology, with an increasing number of structures being determined by cryo-EM each year, many at higher resolutions. However, challenges remain in accurately interpreting cryo-EM maps. Inaccuracies can arise in regions of locally low resolution, where manual model building is more prone to errors. Validation scores for structure models have been developed to assess both the compatibility between map density and the structure, as well as the geometric and stereochemical properties of protein models. Recent advancements have introduced artificial intelligence (AI) into this field. These emerging AI-driven tools offer unique capabilities in the validation and refinement of cryo-EM-derived protein atomic models, potentially leading to more accurate protein structures and deeper insights into complex biological systems.
Collapse
Affiliation(s)
- Han Zhu
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Genki Terashi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Farhanaz Farheen
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Tsukasa Nakamura
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Structural Biology Research Center, High Energy Accelerator Research Organization (KEK), Tsukuba, Ibaraki, 305-0801, Japan
| | - Daisuke Kihara
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
2
|
Chatziefthymiou SD, Kuzikov M, Afandi S, Kovacs G, Srivastava S, Zaliani A, Gruzinov A, Pompidor G, Lunelli M, Ahmed GR, Labahn J, Hakanpää J, Windshügel B, Kolbe M. Identification, validation, and characterization of approved and investigational drugs interfering with the SARS-CoV-2 endoribonuclease Nsp15. Protein Sci 2025; 34:e70156. [PMID: 40371758 PMCID: PMC12079475 DOI: 10.1002/pro.70156] [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: 12/18/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
Since the emergence of SARS-CoV-2 at the end of 2019, the virus has caused significant global health and economic disruptions. Despite the rapid development of antiviral vaccines and some approved treatments such as remdesivir and paxlovid, effective antiviral pharmacological treatments for COVID-19 patients remain limited. This study explores Nsp15, a 3'-uridylate-specific RNA endonuclease, which has a critical role in immune system evasion and hence in escaping the innate immune sensors. We conducted a comprehensive drug repurposing screen and identified 44 compounds that showed more than 55% inhibition of Nsp15 activity in a real-time fluorescence assay. A validation pipeline was employed to exclude unspecific interactions, and dose-response assays confirmed 29 compounds with an IC50 below 10 μM. Structural studies, including molecular docking and x-ray crystallography, revealed key interactions of identified inhibitors, such as TAS-103 and YM-155, with the Nsp15 active site and other critical regions. Our findings show that the identified compounds, particularly those retaining potency under different assay conditions, could serve as promising hits for developing Nsp15 inhibitors. Additionally, the study emphasizes the potential of combination therapies targeting multiple viral processes to enhance treatment efficacy and reduce the risk of drug resistance. This research contributes to the ongoing efforts to develop effective antiviral therapies for SARS-CoV-2 and possibly other coronaviruses.
Collapse
Affiliation(s)
- Spyros D. Chatziefthymiou
- Photon Science, DESYHamburgGermany
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Maria Kuzikov
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
- School of Science, Constructor UniversityBremenGermany
| | - Sara Afandi
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Greta Kovacs
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Sukrit Srivastava
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
| | | | | | - Michele Lunelli
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Golam Rizvee Ahmed
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
| | - Jörg Labahn
- Forschungszentrum Jülich, Institute of Complex Systems (ICS‐6)JülichGermany
| | | | - Björn Windshügel
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPortHamburgGermany
- School of Science, Constructor UniversityBremenGermany
| | - Michael Kolbe
- Department for Structural Infection BiologyCentre for Structural Systems Biology (CSSB) & Helmholtz‐Centre for Infection ResearchHamburgGermany
- Faculty of Mathematics, Informatics and Natural SciencesUniversität HamburgHamburgGermany
| |
Collapse
|
3
|
Zvornicanin SN, Shaqra AM, Flynn J, Carias Martinez H, Jia W, Moquin S, Dovala D, Bolon DN, Kurt Yilmaz N, Schiffer CA. Molecular mechanisms of drug resistance and compensation in SARS-CoV-2 main protease: the interplay between E166 and L50. mBio 2025; 16:e0406824. [PMID: 40183562 PMCID: PMC12077200 DOI: 10.1128/mbio.04068-24] [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: 01/15/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
The SARS-CoV-2 main protease (Mpro) is essential for viral replication and is a primary target for COVID-19 antivirals. Direct-acting antivirals such as nirmatrelvir, the active component of Paxlovid, target the Mpro active site to block viral polyprotein cleavage and thus replication. However, drug resistance mutations at the active site residue Glu166 (E166) have emerged during in vitro selection studies, raising concerns about the durability of current antiviral strategies. Here, we investigate the molecular basis of drug resistance conferred by E166A and E166V mutations against nirmatrelvir and the related PF-00835231, individually and in combination with the distal mutation L50F. We found that E166 mutations reduce nirmatrelvir potency by up to 3,000-fold while preserving substrate cleavage, with catalytic efficiency reduced by only up to twofold. This loss of catalytic efficiency was compensated for by the addition of L50F in the double-mutant variants. We have determined three cocrystal structures of the E166 variants (E166A, E166V, and E166V/L50F) bound to PF-00835231. Comparison of these structures with wild-type enzyme demonstrated that E166 is crucial for dimerization and for shaping the substrate-binding S1 pocket. Our findings highlight the mutability of E166, a prime site for resistance for inhibitors that leverage direct interactions with this position, and the potential emergence of highly resistant and active variants in combination with the compensatory mutation L50F. These insights support the design of inhibitors that target conserved protease features and avoid E166 side-chain interactions to minimize susceptibility to resistance. IMPORTANCE Drug resistance remains a great challenge to modern medicine. This study investigates SARS-CoV-2 main protease variants E166A and E166V which confer nirmatrelvir resistance. These variants can retain considerable enzymatic activity through combination with the compensatory mutation L50F. For single- and double-mutant variant enzymes, we assessed catalytic efficiency, measured loss in potency for nirmatrelvir and its analog PF-00835231, and cocrystallized with inhibitors to investigate drug resistance caused by these mutations. Our results contribute toward understanding of molecular mechanisms of resistance and combinations of mutations, which pushes toward resistance-thwarting inhibitor design. These principles also apply broadly to many quickly evolving drug targets in infectious diseases.
Collapse
Affiliation(s)
- Sarah N. Zvornicanin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Ala M. Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Julia Flynn
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Weiping Jia
- Biomedical Research, Novartis, Emeryville, California, USA
| | | | - Dustin Dovala
- Biomedical Research, Novartis, Emeryville, California, USA
| | - Daniel N. Bolon
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
4
|
Luang S, Fernández-Luengo X, Streltsov VA, Maréchal JD, Masgrau L, Hrmova M. The structure and dynamics of water molecule networks underlie catalytic efficiency in a glycoside exo-hydrolase. Commun Biol 2025; 8:729. [PMID: 40348901 PMCID: PMC12065899 DOI: 10.1038/s42003-025-08113-9] [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: 09/18/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
Glycoside hydrolases break glycosidic bonds by transferring a water molecule onto the glycosidic oxygen of carbohydrates, but on the nanoscale, the dynamics of water molecules remains unclear. We investigate the role of the non-nucleophilic E220 glutamate, essential for maintaining the water molecule network in a family 3 β-D-glucan glucohydrolase, but not involved directly in catalysis. Kinetic data disclose that the E220A mutant retains substrate poly-specificity but has drastically reduced catalytic efficiency compared to the wild-type. High-resolution structures in-complex with a hydrolytic product and a mechanism-based inhibitor reveal that in wild-type, the concatenated water molecules near acid/base E491 and neighbouring N219 and E220 form a harmonised network. In contrast, in the E220A mutant, this network is uncoordinated. Computational models of covalent complexes show that water flux through the wild-type protein correlates with high catalytic efficiency dissimilar to E220A, where this correlation is lost. Ancestral sequence reconstructions of family 3 enzymes divulge the evolutionary conservation of residues participating in water molecule networks, which underlie substrate-product-assisted processivity. Our findings provide a blueprint for the dynamics of catalysis mediated by hydrolytic enzymes, which could inspire bioengineering to create a sustainable bio-economy.
Collapse
Affiliation(s)
- Sukanya Luang
- School of Agriculture, Food and Wine, and Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, SA, Australia
| | | | - Victor A Streltsov
- The Florey Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Jean-Didier Maréchal
- Department de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laura Masgrau
- Department de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| | - Maria Hrmova
- School of Agriculture, Food and Wine, and Waite Research Institute, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
5
|
Geng A, Roy R, Gu S, Guseva S, Pratihar S, Lee Y, Li L, Kimsey IJ, Wilson MA, Al-Hashimi HM. Insight into the Conformational Ensembles Formed by U-U and T-T Mismatches in RNA and DNA Duplexes From a Structure-based Survey, NMR, and Molecular Dynamics Simulations. J Mol Biol 2025; 437:169197. [PMID: 40345379 DOI: 10.1016/j.jmb.2025.169197] [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: 03/25/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Nucleic acid base pairs interconvert between alternative conformations on a free energy landscape, and these dynamics play critical roles in recognition, folding, and catalysis. U-U and T-T mismatches can adopt two nearly isoenergetic wobble conformations, distinguished by their relative shearing displacements. Experimental NMR evidence suggests that these conformations dynamically interconvert in RNA motifs containing tandem U-U mismatches. However, whether such motions occur ubiquitously across U-U and T-T mismatches remains unknown, as high-resolution nucleic acid structures typically report only a single conformation. Here, we used NMR spectroscopy, a structure-based survey of the Protein Data Bank, and molecular dynamics (MD) simulations to investigate wobble dynamics in U-U and T-T mismatches when flanked by canonical Watson-Crick base pairs in RNA and DNA duplexes. The structure-based survey revealed that U-U mismatches have propensities to adopt alternative wobble conformations even when controlling for sequence and identified potential intermediates along the wobble transition. Off-resonance R1ρ relaxation dispersion experiments detected no micro- to millisecond dynamics for U-U mismatches in duplex RNA and T-T mismatches in duplex DNA. However, alternative conformer refinement of the electron density in X-ray structures, inter-proton NOEs, carbonyl carbon chemical shifts, an RDC-derived conformational ensemble, and MD simulations indicated that U-U and T-T mismatches exist in a dynamic equilibrium between two wobble conformations, with the minor state exceeding 30% and the transitions occurring on the nanosecond timescale. Our findings suggest that U-U and T-T ubiquitously undergo sub-microsecond wobble motions, contributing to the energetic landscape and dynamic plasticity of nucleic acids, with important implications for processes that generate and act on these mismatches.
Collapse
Affiliation(s)
- Ainan Geng
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27701, USA
| | - Rohit Roy
- Center for Genomic and Computational Biology, Duke University School of Medicine, Durham, NC 27701, USA
| | - Stephanie Gu
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27701, USA
| | - Serafima Guseva
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Supriya Pratihar
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Yeongjoon Lee
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Linshu Li
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Isaac J Kimsey
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27701, USA
| | - Mark A Wilson
- Department of Biochemistry and Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Hashim M Al-Hashimi
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
6
|
Basanta B, Nugroho K, Yan NL, Kline GM, Powers ET, Tsai FJ, Wu M, Hansel-Harris A, Chen JS, Forli S, Kelly JW, Lander GC. The conformational landscape of human transthyretin revealed by cryo-EM. Nat Struct Mol Biol 2025; 32:876-883. [PMID: 39843982 DOI: 10.1038/s41594-024-01472-7] [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: 02/09/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025]
Abstract
Transthyretin (TTR) is a natively tetrameric thyroxine transporter in blood and cerebrospinal fluid whose misfolding and aggregation causes TTR amyloidosis. A rational drug design campaign identified the small molecule tafamidis (Vyndamax) as a stabilizer of the native TTR fold, and this aggregation inhibitor is regulatory agency approved for the treatment of TTR amyloidosis. Here we used cryo-EM to investigate the conformational landscape of this 55 kDa tetramer in the absence and presence of one or two ligands, revealing inherent asymmetries in the tetrameric architecture and previously unobserved conformational states. These findings provide critical mechanistic insights into negatively cooperative ligand binding and the structural pathways responsible for TTR amyloidogenesis, underscoring the capacity of cryo-EM to identify pharmacological targets suppressed by the confines of the crystal lattice, opening uncharted territory in structure-based drug design.
Collapse
Affiliation(s)
- Benjamin Basanta
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
- Arzeda, Seattle, WA, USA
| | - Karina Nugroho
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Nicholas L Yan
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Gabriel M Kline
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Evan T Powers
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Felix J Tsai
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Mengyu Wu
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
- Neomorph, San Diego, CA, USA
| | - Althea Hansel-Harris
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Jason S Chen
- Automated Synthesis Facility, Scripps Research, La Jolla, CA, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Jeffrey W Kelly
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Gabriel C Lander
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
7
|
Saha S, Kanaujia SP. Structural analysis of the ribosome assembly factor Nep1, an N1-specific pseudouridine methyltransferase, reveals mechanistic insights. FEBS J 2025; 292:2338-2358. [PMID: 39918246 DOI: 10.1111/febs.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/01/2024] [Accepted: 12/30/2024] [Indexed: 05/11/2025]
Abstract
Nucleolar essential protein 1 (Nep1; also known as ribosomal RNA small subunit methyltransferase Nep1) is a crucial factor in forming small ribosomal subunits in eukaryotes and archaea. Nep1 possesses an S-adenosyl-L-methionine (SAM)-dependent SpoU-TrmD (SPOUT) ribosomal RNA (rRNA) methyltransferase (MTase) fold and catalyzes pseudouridine (Ψ) methylation at specific sites of the small subunit (SSU) rRNA. Mutations in Nep1 proteins result in a severe developmental disorder in humans and reduced growth in yeast, suggesting its role in ribosome biogenesis. In this study, the crystal structures of Nep1 from the archaebacterium Pyrococcus horikoshii (PhNep1), both in its apo and holo (adenosine or 5-methylthioadenosine bound) forms have been reported. The structural analysis of PhNep1 revealed an α/β fold featuring a deep trefoil knot akin to the SPOUT domain, with two novel extensions-a globular loop and a β-α-β extension. Moreover, the cofactor-binding site of PhNep1 exhibits a preformed pocket, topologically similar to that of other SPOUT-class MTases. Further, structural analysis of PhNep1 revealed that it forms a homodimer coordinated by inter-subunit hydrogen bonds and hydrophobic interactions. Moreover, the results of this study indicate that PhNep1 can specifically methylate consensus RNAs, having a pseudouridine (ψ) located at position 926 of helix 35 (h35) of 16S rRNA in P. horikoshii. The stability of the Nep1-RNA complex seems to be primarily assisted by the conserved arginine residues located at the dimeric interface.
Collapse
Affiliation(s)
- Sayan Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, India
| | - Shankar Prasad Kanaujia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, India
| |
Collapse
|
8
|
Horton JR, Yu M, Zhou J, Tran M, Anakal RR, Lu Y, Blumenthal RM, Zhang X, Huang Y, Zhang X, Cheng X. Multimeric transcription factor BCL11A utilizes two zinc-finger tandem arrays to bind clustered short sequence motifs. Nat Commun 2025; 16:3672. [PMID: 40246927 PMCID: PMC12006351 DOI: 10.1038/s41467-025-58998-7] [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: 11/01/2024] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
BCL11A, a transcription factor, is vital for hematopoiesis, including B and T cell maturation and the fetal-to-adult hemoglobin switch. Mutations in BCL11A are linked to neurodevelopmental disorders. BCL11A contains two DNA-binding zinc-finger arrays, low-affinity ZF2-3 and high-affinity ZF4-6, separated by a 300-amino-acid linker. ZF2-3 and ZF4-5 share 73% identity, including five out of six DNA base-interacting residues. These arrays bind similar short sequence motifs in clusters, with the linker enabling a broader binding span. Crystallographic structures of ZF4-6, in complex with oligonucleotides from the β-globin locus region, reveal DNA sequence recognition by residues Asn756 (ZF4), Lys784 and Arg787 (ZF5). A Lys784-to-Thr mutation, linked to a neurodevelopmental disorder with persistent fetal globin expression, reduces DNA binding over 10-fold but gains interaction with a variable base pair. BCL11A isoforms may form oligomers, enhancing chromatin occupancy and repressor functions by allowing multiple copies of both low- and high-affinity ZF arrays to bind DNA. These distinctive properties, apparently conserved among vertebrates, provide essential functional flexibility to this crucial regulator.
Collapse
Affiliation(s)
- John R Horton
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Meigen Yu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jujun Zhou
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Melody Tran
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rithvi R Anakal
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Robert M Blumenthal
- Department of Medical Microbiology and Immunology, and Program in Bioinformatics, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, 43614, USA
| | - Xiaotian Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center Houston, McGovern Medical School, Houston, TX, 77030, USA
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, 77030, USA
| | - Xing Zhang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaodong Cheng
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
9
|
Peng W, Wang X, Liu Q, Xiao Z, Li F, Ji N, Chen Z, He J, Wang J, Deng Z, Lin S, Liang R. The GntR/VanR transcription regulator AlkR represses AlkB2 monooxygenase expression and regulates n-alkane degradation in Pseudomonas aeruginosa SJTD-1. MLIFE 2025; 4:126-142. [PMID: 40313978 PMCID: PMC12042122 DOI: 10.1002/mlf2.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/17/2024] [Accepted: 12/21/2024] [Indexed: 05/03/2025]
Abstract
Transmembrane alkane monooxygenase (AlkB)-type monooxygenases, especially AlkB2 monooxygenases, are crucial for aerobic degradation of the medium-to-long-chain n-alkanes in hydrocarbon-utilizing microorganisms. In this study, we identified a GntR/VanR transcription regulator AlkR of Pseudomonas aeruginosa SJTD-1 involved in the negative regulation of AlkB2 and deciphered its nature of DNA binding and ligand release. The deletion of alkR enhanced the transcription levels of the alkB2 gene and the utilization efficiency of the medium-to-long-chain n-alkanes by strain SJTD-1. The dimer of AlkR recognizes and binds to a conserved palindromic motif in the promoter of the alkB2 gene, and structural symmetry is vital for DNA binding and transcription repression. The long-chain fatty acyl coenzyme A compounds can release AlkR and stimulate transcription of alkB2, reflecting the effect of alkane catabolic metabolites. Structural insights unveiled that the arginine residues and scaffold residues of AlkR are critical for DNA binding. Further bioinformatics analysis of AlkR revealed the widespread VanR-AlkB couples distributed in Pseudomonadaceae with high conservation in the sequences of functional genes and intergenic regions, highlighting a conserved regulatory pattern for n-alkane utilization across this family. These findings demonstrate the regulatory mechanism and structural basis of GntR/VanR transcription regulators in modulating n-alkane biodegradation and provide valuable insights in improving the bioremediation efficiency of hydrocarbon pollution.
Collapse
Affiliation(s)
- Wanli Peng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Xiuli Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Qinchen Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Zhihong Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Fulin Li
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Nannan Ji
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Zhuo Chen
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Jiaying He
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Junhao Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Shuangjun Lin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| | - Rubing Liang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences & BiotechnologyShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
10
|
Li Z, Han X, Cong L, Singh P, Paiva P, Branson Y, Li W, Chen Y, Jaradat DMM, Lennartz F, Bayer T, Schmidt L, Garscha U, You S, Fernandes PA, Ramos MJ, Bornscheuer UT, Weber G, Wei R, Liu W. Structure-Guided Engineering of a Versatile Urethanase Improves Its Polyurethane Depolymerization Activity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416019. [PMID: 39921299 PMCID: PMC11967865 DOI: 10.1002/advs.202416019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/22/2025] [Indexed: 02/10/2025]
Abstract
Polyurethane (PUR), the fifth most prevalent synthetic polymer, substantially contributes to the global plastic waste problem. Biotechnology-based recycling methods have recently emerged as innovative solutions to plastic waste disposal and sparked interest among scientific communities and industrial stakeholders in discovering and designing highly active plastic-degrading enzymes. Here, the ligand-free crystal structure of UMG-SP2, a metagenome-derived urethanase with depolymerization activities, at 2.59 Å resolution, as well as its (co-)structures bound to a suicide hydrolase inhibitor and a short-chain carbamate substrate at 2.16 and 2.40 Å resolutions, respectively, is reported. Structural analysis and molecular dynamics simulations reveal that the flexible loop L3 consisting of residues 219-226 is crucial for regulating the hydrolytic activity of UMG-SP2. The semi-rational redesign of UMG-SP2 reveals superior variants, A141G and Q399A, exhibiting over 30.7- and 7.4-fold increased activities on polyester-PUR and a methylene diamine derivative of PUR, respectively, compared to the wild-type enzyme. These findings advance the understanding of the structure-function relationship of PUR-hydrolyzing enzymes, which hold great promise for developing effective industrial PUR recycling processes and mitigating the environmental footprint of plastic waste.
Collapse
Affiliation(s)
- Zhishuai Li
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
| | - Xu Han
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
| | - Lin Cong
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
- School of Life Sciences and Biopharmaceutical SciencesShenyang Pharmaceutical University103 Wenhua RoadShenhe DistrictShenyangLiaoning110016China
| | - Parinita Singh
- Helmholtz‐Zentrum Berlin für Materialien und EnergieAlbert‐Einstein‐Str. 15D‐12489BerlinGermany
| | - Pedro Paiva
- LAQV@REQUIMTEDepartamento de Química e BioquímicaFaculdade de Ciências da Universidade do PortoRua do Campo AlegrePorto4169‐007Portugal
| | - Yannick Branson
- Department of Biotechnology and Enzyme CatalysisInstitute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 417487GreifswaldGermany
| | - Wenshuo Li
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
| | - Yangyang Chen
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
| | - Da'san M. M. Jaradat
- Helmholtz‐Zentrum Berlin für Materialien und EnergieAlbert‐Einstein‐Str. 15D‐12489BerlinGermany
- Department of ChemistryFaculty of ScienceAl‐Balqa Applied UniversityP.O. Box 206Al‐Salt19117Jordan
| | - Frank Lennartz
- Helmholtz‐Zentrum Berlin für Materialien und EnergieAlbert‐Einstein‐Str. 15D‐12489BerlinGermany
| | - Thomas Bayer
- Department of Biotechnology and Enzyme CatalysisInstitute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 417487GreifswaldGermany
| | - Louis Schmidt
- Department of Pharmaceutical and Medicinal ChemistryInstitute of PharmacyUniversity of GreifswaldFriedrich‐Ludwig‐Jahn‐Str. 1717489GreifswaldGermany
| | - Ulrike Garscha
- Department of Pharmaceutical and Medicinal ChemistryInstitute of PharmacyUniversity of GreifswaldFriedrich‐Ludwig‐Jahn‐Str. 1717489GreifswaldGermany
| | - Song You
- School of Life Sciences and Biopharmaceutical SciencesShenyang Pharmaceutical University103 Wenhua RoadShenhe DistrictShenyangLiaoning110016China
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTEDepartamento de Química e BioquímicaFaculdade de Ciências da Universidade do PortoRua do Campo AlegrePorto4169‐007Portugal
| | - Maria João Ramos
- LAQV@REQUIMTEDepartamento de Química e BioquímicaFaculdade de Ciências da Universidade do PortoRua do Campo AlegrePorto4169‐007Portugal
| | - Uwe T. Bornscheuer
- Department of Biotechnology and Enzyme CatalysisInstitute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 417487GreifswaldGermany
| | - Gert Weber
- Helmholtz‐Zentrum Berlin für Materialien und EnergieAlbert‐Einstein‐Str. 15D‐12489BerlinGermany
| | - Ren Wei
- Department of Biotechnology and Enzyme CatalysisInstitute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 417487GreifswaldGermany
| | - Weidong Liu
- Key Laboratory of Engineering Biology for Low‐carbon ManufacturingNational Engineering Research Center for Industrial EnzymesNational Technology Innovation Center of Synthetic BiologyTianjin Institute of Industrial BiotechnologyChinese Academy of Sciences32 West Seventh Avenue, Tianjin Airport Economic AreaTianjin300308China
- University of Chinese Academy of Sciences19A Yuquan RoadBeijing100049China
| |
Collapse
|
11
|
Trindade IB, Fonseca BM, Catarino T, Matias PM, Moe E, Louro RO. Flavin-containing siderophore-interacting protein of Shewanella putrefaciens DSM 9451 reveals common structural and functional aspects of ferric-siderophore reduction. J Biol Inorg Chem 2025; 30:241-255. [PMID: 40080164 PMCID: PMC11965169 DOI: 10.1007/s00775-025-02106-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: 12/04/2024] [Accepted: 02/19/2025] [Indexed: 03/15/2025]
Abstract
Shewanella are bacteria widespread in marine and brackish water environments and emergent opportunistic pathogens. Their environmental versatility depends on the ability to produce numerous iron-rich proteins, mainly multiheme c-type cytochromes. Although iron plays a vital role in the versatility of Shewanella species, very few studies exist regarding the strategies by which these bacteria scavenge iron from the environment. Siderophore-mediated iron transport is a commonly employed strategy for iron acquisition, and it was identified among Shewanella spp. over two decades ago. Shewanella species produce hydroxamate-type siderophores and iron removal from these compounds can occur in the cytoplasm via Fe(III)-siderophore reduction mediated by siderophore-interacting proteins (SIPs). The genome of Shewanella putrefaciens DSM 9451 isolated from an infected child contains representatives of the two different families of SIPs: the flavin-containing siderophore reductase (SbSIP) and the iron-sulfur cluster-containing ferric-siderophore reductase (SbFSR). Here, we report their expression, purification, and further biochemical characterization of SbSIP. The structural and functional characterization of SbSIP and comparison with the homologous SIP from Shewanella frigidimarina (SfSIP) revealed similarities between these proteins including a common binding pocket for NADH, NADPH, and siderophore substrates plus a pronounced redox-Bohr effect that ensures coupled transfer of electrons and protons in the physiological pH range. These mechanistic aspects open the door for further investigations on developing drugs that interfere with the iron metabolism of these bacteria and thereby prevent their spread.
Collapse
Affiliation(s)
- Inês B Trindade
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Bruno M Fonseca
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
| | - Teresa Catarino
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
- Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516, Caparica, Portugal
| | - Pedro M Matias
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
- iBET-Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901, Oeiras, Portugal
| | - Elin Moe
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal
| | - Ricardo O Louro
- Avenida da República (EAN), Instituto de Tecnologia Química e Biológica António Xavier da Universidade Nova de Lisboa, 2780-157, Oeiras, Portugal.
| |
Collapse
|
12
|
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
|
13
|
de Oliveira Silva YR, Contreras-Martel C, Rodrigues de Melo R, Zanphorlin LM, Trindade DM, Dessen A. Architecture of an embracing lipase-foldase complex of the type II secretion system of Acinetobacter baumannii. Structure 2025; 33:601-612.e4. [PMID: 39904335 DOI: 10.1016/j.str.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/23/2024] [Accepted: 12/28/2024] [Indexed: 02/06/2025]
Abstract
Acinetobacter baumannii is a major human pathogen responsible for a growing number of multi-antibiotic-resistant infections, and of critical priority for the World Health Organization (WHO). A. baumannii employs a type II secretion system (T2SS) to secrete toxins extracellularly to enable cytotoxicity and colonization. Lipase LipA, secreted by the A. baumannii T2SS, is required for virulence and fitness, and in the periplasm is maintained in an active state by its essential foldase, LipB. Here we report that LipA is able to recognize lipids of different chain lengths at extremes of pH and temperature, thanks to its stabilization by LipB through an extended, highly helical "embrace." A vast bioinformatic analysis indicates that LipB-like foldases are widespread over numerous proteobacteria, and thus the extended foldase architecture shown here could be widespread. These results provide new insight into A. baumannii's adaptability as a pathogen in different environments and could facilitate the development of novel antibacterials.
Collapse
Affiliation(s)
- Yuri Rafael de Oliveira Silva
- Brazilian Biosciences National Laboratory (LNBio), CNPEM, Campinas São Paulo 13084-971, Brazil; Departamento de Genética, Evolução, Microbiologia e Imunologia, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Carlos Contreras-Martel
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38044 Grenoble, France
| | | | | | - Daniel Maragno Trindade
- Brazilian Biosciences National Laboratory (LNBio), CNPEM, Campinas São Paulo 13084-971, Brazil.
| | - Andréa Dessen
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale (IBS), 38044 Grenoble, France.
| |
Collapse
|
14
|
Eun HJ, Jang SW, Park JH, Lee J, Lee KY, Lee EJ, Lee BJ. Structural and functional analyses of STM14_5441-STM14_5442: A potential mechanism for persister formation against aminoglycosides. Drug Resist Updat 2025; 79:101210. [PMID: 39908597 DOI: 10.1016/j.drup.2025.101210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
AIMS The ability to eliminate bacterial persister cells is still a medical challenge that has yet to be overcome. These cells represent a unique subpopulation within bacterial communities and are characterized by a reduced susceptibility to antibiotics with growth retardation. In this study, we investigated the molecular basis of persister formation in Salmonella Typhimurium 14028 s under aminoglycoside stress. METHODS We analyzed the crystal structure of the STM14_5441-STM14_5442 complex, which belongs to the type II toxin-antitoxin system, and identified key ribosome-binding residues in STM14_5441. Changes in the antibiotic susceptibility of Salmonella caused by the loss of the ribosome-binding property of STM14_5441 were assessed. We conducted intracellular ATP assays under aminoglycoside stress and RNA-seq analysis following STM14_5441 induction. RESULTS Our studies demonstrated the critical role of STM14_5441 in the formation of persister cells in Salmonella, particularly those under aminoglycoside stress. We observed that a loss of ribosome binding in STM14_5441 resulted in increased antibiotic susceptibility. Additionally, intracellular ATP assays revealed increased ATP levels in STM14_5441 induced group, and RNA-seq analysis identified several genes that play a role in this phenomenon. CONCLUSIONS The present data suggest that persister forms under aminoglycoside stress through the following mechanisms: i) inhibition of membrane hyperpolarization by impeding F1Fo ATP synthase activity and ii) enhanced poststress recovery by ATP storage and increased protein synthesis capacity. Based on this suggestion, we reannotated the STM14_5441-STM14_5442 TA pair as the ResTA (RNA cleavage-induced energy storage toxin-antitoxin) system. Furthermore, new insights into the function of TA systems may lay the groundwork for developing novel strategies to target bacterial persister cells, thereby preventing the accelerated emergence of antibiotic resistance in bacterial populations.
Collapse
Affiliation(s)
- Hyun-Jong Eun
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seok-Won Jang
- Bioinformatics Branch, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Ju-Hyun Park
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Jooyeon Lee
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Ki-Young Lee
- School of Pharmacy, Sungkyunkwan University, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Eun-Jin Lee
- Department of Life Sciences, Korea University, Seoul 02481, Republic of Korea
| | - Bong-Jin Lee
- College of Pharmacy, Ajou University, Suwon-si, Gyeonggi-do 16499, Republic of Korea; MasterMediTech, Seoul 07795, Republic of Korea.
| |
Collapse
|
15
|
Mathpal S, Joshi T, Priyamvada P, Ramaiah S, Anbarasu A. Machine learning and cheminformatics-based Identification of lichen-derived compounds targeting mutant PBP4 R200L in Staphylococcus aureus. Mol Divers 2025:10.1007/s11030-025-11125-6. [PMID: 39954181 DOI: 10.1007/s11030-025-11125-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Penicillin-binding protein 4 (PBP4) is essential in imparting significant β-lactam antibiotics resistance in Staphylococcus aureus (S. aureus) and the mutation R200L in PBP4 is linked to β-lactam non-susceptibility in natural strains, complicating treatment options. Therefore, discovering novel therapeutics against the mutant PBP4 is crucial, and natural compounds from lichen have found relevance in this regard. The aim of our study was to identify novel inhibitors against the R200L mutation by applying machine learning (ML) approach. Predictive classification models were developed using six machine learning algorithms to categorize lichen-derived compounds as either active or inactive. The models were evaluated using ROC curves, confusion matrices, and relevant statistical parameters. Among these, the Extra Trees algorithm showed superior predictive accuracy at 81%. The model identified 115 potentially active compounds from lichen, which were further evaluated for drug-likeness and structural similarity to β-lactam antibiotics. The top 23 compounds, showing similarity to β-lactam drug, were subjected to molecular docking. Among the top 10 compounds, two compounds, Barbatolic acid and Orcinyl lecanorate, displayed promising results in 200 ns molecular dynamics (MD) simulations and MM-PBSA analysis, exhibiting better docking score compare to reference compound. Additionally, DFT calculations revealed negative binding energies and smaller HOMO-LUMO gaps for both compounds. The obtained results prove the utility of ML in screening natural compounds, and provide novel opportunities for the design of antimicrobial compounds in the future.
Collapse
Affiliation(s)
- Shalini Mathpal
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biosciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Tushar Joshi
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - P Priyamvada
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biosciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
- Department of Biosciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| |
Collapse
|
16
|
Fu ZQ, Geisbrecht BV, Bouyain S, Dyda F, Chrzas J, Kandavelu P, Miller DJ, Wang BC. I / σ I vs {Rmerg, Rmeas, Rpim, CC1/2} for Crystal Diffraction Data Quality Evaluation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.10.627855. [PMID: 39713313 PMCID: PMC11661158 DOI: 10.1101/2024.12.10.627855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
X-ray crystal diffraction has provided atomic-level structural information on biological macromolecules. Data quality determines the reliability of structural models. In most cases, multiple data sets are available from different crystals and/or collected with different experimental settings. Reliable metrics are critical to rank and select the data set with the highest quality. Many measures have been created or modified for data quality evaluation. However, some are duplicate in functionality, and some are likely misused due to misunderstanding, which causes confusion or problems, especially at synchrotron beamlines where experiments proceed quickly. In this work, these measures are studied through both theoretical analysis and experimental data with various characteristics, which demonstrated that: 1). {Rmerg, Rmeas, Rpim, CC1/2} all measure the equivalence of reflections, and the low-shell values of these metrics can be used as reliable indicators for correctness (or trueness) of Laue symmetry; 2). High-shell I / σ I is a reliable and better indicator to select resolution cutoff while the overall value measures the overall strength of the data.
Collapse
Affiliation(s)
- Zheng-Qing Fu
- SER-CAT, Advanced Photon Source, Argonne National Laboratory Argonne, IL 60439, USA
- Department of Biochemistry & Molecular Biology, University of Georgia Athens, GA 30602, USA
| | - Brian V Geisbrecht
- Department of Biochemistry & Molecular Biophysics, Kansas State University Manhattan, KS 66506 USA
| | - Samuel Bouyain
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri - Kansas City Kansas City, MO 64110, USA
| | - Fred Dyda
- Laboratory of Molecular Biology, The National Institute of Diabetes and Digestive and Kidney Diseases Bethesda, MD 20892-0560. USA
| | - John Chrzas
- SER-CAT, Advanced Photon Source, Argonne National Laboratory Argonne, IL 60439, USA
- Department of Biochemistry & Molecular Biology, University of Georgia Athens, GA 30602, USA
| | - Palani Kandavelu
- SER-CAT, Advanced Photon Source, Argonne National Laboratory Argonne, IL 60439, USA
- Department of Biochemistry & Molecular Biology, University of Georgia Athens, GA 30602, USA
| | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital Memphis, TN 38105
| | - Bi-Cheng Wang
- SER-CAT, Advanced Photon Source, Argonne National Laboratory Argonne, IL 60439, USA
- Department of Biochemistry & Molecular Biology, University of Georgia Athens, GA 30602, USA
| |
Collapse
|
17
|
Kieffer J, Orlans J, Coquelle N, Debionne S, Basu S, Homs A, Santoni G, De Sanctis D. Application of signal separation to diffraction image compression and serial crystallography. J Appl Crystallogr 2025; 58:138-153. [PMID: 39917186 PMCID: PMC11798513 DOI: 10.1107/s1600576724011038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/13/2024] [Indexed: 02/09/2025] Open
Abstract
We present here a methodology for real-time analysis of diffraction images acquired at a high frame rate (925 Hz) and its application to macromolecular serial crystallography at ESRF. We introduce a new signal-separation algorithm, able to distinguish the amorphous (or powder diffraction) component from the diffraction signal originating from single crystals. It relies on the ability to work efficiently in azimuthal space and is implemented in pyFAI, the fast azimuthal integration library. Two applications are built upon this separation algorithm: a lossy compression algorithm and a peak-picking algorithm. The performances of both are assessed by comparing data quality after reduction with XDS and CrystFEL.
Collapse
Affiliation(s)
- Jérôme Kieffer
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Julien Orlans
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Nicolas Coquelle
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Samuel Debionne
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Shibom Basu
- EMBL Grenoble, 71 avenue des Martyrs, CS 90181, 38042 Grenoble Cedex 9, France
| | - Alejandro Homs
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Gianluca Santoni
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| | - Daniele De Sanctis
- European Synchrotron Radiation Facility, 71 avenue des Martyrs, CS 40220, 38043 Grenoble Cedex 9, France
| |
Collapse
|
18
|
Azzolino VN, Shaqra AM, Ali A, Kurt Yilmaz N, Schiffer CA. Structural Analysis of Inhibitor Binding to Enterovirus-D68 3C Protease. Viruses 2025; 17:75. [PMID: 39861864 PMCID: PMC11768739 DOI: 10.3390/v17010075] [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: 12/18/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Enterovirus-D68 (EV68) continues to present as a global health issue causing respiratory illness and outbreaks associated with long-lasting neurological disease, with no antivirals or specific treatment options. The development of antiviral therapeutics, such as small-molecule inhibitors that target conserved proteins like the enteroviral 3C protease, remains to be achieved. While various 3C inhibitors have been investigated, their design does not consider the potential emergence of drug resistance mutations. For other antivirals where resistance has been a challenge, we have demonstrated that the likelihood of resistance can be minimized by designing inhibitors that leverage the evolutionary constraints of the target. Here, we characterize a series of 3C inhibitors against EV68-3C protease through enzyme inhibition, protein crystallography, and structural analysis. We have determined and analyzed three high-resolution inhibitor-bound crystal structures of EV68-3C protease, which revealed possible sites of resistance mutations, a key structural water molecule conserved during ligand binding, and the conformational flexibility of the catalytic histidine H40. This structural analysis combined with enzymatic assays provides insights for the rational design of inhibitors that are robust against resistance toward developing antiviral treatments for EV68 infections.
Collapse
Affiliation(s)
| | | | | | | | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (V.N.A.); (A.M.S.); (A.A.); (N.K.Y.)
| |
Collapse
|
19
|
Šoltysová M, Güixens-Gallardo P, Sieglová I, Soldánová A, Krejčiříková V, Fábry M, Brynda J, Khoroshyy P, Hocek M, Řezáčová P. Using environment-sensitive tetramethylated thiophene-BODIPY fluorophores in DNA probes for studying effector-induced conformational changes of protein-DNA complexes. RSC Chem Biol 2025:d4cb00260a. [PMID: 39822774 PMCID: PMC11734750 DOI: 10.1039/d4cb00260a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 12/26/2024] [Indexed: 01/19/2025] Open
Abstract
The LutR protein represses the transcription of genes encoding enzymes for the utilization of l-lactate in Bacillus subtilis through binding to a specific DNA region. In this study, we employed oligonucleotide probes modified by viscosity-sensitive tetramethylated thiophene-BODIPY fluorophores to investigate the impact of selected metabolites on the LutR-DNA complex. Our goal was to identify the effector molecule whose binding alters the protein-DNA affinity, thereby enabling gene transcription. The designed DNA probes exhibited distinctive responses to the binding and release of the protein, characterized by significant alterations in fluorescence lifetime. Through this method, we have identified l-lactate as the sole metabolite exerting a substantial modulating effect on the protein-DNA interaction and thus confirmed its role as an effector molecule. Moreover, we showed that our approach was able to follow conformation changes affecting affinity, which were not captured by other methods commonly used to study the protein-DNA interaction, such as electro-mobility shift assays and florescence anisotropy binding studies. This work underlines the potential of environment-sensitive fluorophore-linked nucleotide modifications, i.e. dCTBdp, for studying the dynamics and subtle changes of protein-DNA interactions.
Collapse
Affiliation(s)
- Markéta Šoltysová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Pedro Güixens-Gallardo
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Irena Sieglová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Anna Soldánová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Veronika Krejčiříková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Milan Fábry
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Jiří Brynda
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Petro Khoroshyy
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| | - Michal Hocek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
- Department of Organic Chemistry, Faculty of Science, Charles University Hlavova 8 CZ-12843 Prague 2 Czechia
| | - Pavlína Řezáčová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences Flemingovo n. 2 Prague 6 Czechia
| |
Collapse
|
20
|
Kim DH, Lee YC, Jin C, Kang SM, Kang SJ, Kang HS, Lee BJ. Structural and Functional Insight Into YefM-YoeB Complex of Toxin-Antitoxin System From Streptococcus pneumoniae. J Cell Biochem 2025; 126:e30672. [PMID: 39530329 DOI: 10.1002/jcb.30672] [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: 08/09/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Streptococcus pneumonia is a Gram-positive and facultative anaerobic bacterium that causes a number of diseases, including otitis media, community-acquired pneumonia, sepsis, and meningitis. With the emergence of antibiotic-resistant strains, there is an urgent need to develop antibiotics with a novel mechanism. The toxin-antitoxin (TA) system, which is primarily found in prokaryotes, consists of a toxin and its equivalent antitoxin genes. The YefM-YoeB module is a Type II TA system, where the YoeB toxin functions as a putative mRNA interferase upon activation, while the YefM antitoxin acts as a transcription repressor by binding to its promoter region along with YoeB. In this study, we determined the crystal structure of the YefM-YoeB complex from S. pneumoniae TIGR4 to comprehend the binding mechanism of the TA system. Furthermore, an in vitro ribonuclease activity assay was conducted to identify the ribonuclease activity of the YoeB toxin. Additionally, furthermore, the oligomeric state of the YefM-YoeB complex in solution was investigated, and a DNA-binding mode was proposed. These structural and functional insights into the YefM-YoeB complex could provide valuable information for the development of novel antibiotics targeting S. pneumonia-associated diseases.
Collapse
Affiliation(s)
- Do-Hee Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Yong-Chan Lee
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Chenglong Jin
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Mastermeditech Ltd., Seoul, Republic of Korea
| | - Sung-Min Kang
- College of Pharmacy, Duksung Women's University, Seoul, Republic of Korea
| | - Su-Jin Kang
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| | - Hoon-Seok Kang
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea
| | - Bong-Jin Lee
- Mastermeditech Ltd., Seoul, Republic of Korea
- College of Pharmacy, Ajou University, Suwon, Republic of Korea
| |
Collapse
|
21
|
Neißner K, Keller H, Kirchner L, Düsterhus S, Duchardt-Ferner E, Averhoff B, Wöhnert J. The structural basis for high-affinity c-di-GMP binding to the GSPII-B domain of the traffic ATPase PilF from Thermus thermophilus. J Biol Chem 2025; 301:108041. [PMID: 39615687 PMCID: PMC11731258 DOI: 10.1016/j.jbc.2024.108041] [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: 06/10/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
c-di-GMP is an important second messenger in bacteria regulating, for example motility, biofilm formation, cell wall biosynthesis, infectivity, and natural transformability. It binds to a multitude of intracellular receptors. This includes proteins containing general secretory pathway II (GSPII) domains such as the N-terminal domain of the Vibrio cholerae ATPase MshE (MshEN) which binds c-di-GMP with two copies of a 24-amino acids sequence motif. The traffic ATPase PilF from Thermus thermophilus is important for type IV pilus biogenesis, twitching motility, surface attachment, and natural DNA-uptake and contains three consecutive homologous GPSII domains. We show that only two of these domains bind c-di-GMP and define the structural basis for the exceptional high affinity of the GSPII-B domain for c-di-GMP, which is 83-fold higher than that of the prototypical MshEN domain. Our work establishes an extended consensus sequence for the c-di-GMP-binding motif and highlights the role of hydrophobic residues for high-affinity recognition of c-di-GMP. Our structure is the first example for a c-di-GMP-binding domain not relying on arginine residues for ligand recognition. We also show that c-di-GMP-binding induces local unwinding of an α-helical turn as well as subdomain reorientation to reinforce intermolecular contacts between c-di-GMP and the C-terminal subdomain. Abolishing c-di-GMP binding to GSPII-B reduces twitching motility and surface attachment but not natural DNA-uptake. Overall, our work contributes to a better characterization of c-di-GMP binding in this class of effector domains, allows the prediction of high-affinity c-di-GMP-binding family members, and advances our understanding of the importance of c-di-GMP binding for T4P-related functions.
Collapse
Affiliation(s)
- Konstantin Neißner
- Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Heiko Keller
- Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Lennart Kirchner
- Molecular Microbiology and Bioenergetics, Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Stefanie Düsterhus
- Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Elke Duchardt-Ferner
- Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Beate Averhoff
- Molecular Microbiology and Bioenergetics, Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany
| | - Jens Wöhnert
- Institute for Molecular Biosciences, Goethe-University Frankfurt/M., Frankfurt, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-University Frankfurt/M., Frankfurt, Germany.
| |
Collapse
|
22
|
Trujillo J, Fung R, Shankar MK, Schwander P, Hosseinizadeh A. Filling data analysis gaps in time-resolved crystallography by machine learning. STRUCTURAL DYNAMICS (MELVILLE, N.Y.) 2025; 12:014101. [PMID: 39868355 PMCID: PMC11758283 DOI: 10.1063/4.0000280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025]
Abstract
There is a growing understanding of the structural dynamics of biological molecules fueled by x-ray crystallography experiments. Time-resolved serial femtosecond crystallography (TR-SFX) with x-ray Free Electron Lasers allows the measurement of ultrafast structural changes in proteins. Nevertheless, this technique comes with some limitations. One major challenge is the quality of data from TR-SFX measurements, which often faces issues like data sparsity, partial recording of Bragg reflections, timing errors, and pixel noise. To overcome these difficulties, conventionally, large volumes of data are collected and grouped into a few temporal bins. The data in each bin are then averaged and paired with the mean of their corresponding jittered timestamps. This procedure provides one structure per bin, resulting in a limited number of averaged structures for the entire time interval spanned by the experiment. Therefore, the information on ultrafast structural dynamics at high temporal resolution is lost. This has initiated research for advanced methods of analyzing experimental TR-SFX data beyond the standard binning and averaging method. To address this problem, we use a machine learning algorithm called Nonlinear Laplacian Spectral Analysis (NLSA), which has emerged as a promising technique for studying the dynamics of complex systems. In this work, we demonstrate the power of this algorithm using synthetic x-ray diffraction snapshots from a protein with significant data incompleteness, timing uncertainties, and noise. Our study confirms that NLSA is a suitable approach that effectively mitigates the effects of these artifacts in TR-SFX data and recovers accurate structural dynamics information hidden in such data.
Collapse
Affiliation(s)
- Justin Trujillo
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N. Maryland Ave, Milwaukee, Wisconsin 53211, USA
| | - Russell Fung
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N. Maryland Ave, Milwaukee, Wisconsin 53211, USA
| | - Madan Kumar Shankar
- Department of Chemistry-BMC Biochemistry, Uppsala University, Husargatan 3, Uppsala 75237, Sweden
| | - Peter Schwander
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N. Maryland Ave, Milwaukee, Wisconsin 53211, USA
| | - Ahmad Hosseinizadeh
- Department of Physics, University of Wisconsin-Milwaukee, 3135 N. Maryland Ave, Milwaukee, Wisconsin 53211, USA
| |
Collapse
|
23
|
Oda K, Komaguchi K, Matoba Y. Copper inactivates DcsB by oxidizing the metal ligand Cys86 to sulfinic acid. FEBS J 2024; 291:5486-5505. [PMID: 39563074 DOI: 10.1111/febs.17325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/25/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024]
Abstract
Nω-hydroxy-l-arginine amidinohydrolase (EC:3.5.3.25), an enzyme in the d-cycloserine (d-CS) biosynthetic pathway of Streptomyces lavendulae, catalyzes the hydrolysis of an arginase inhibitor, Nω-hydroxy-l-arginine, to produce l-ornithine and hydroxyurea, despite being homologous to arginase. Like arginase, the enzyme (DcsB) possesses two manganese ions (MnA and MnB) essential for the enzymatic reaction at the bottom of the cavity formed within the molecule. However, one of the MnA ligands in DcsB is Cys86, whereas the corresponding residues in arginase are histidine. In this study, we determined the crystal structure of Mn-free DcsB to elucidate the installation mechanism of the manganese ions. The flipping of the His111 residue after the formation of the coordination bond to the second manganese ion may facilitate the installation of MnB and the closing of the cavity entrance to retain MnA and MnB at the active site. Copper ions, which are known to be a positive regulator of many secondary metabolites in Streptomyces species, were found to irreversibly inactivate the catalytic activity of DcsB. Mass spectrometric and crystallographic analyses of the Cu(II)-treated DcsB indicated that Cys86 is oxidized to sulfinic acid. The d-CS biosynthesis in the producing microorganism may be negatively regulated by the concentration of intracellular copper ions, which mediates the oxidative stress.
Collapse
Affiliation(s)
- Kosuke Oda
- Department of Virology, Institute of Biomedical and Health Sciences, Hiroshima University, Japan
- Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| | - Kenji Komaguchi
- Department of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, Japan
| | - Yasuyuki Matoba
- Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| |
Collapse
|
24
|
Bilev E, Wild N, Momayyezi P, Sala BM, Sun R, Sandalova T, Marquardt N, Ljunggren HG, Achour A, Hammer Q. Emerging mutation in SARS-CoV-2 facilitates escape from NK cell recognition and associates with enhanced viral fitness. PLoS Pathog 2024; 20:e1012755. [PMID: 39652590 DOI: 10.1371/journal.ppat.1012755] [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: 03/05/2024] [Revised: 12/19/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024] Open
Abstract
In addition to adaptive immunity, natural killer (NK) cells of the innate immune system contribute to the control of viral infections. The HLA-E-restricted SARS-CoV-2 Nsp13232-240 epitope VMPLSAPTL renders infected cells susceptible to NK cells by preventing binding to the inhibitory receptor NKG2A. Here, we report that a recently emerged methionine to isoleucine substitution at position 2 (pM2I) of Nsp13232-240 impairs binding of the mutated epitope to HLA-E and diminishes HLA-E/peptide complex stability. Structural analyses revealed altered occupancy of the HLA-E B-pocket as the underlying cause for reduced presentation and stability of the mutated epitope. Functionally, the reduced presentation of the mutated epitope correlated with elevated binding to NKG2A as well as with increased NK cell inhibition. Moreover, the pM2I mutation associated with enhanced estimated viral fitness and was transmitted to descendants of the SARS-CoV-2 BQ.1 variant. Interestingly, the mutated epitope resembles sequences of related peptides found in endemic common cold-causing human coronaviruses. Altogether, these findings indicate compromised peptide presentation as a viral adaptation to evade NK cell-mediated immunosurveillance by enabling enhanced presentation of self-peptide and restoring NKG2A-dependent inhibition of NK cells.
Collapse
Affiliation(s)
- Eleni Bilev
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nicole Wild
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Pouria Momayyezi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Benedetta Maria Sala
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet & Division of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Renhua Sun
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet & Division of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet & Division of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nicole Marquardt
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet & Division of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
25
|
Jin C, Jeon CH, Kim HW, Kang JM, Choi Y, Kang SM, Lee HH, Kim DH, Han BW, Lee BJ. Structural insight into the distinct regulatory mechanism of the HEPN-MNT toxin-antitoxin system in Legionella pneumophila. Nat Commun 2024; 15:10188. [PMID: 39582057 PMCID: PMC11586414 DOI: 10.1038/s41467-024-54551-0] [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: 01/08/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
HEPN-MNT, a type VII TA module, comprises the HEPN toxin and the MNT antitoxin, which acts as a nucleotidyltransferase that transfers the NMP moiety to the corresponding HEPN toxin, thereby interfering with its toxicity. Here, we report crystal structures of the Legionella pneumophila HEPN-MNT module, including HEPN, AMPylated HEPN, MNT, and the HEPN-MNT complex. Our structural analysis and biochemical assays, suggest that HEPN is a metal-dependent RNase and identify its active site residues. We also elucidate the oligomeric state of HEPN in solution. Interestingly, L. pneumophila MNT, which lacks a long C-terminal α4 helix, controls the toxicity of HEPN toxin via a distinct binding mode with HEPN. Finally, we propose a comprehensive regulatory mechanism of the L. pneumophila HEPN-MNT module based on structural and functional studies. These results provide insight into the type VII HEPN-MNT TA system.
Collapse
Affiliation(s)
- Chenglong Jin
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- MasterMediTech, Seoul, Republic of Korea
| | - Cha-Hee Jeon
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Heung Wan Kim
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Jin Mo Kang
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yuri Choi
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung-Min Kang
- College of Pharmacy, Duksung Women's University, Seoul, Republic of Korea
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Do-Hee Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea.
| | - Byung Woo Han
- The Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Bong-Jin Lee
- MasterMediTech, Seoul, Republic of Korea.
- College of Pharmacy, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
26
|
Cossu M, Catlin D, Elliott SJ, Metcalf WW, Nair SK. Structural organization of pyruvate: ferredoxin oxidoreductase from the methanogenic archaeon Methanosarcina acetivorans. Structure 2024; 32:1963-1972.e3. [PMID: 39265575 PMCID: PMC11956543 DOI: 10.1016/j.str.2024.08.011] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024]
Abstract
Enzymes of the 2-oxoacid:ferredoxin oxidoreductase (OFOR) superfamily catalyze the reversible oxidation of 2-oxoacids to acyl-coenzyme A esters and carbon dioxide (CO2)using ferredoxin or flavodoxin as the redox partner. Although members of the family share primary sequence identity, a variety of domain and subunit arrangements are known. Here, we characterize the structure of a four-subunit family member: the pyruvate:ferredoxin oxidoreductase (PFOR) from the methane producing archaeon Methanosarcina acetivorans (MaPFOR). The 1.92 Å resolution crystal structure of MaPFOR shows a protein fold like those of single- or two-subunit PFORs that function in 2-oxoacid oxidation, including the location of the requisite thiamine pyrophosphate (TPP), and three [4Fe-4S] clusters. Of note, MaPFOR typically functions in the CO2 reductive direction, and structural comparisons to the pyruvate oxidizing PFORs show subtle differences in several regions of catalytical relevance. These studies provide a framework that may shed light on the biochemical mechanisms used to facilitate reductive pyruvate synthesis.
Collapse
Affiliation(s)
- Matteo Cossu
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Daniel Catlin
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sean J Elliott
- Department of Chemistry, Boston University, Boston, MA 02215, USA
| | - William W Metcalf
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Satish K Nair
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| |
Collapse
|
27
|
Banasik M, Napolitano V, Blat A, Abdulkarim K, Plewka J, Czaplewski C, Gieldon A, Kozak M, Wladyka B, Popowicz G, Dubin G. Structural dynamics of the TPR domain of the peroxisomal cargo receptor Pex5 in Trypanosoma. Int J Biol Macromol 2024; 280:135510. [PMID: 39304044 DOI: 10.1016/j.ijbiomac.2024.135510] [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: 06/24/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024]
Abstract
Peroxisomal protein import has been identified as a valid target in trypanosomiases, an important health threat in Central and South America. The importomer is built of multiple peroxins (Pex) and structural characterization of these proteins facilitates rational inhibitor development. We report crystal structures of the Trypanosoma brucei and T. cruzi tetratricopeptide repeat domain (TPR) of the cytoplasmic peroxisomal targeting signal 1 (PTS1) receptor Pex5. The structure of the TPR domain of TbPex5 represents an apo-form of the receptor which, together with the previously determined structure of the complex of TbPex5 TPR and PTS1 demonstrate significant receptor dynamics associated with signal peptide recognition. The structure of the complex of TPR domain of TcPex5 with PTS1 provided in this study details the molecular interactions that guide signal peptide recognition at the atomic level in the pathogenic species currently perceived as the most relevant among Trypanosoma. Small - angle X - ray scattering (SAXS) data obtained in solution supports the crystallographic findings on the compaction of the TPR domains of TbPex5 and TcPex5 upon interaction with the cargo.
Collapse
Affiliation(s)
- Michal Banasik
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
| | - Valeria Napolitano
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Artur Blat
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Karim Abdulkarim
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; Department of Biology, College of Science, Salahaddin University-Erbil, Kirkuk Road, 44002 Erbil, Kurdistan Region, Iraq
| | - Jacek Plewka
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Cezary Czaplewski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland; School of Computational Sciences, Korea Institute for Advanced Study, 85 Hoegiro Dongdaemun-gu, Seoul 02455, Republic of Korea
| | - Artur Gieldon
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland
| | - Maciej Kozak
- Faculty of Physics, Adam Mickiewicz University, 61-614 Poznan, Poland; National Synchrotron Radiation Centre SOLARIS, Jagiellonian University, 30-392 Kraków, Poland
| | - Benedykt Wladyka
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | | | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland.
| |
Collapse
|
28
|
Lo PS, Nisar M, Lakerveld R. Temperature Cycling Induced Deracemization of p-Synephrine in the Presence of Degradation. ACS OMEGA 2024; 9:41936-41943. [PMID: 39398154 PMCID: PMC11465440 DOI: 10.1021/acsomega.4c06807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
The acquisition of enantioenriched organic molecules is crucial in processes where the enantiomeric purity of active ingredients impacts efficacy and safety. Temperature cycling-induced deracemization (TCID) can achieve deracemization, but its effectiveness can be hindered by degradation reactions that influence the kinetics and the achievable enantioenrichment. This work characterizes the impact of degradation on the dynamic development of enantiomeric excess during the TCID process for the p-synephrine hydrochloride salt. The pilot study demonstrates that a maximum enantiomeric excess of 86% R-(-)-p-synephrine can be achieved at an intermediate batch time among all tested conditions. Degradation promoted the crystallization of a dimer with novel solid-state form, disynephrine ether dihydrochloride, which led to a substantial decrease in the slurry density of synephrine, potentially contributing to the observed decline in enantiomeric excess during the TCID process. Batch-to-batch variability in process dynamics and maximum attainable enantiomeric excess was observed, potentially attributable to the sensitivity of the process to uncontrolled initial conditions. These findings underscore the importance of accounting for degradation kinetics in the design and optimization of TCID processes for enantioenrichment.
Collapse
Affiliation(s)
- Po Sang Lo
- Department
of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Madiha Nisar
- Department
of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Department
of Chemistry, The
Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Richard Lakerveld
- Department
of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
29
|
Saha S, Kanaujia SP. Structural and functional characterization of archaeal DIMT1 unveils distinct protein dynamics essential for efficient catalysis. Structure 2024; 32:1760-1775.e7. [PMID: 39146930 DOI: 10.1016/j.str.2024.07.013] [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: 04/10/2024] [Revised: 06/29/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Dimethyladenosine transferase 1 (DIMT1), an ortholog of bacterial KsgA is a conserved protein that assists in ribosome biogenesis by modifying two successive adenosine bases near the 3' end of small subunit (SSU) rRNA. Although KsgA/DIMT1 proteins have been characterized in bacteria and eukaryotes, they are yet unexplored in archaea. Also, their dynamics are not well understood. Here, we structurally and functionally characterized the apo and holo forms of archaeal DIMT1 from Pyrococcus horikoshii. Wild-type protein and mutants were analyzed to capture different transition states, including open, closed, and intermediate states. This study reports a unique inter-domain movement that is needed for substrate (RNA) positioning in the catalytic pocket, and is only observed in the presence of the cognate cofactors S-adenosyl-L-methionine (SAM) or S-adenosyl-L-homocysteine (SAH). The binding of the inhibitor sinefungine, an analog of SAM or SAH, to archaeal DIMT1 blocks the catalytic pocket and renders the enzyme inactive.
Collapse
Affiliation(s)
- Sayan Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Shankar Prasad Kanaujia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
30
|
Dong H, Zhang J, Zhang K, Zhang F, Wang S, Wang Q, Xu C, Yin K, Gu L. The cAMP receptor protein from Gardnerella vaginalis is not regulated by ligands. Commun Biol 2024; 7:1233. [PMID: 39354127 PMCID: PMC11445507 DOI: 10.1038/s42003-024-06957-1] [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: 02/02/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024] Open
Abstract
Overgrowth of Gardnerella vaginalis causes an imbalance in vaginal microecology. The pathogenicity of G. vaginalis is directly regulated by the cAMP receptor protein (CRP). In this study, we resolve the crystal structure of CRPGv at a resolution of 2.22 Å and find some significant differences from homologous proteins. The first 23 amino acids of CRPGv are inserted into the ligand binding pocket, creating a strong steric barrier to ligand entry that has not been seen previously in its homologues. In the absence of ligands, the two α helices used by CRPGv to bind oligonucleotide chains are exposed and can specifically bind TGTGA-N6-TCACA sequences. cAMP and other ligands of CRP homologs are not cofactors of CRPGv. There is no coding gene of the adenylate cyclase, and cAMP could not be identified in G. vaginalis by liquid chromatography tandem mass spectrometry. We speculate that CRPGv may achieve fine regulation through a conformational transformation different from that of its homologous proteins, and this conformational transformation is no longer dependent on small molecules, but may be aided by accessory proteins. CRPGv is the first discovered CRP that is not ligand-regulated, and its active conformation provides a structural basis for drug screening.
Collapse
Affiliation(s)
- Hongjie Dong
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, PR China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Junmei Zhang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, PR China
| | - Kundi Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, PR China
| | - Fengyu Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, PR China
| | - Shuai Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, PR China
| | - Qi Wang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, PR China
| | - Chao Xu
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, PR China
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Kun Yin
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, PR China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China.
| | - Lichuan Gu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, PR China.
| |
Collapse
|
31
|
Medley BJ, Low KE, Irungu JDW, Kipchumba L, Daneshgar P, Liu L, Garber JM, Klassen L, Inglis GD, Boons GJ, Zandberg WF, Abbott DW, Boraston AB. A "terminal" case of glycan catabolism: Structural and enzymatic characterization of the sialidases of Clostridium perfringens. J Biol Chem 2024; 300:107750. [PMID: 39251137 PMCID: PMC11525138 DOI: 10.1016/j.jbc.2024.107750] [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: 07/01/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Sialic acids are commonly found on the terminal ends of biologically important carbohydrates, including intestinal mucin O-linked glycans. Pathogens such as Clostridium perfringens, the causative agent of necrotic enteritis in poultry and humans, have the ability to degrade host mucins and colonize the mucus layer, which involves removal of the terminal sialic acid by carbohydrate-active enzymes (CAZymes). Here, we present the structural and biochemical characterization of the GH33 catalytic domains of the three sialidases of C. perfringens and probe their substrate specificity. The catalytically active domains, which we refer to as NanHGH33, NanJGH33, and NanIGH33, displayed differential activity on various naturally occurring forms of sialic acid. We report the X-ray crystal structures of these domains in complex with relevant sialic acid variants revealing the molecular basis of how each catalytic domain accommodates different sialic acids. NanHGH33 displays a distinct preference for α-2,3-linked sialic acid, but can process α-2,6-linked sialic acid. NanJGH33 and NanIGH33 both exhibit the ability to process α-2,3- and α-2,6-linked sialic acid without any significant apparent preference. All three enzymes were sensitive to generic and commercially available sialidase inhibitors, which impeded sialidase activity in cultures as well as the growth of C. perfringens on sialylated glycans. The knowledge gained in these studies can be applied to in vivo models for C. perfringens growth and metabolism of mucin O-glycans, with a view toward future mitigation of bacterial colonization and infection of intestinal tissues.
Collapse
Affiliation(s)
- Brendon J Medley
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Kristin E Low
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Jackline D W Irungu
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Linus Kipchumba
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Parandis Daneshgar
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Lin Liu
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Jolene M Garber
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada; Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada
| | - Leeann Klassen
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - G Douglas Inglis
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA; Chemical Biology and Drug Discovery, Utrecht University, Utrecht, The Netherlands
| | - Wesley F Zandberg
- Department of Chemistry, Irving K. Barber Faculty of Science, University of British Columbia, Kelowna, British Columbia, Canada.
| | - D Wade Abbott
- Agriculture and Agri-Food Canada, Lethbridge Research and Development Centre, Lethbridge, Alberta, Canada.
| | - Alisdair B Boraston
- Department of Biochemistry & Microbiology, University of Victoria, Victoria, British Columbia, Canada.
| |
Collapse
|
32
|
Grzechowiak M, Sliwiak J, Link A, Ruszkowski M. Legume-type glutamate dehydrogenase: Structure, activity, and inhibition studies. Int J Biol Macromol 2024; 278:134648. [PMID: 39142482 DOI: 10.1016/j.ijbiomac.2024.134648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024]
Abstract
Glutamate dehydrogenases (GDHs) are key enzymes at the crossroads of N and C metabolism in plants. Legumes, whose N metabolism is particularly intricate, possess a unique type of GDH. This study presents an analysis of a legume-type GDH (isoform 2) from Medicago truncatula (MtGDH2). We measured MtGDH2 activity in both the Glu → 2-oxoglutarate (2OG) and 2OG → Glu reaction directions and obtained kinetic parameters for Glu, 2OG, NAD+, and NADH. Inhibition assays revealed that compounds possessing di- or tricarboxylates act as inhibitors of plant GDHs. Interestingly, 2,6-pyridinedicarboxylate (PYR) weakly inhibits MtGDH2 compared to Arabidopsis thaliana homologs. Furthermore, we explored tetrazole derivatives to discover 3-(1H-tetrazol-5-yl)benzoic acid (TBA) as an MtGDH2 inhibitor. The kinetic experiments are supported by six crystal structures, solved as: (i) unliganded enzyme, (ii) trapping the reaction intermediate 2-amino-2-hydroxyglutarate and NAD+, and also complexed with NAD+ and inhibitors such as (iii) citrate, (iv) PYR, (v) isophthalate, and (vi) TBA. The complex with TBA revealed a new mode of action that, in contrast to other inhibitors, prevents domain closure. This discovery points to TBA as a starting point for the development of novel GDH inhibitors to study the functions of GDH in plants and potentially boost biomass production.
Collapse
Affiliation(s)
- Marta Grzechowiak
- Department of Structural Biology of Eukaryotes, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan 61-704, Poland
| | - Joanna Sliwiak
- Laboratory of Protein Engineering, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan 61-704, Poland
| | - Andreas Link
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Milosz Ruszkowski
- Department of Structural Biology of Eukaryotes, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan 61-704, Poland.
| |
Collapse
|
33
|
Saha S, Kanaujia SP. Decoding Substrate Selectivity of an Archaeal RlmCD-like Methyltransferase Through Its Salient Traits. Biochemistry 2024; 63:2477-2492. [PMID: 39350642 DOI: 10.1021/acs.biochem.4c00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2024]
Abstract
5-Methyluridine (m5U) rRNA modifications frequently occur at U747 and U1939 (Escherichia coli numbering) in domains II and IV of the 23S rRNA in Gram-negative bacteria, with the help of S-adenosyl-l-methionine (SAM)-dependent rRNA methyltransferases (MTases), RlmC and RlmD, respectively. In contrast, Gram-positive bacteria utilize a single SAM-dependent rRNA MTase, RlmCD, to modify both corresponding sites. Notably, certain archaea, specifically within the Thermococcales group, have been found to possess two genes encoding SAM-dependent archaeal (tRNA and rRNA) m5U (Arm5U) MTases. Among these, a tRNA-specific Arm5U MTase (PabTrmU54) has already been characterized. This study focused on the structural and functional characterization of the rRNA-specific Arm5U MTase from the hyperthermophilic archaeon Pyrococcus horikoshii (PhRlmCD). An in-depth structural examination revealed a dynamic hinge movement induced by the replacement of the iron-sulfur cluster with disulfide bonds, obstructing the substrate-binding site. It revealed distinctive characteristics of PhRlmCD, including elongated positively charged loops in the central domain and rotational variations in the TRAM domain, which influence substrate selectivity. Additionally, the results suggested that two potential mini-rRNA fragments interact in a similar manner with PhRlmCD at a positively charged cleft at the interface of domains and facilitate dual MTase activities akin to the protein RlmCD. Altogether, these observations showed that Arm5U MTases originated from horizontal gene transfer events, most likely from Gram-positive bacteria.
Collapse
Affiliation(s)
- Sayan Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Shankar Prasad Kanaujia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
34
|
Jana S, Shang J, Hong JY, Fenwick MK, Puri R, Lu X, Melnick AM, Li M, Lin H. A Mitochondria-Targeting SIRT3 Inhibitor with Activity against Diffuse Large B Cell Lymphoma. J Med Chem 2024; 67:15428-15437. [PMID: 39191393 PMCID: PMC11403614 DOI: 10.1021/acs.jmedchem.4c01053] [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: 05/03/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024]
Abstract
Diffuse large B-cell lymphomas (DLBCLs) are heterogeneous cancers that still require better and less toxic treatments. SIRT3, a member of the sirtuin family of NAD+-dependent protein deacylase, is critical for DLBCL growth and survival. A mitochondria-targeted SIRT3 small-molecule inhibitor, YC8-02, exhibits promising activity against DLBCL. However, YC8-02 has several limitations including poor solubility. Here, we report our medicinal chemistry efforts that led to an improved mitochondria-targeted SIRT3 inhibitor, SJ-106C, achieved by using a triethylammonium group, which helps to increase both solubility and SIRT3 inhibition potency. SJ-106C, while still inhibiting SIRT1 and SIRT2, is enriched in the mitochondria to help with SIRT3 inhibition. It is more active against DLBCL than other solid tumor cells and effectively inhibits DLBCL xenograft tumor growth. The findings provide useful insights for the development of SIRT3 inhibitors and mitochondrial targeting agents and further support the notion that SIRT3 is a promising druggable target for DLBCL.
Collapse
Affiliation(s)
- Sadhan Jana
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Jialin Shang
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Jun Young Hong
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Michael K. Fenwick
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Rishi Puri
- College
of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Xuan Lu
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
| | - Ari M. Melnick
- Department
of Medicine, Division of Hematology &
Medical Oncology, Weill Cornell Medicine, New York, New York 10065, United States
| | - Meng Li
- Department
of Medicine, Division of Hematology &
Medical Oncology, Weill Cornell Medicine, New York, New York 10065, United States
| | - Hening Lin
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United States
- Howard
Hughes Medical Institute; Department of Chemistry and Chemical Biology;
Department of Molecular Biology and Genetics Cornell University Ithaca New York 14853 United States
| |
Collapse
|
35
|
Azzolino VN, Shaqra AM, Ali A, Kurt Yilmaz N, Schiffer CA. Elucidating the Substrate Envelope of Enterovirus 68-3C Protease: Structural Basis of Specificity and Potential Resistance. Viruses 2024; 16:1419. [PMID: 39339895 PMCID: PMC11437433 DOI: 10.3390/v16091419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/30/2024] Open
Abstract
Enterovirus-D68 (EV68) has emerged as a global health concern over the last decade with severe symptomatic infections resulting in long-lasting neurological deficits and death. Unfortunately, there are currently no FDA-approved antiviral drugs for EV68 or any other non-polio enterovirus. One particularly attractive class of potential drugs are small molecules inhibitors, which can target the conserved active site of EV68-3C protease. For other viral proteases, we have demonstrated that the emergence of drug resistance can be minimized by designing inhibitors that leverage the evolutionary constraints of substrate specificity. However, the structural characterization of EV68-3C protease bound to its substrates has been lacking. Here, we have determined the substrate specificity of EV68-3C protease through molecular modeling, molecular dynamics (MD) simulations, and co-crystal structures. Molecular models enabled us to successfully characterize the conserved hydrogen-bond networks between EV68-3C protease and the peptides corresponding to the viral cleavage sites. In addition, co-crystal structures we determined have revealed substrate-induced conformational changes of the protease which involved new interactions, primarily surrounding the S1 pocket. We calculated the substrate envelope, the three-dimensional consensus volume occupied by the substrates within the active site. With the elucidation of the EV68-3C protease substrate envelope, we evaluated how 3C protease inhibitors, AG7088 and SG-85, fit within the active site to predict potential resistance mutations.
Collapse
Affiliation(s)
| | | | | | | | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (V.N.A.); (A.M.S.); (A.A.); (N.K.Y.)
| |
Collapse
|
36
|
Zubatyuk R, Biczysko M, Ranasinghe K, Moriarty NW, Gokcan H, Kruse H, Poon BK, Adams PD, Waller MP, Roitberg AE, Isayev O, Afonine PV. AQuaRef: Machine learning accelerated quantum refinement of protein structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.21.604493. [PMID: 39071315 PMCID: PMC11275739 DOI: 10.1101/2024.07.21.604493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Cryo-EM and X-ray crystallography provide crucial experimental data for obtaining atomic-detail models of biomacromolecules. Refining these models relies on library-based stereochemical restraints, which, in addition to being limited to known chemical entities, do not include meaningful noncovalent interactions relying solely on nonbonded repulsions. Quantum mechanical (QM) calculations could alleviate these issues but are too expensive for large molecules. We present a novel AI-enabled Quantum Refinement (AQuaRef) based on AIMNet2 neural network potential mimicking QM at substantially lower computational costs. By refining 41 cryo-EM and 30 X-ray structures, we show that this approach yields atomic models with superior geometric quality compared to standard techniques, while maintaining an equal or better fit to experimental data.
Collapse
Affiliation(s)
- Roman Zubatyuk
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Malgorzata Biczysko
- Faculty of Chemistry, University of Wrocław, F. Joliot-Curie 14, 50-383 Wrocław, Poland
| | | | - Nigel W. Moriarty
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| | - Hatice Gokcan
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - Billy K. Poon
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| | - Paul D. Adams
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA 94720, USA
| | | | - Adrian E. Roitberg
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA
| | - Olexandr Isayev
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Pavel V. Afonine
- Molecular Biophysics & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720-8235, USA
| |
Collapse
|
37
|
Šoltysová M, Škerlová J, Pachl P, Škubník K, Fábry M, Sieglová I, Farolfi M, Grishkovskaya I, Babiak M, Nováček J, Krásný L, Řezáčová P. Structural characterization of two prototypical repressors of SorC family reveals tetrameric assemblies on DNA and mechanism of function. Nucleic Acids Res 2024; 52:7305-7320. [PMID: 38842936 PMCID: PMC11229326 DOI: 10.1093/nar/gkae434] [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: 12/05/2023] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
The SorC family of transcriptional regulators plays a crucial role in controlling the carbohydrate metabolism and quorum sensing. We employed an integrative approach combining X-ray crystallography and cryo-electron microscopy to investigate architecture and functional mechanism of two prototypical representatives of two sub-classes of the SorC family: DeoR and CggR from Bacillus subtilis. Despite possessing distinct DNA-binding domains, both proteins form similar tetrameric assemblies when bound to their respective DNA operators. Structural analysis elucidates the process by which the CggR-regulated gapA operon is derepressed through the action of two effectors: fructose-1,6-bisphosphate and newly confirmed dihydroxyacetone phosphate. Our findings provide the first comprehensive understanding of the DNA binding mechanism of the SorC-family proteins, shedding new light on their functional characteristics.
Collapse
Affiliation(s)
- Markéta Šoltysová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Jana Škerlová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Petr Pachl
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Karel Škubník
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Milan Fábry
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Irena Sieglová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Martina Farolfi
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 20, Czechia
| | - Irina Grishkovskaya
- Research Institute of Molecular Pathology, Campus-ViennaBiocenter 1, 1030 Vienna, Austria
| | - Michal Babiak
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Jiří Nováček
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Libor Krásný
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 20, Czechia
| | - Pavlína Řezáčová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| |
Collapse
|
38
|
Hobbs JK, Boraston AB. The structure of a pectin-active family 1 polysaccharide lyase from the marine bacterium Pseudoalteromonas fuliginea. Acta Crystallogr F Struct Biol Commun 2024; 80:142-147. [PMID: 38935515 PMCID: PMC11229556 DOI: 10.1107/s2053230x2400596x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Pseudoalteromonas fuliginea sp. PS47 is a recently identified marine bacterium that has extensive enzymatic machinery to metabolize polysaccharides, including a locus that targets pectin-like substrates. This locus contains a gene (locus tag EU509_03255) that encodes a pectin-degrading lyase, called PfPL1, that belongs to polysaccharide lyase family 1 (PL1). The 2.2 Å resolution X-ray crystal structure of PfPL1 reveals the compact parallel β-helix fold of the PL1 family. The back side of the core parallel β-helix opposite to the active site is a meandering set of five α-helices joined by lengthy loops. A comparison of the active site with those of other PL1 enzymes suggests a catalytic mechanism that is independent of metal ions, such as Ca2+, but that substrate recognition may require metal ions. Overall, this work provides the first structural insight into a pectinase of marine origin and the first structure of a PL1 enzyme in subfamily 2.
Collapse
Affiliation(s)
- Joanne K. Hobbs
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 1700 STN CSCVictoriaBCV8W 2Y2Canada
| | - Alisdair B. Boraston
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 1700 STN CSCVictoriaBCV8W 2Y2Canada
| |
Collapse
|
39
|
Wlodawer A, Dauter Z, Lubkowski J, Loch JI, Brzezinski D, Gilski M, Jaskolski M. Towards a dependable data set of structures for L-asparaginase research. Acta Crystallogr D Struct Biol 2024; 80:506-527. [PMID: 38935343 PMCID: PMC11220836 DOI: 10.1107/s2059798324005461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The Protein Data Bank (PDB) includes a carefully curated treasury of experimentally derived structural data on biological macromolecules and their various complexes. Such information is fundamental for a multitude of projects that involve large-scale data mining and/or detailed evaluation of individual structures of importance to chemistry, biology and, most of all, to medicine, where it provides the foundation for structure-based drug discovery. However, despite extensive validation mechanisms, it is almost inevitable that among the ∼215 000 entries there will occasionally be suboptimal or incorrect structure models. It is thus vital to apply careful verification procedures to those segments of the PDB that are of direct medicinal interest. Here, such an analysis was carried out for crystallographic models of L-asparaginases, enzymes that include approved drugs for the treatment of certain types of leukemia. The focus was on the adherence of the atomic coordinates to the rules of stereochemistry and their agreement with the experimental electron-density maps. Whereas the current clinical application of L-asparaginases is limited to two bacterial proteins and their chemical modifications, the field of investigations of such enzymes has expanded tremendously in recent years with the discovery of three entirely different structural classes and with numerous reports, not always quite reliable, of the anticancer properties of L-asparaginases of different origins.
Collapse
Affiliation(s)
- Alexander Wlodawer
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Zbigniew Dauter
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Jacek Lubkowski
- Center for Structural Biology, Center for Cancer ResearchNational Cancer InstituteMarylandUSA
| | - Joanna I. Loch
- Department of Crystal Chemistry and Crystal Physics, Faculty of ChemistryJagiellonian UniversityCracowPoland
| | - Dariusz Brzezinski
- Institute of Computing SciencePoznan University of TechnologyPoznanPoland
| | - Miroslaw Gilski
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | - Mariusz Jaskolski
- Institute of Bioorganic ChemistryPolish Academy of SciencesPoznanPoland
- Department of Crystallography, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
40
|
Matoba Y, Oda K, Wataeda M, Kanemori H, Matsuo K. pH-dependent regulation of an acidophilic O-acetylhomoserine sulfhydrylase from Lactobacillus plantarum. Appl Environ Microbiol 2024; 90:e0011824. [PMID: 38568076 PMCID: PMC11107162 DOI: 10.1128/aem.00118-24] [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: 01/23/2024] [Accepted: 03/16/2024] [Indexed: 05/22/2024] Open
Abstract
Bacteria have two routes for the l-methionine biosynthesis. In one route called the direct sulfuration pathway, acetylated l-homoserine is directly converted into l-homocysteine. The reaction using H2S as the second substrate is catalyzed by a pyridoxal 5'-phosphate-dependent enzyme, O-acetylhomoserine sulfhydrylase (OAHS). In the present study, we determined the enzymatic functions and the structures of OAHS from Lactobacillus plantarum (LpOAHS). The LpOAHS enzyme exhibited the highest catalytic activity under the weak acidic pH condition. In addition, crystallographic analysis revealed that the enzyme takes two distinct structures, open and closed forms. In the closed form, two acidic residues are sterically clustered. The proximity may cause the electrostatic repulsion, inhibiting the formation of the closed form under the neutral to the basic pH conditions. We concluded that the pH-dependent regulation mechanism using the two acidic residues contributes to the acidophilic feature of the enzyme. IMPORTANCE In the present study, we can elucidate the pH-dependent regulation mechanism of the acidophilic OAHS. The acidophilic feature of the enzyme is caused by the introduction of an acidic residue to the neighborhood of the key acidic residue acting as a switch for the structural interconversion. The strategy may be useful in the field of protein engineering to change the optimal pH of the enzymes. In addition, this study may be useful for the development of antibacterial drugs because the l-methionine synthesis essential for bacteria is inhibited by the OAHS inhibitors. The compounds that can inhibit the interconversion between the open and closed forms of OAHS may become antibacterial drugs.
Collapse
Affiliation(s)
- Yasuyuki Matoba
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima, Japan
| | - Kosuke Oda
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima, Japan
| | - Maho Wataeda
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima, Japan
| | - Hina Kanemori
- Faculty of Pharmacy, Yasuda Women’s University, Hiroshima, Japan
| | - Koichi Matsuo
- Hiroshima Synchrotron Radiation Center, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
41
|
Flynn JM, Zvornicanin SN, Tsepal T, Shaqra AM, Kurt Yilmaz N, Jia W, Moquin S, Dovala D, Schiffer CA, Bolon DN. Contributions of Hyperactive Mutations in M pro from SARS-CoV-2 to Drug Resistance. ACS Infect Dis 2024; 10:1174-1184. [PMID: 38472113 PMCID: PMC11179160 DOI: 10.1021/acsinfecdis.3c00560] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The appearance and spread of mutations that cause drug resistance in rapidly evolving diseases, including infections by the SARS-CoV-2 virus, are major concerns for human health. Many drugs target enzymes, and resistance-conferring mutations impact inhibitor binding or enzyme activity. Nirmatrelvir, the most widely used inhibitor currently used to treat SARS-CoV-2 infections, targets the main protease (Mpro) preventing it from processing the viral polyprotein into active subunits. Our previous work systematically analyzed resistance mutations in Mpro that reduce binding to inhibitors; here, we investigate mutations that affect enzyme function. Hyperactive mutations that increase Mpro activity can contribute to drug resistance but have not been thoroughly studied. To explore how hyperactive mutations contribute to resistance, we comprehensively assessed how all possible individual mutations in Mpro affect enzyme function using a mutational scanning approach with a fluorescence resonance energy transfer (FRET)-based yeast readout. We identified hundreds of mutations that significantly increased the Mpro activity. Hyperactive mutations occurred both proximal and distal to the active site, consistent with protein stability and/or dynamics impacting activity. Hyperactive mutations were observed 3 times more than mutations which reduced apparent binding to nirmatrelvir in recent studies of laboratory-grown viruses selected for drug resistance. Hyperactive mutations were also about three times more prevalent than nirmatrelvir binding mutations in sequenced isolates from circulating SARS-CoV-2. Our findings indicate that hyperactive mutations are likely to contribute to the natural evolution of drug resistance in Mpro and provide a comprehensive list for future surveillance efforts.
Collapse
Affiliation(s)
- Julia M. Flynn
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Sarah N. Zvornicanin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Tenzin Tsepal
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Ala M. Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Weiping Jia
- Novartis Biomedical Research, Emeryville, CA 94608 USA
| | | | - Dustin Dovala
- Novartis Biomedical Research, Emeryville, CA 94608 USA
| | - Celia A. Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Daniel N.A. Bolon
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| |
Collapse
|
42
|
Dutta A, Kanaujia SP. The Structural Features of MlaD Illuminate its Unique Ligand-Transporting Mechanism and Ancestry. Protein J 2024; 43:298-315. [PMID: 38347327 DOI: 10.1007/s10930-023-10179-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2023] [Indexed: 05/01/2024]
Abstract
The membrane-associated solute-binding protein (SBP) MlaD of the maintenance of lipid asymmetry (Mla) system has been reported to help the transport of phospholipids (PLs) between the outer and inner membranes of Gram-negative bacteria. Despite the availability of structural information, the molecular mechanism underlying the transport of PLs and the ancestry of the protein MlaD remain unclear. In this study, we report the crystal structures of the periplasmic region of MlaD from Escherichia coli (EcMlaD) at a resolution range of 2.3-3.2 Å. The EcMlaD protomer consists of two distinct regions, viz. N-terminal β-barrel fold consisting of seven strands (referred to as MlaD domain) and C-terminal α-helical domain (HD). The protein EcMlaD oligomerizes to give rise to a homo-hexameric ring with a central channel that is hydrophobic and continuous with a variable diameter. Interestingly, the structural analysis revealed that the HD, instead of the MlaD domain, plays a critical role in determining the oligomeric state of the protein. Based on the analysis of available structural information, we propose a working mechanism of PL transport, viz. "asymmetric protomer movement (APM)". Wherein half of the EcMlaD hexamer would rise in the periplasmic side along with an outward movement of pore loops, resulting in the change of the central channel geometry. Furthermore, this study highlights that, unlike typical SBPs, EcMlaD possesses a fold similar to EF/AMT-type beta(6)-barrel and a unique ancestry. Altogether, the findings firmly establish EcMlaD to be a non-canonical SBP with a unique ligand-transport mechanism.
Collapse
Affiliation(s)
- Angshu Dutta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Shankar Prasad Kanaujia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
43
|
Kaur D, Reiss K, Wang J, Batista VS, Brudvig GW, Gunner MR. Occupancy Analysis of Water Molecules inside Channels within 25 Å Radius of the Oxygen-Evolving Center of Photosystem II in Molecular Dynamics Simulations. J Phys Chem B 2024; 128:2236-2248. [PMID: 38377592 DOI: 10.1021/acs.jpcb.3c05367] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
At room temperature and neutral pH, the oxygen-evolving center (OEC) of photosystem II (PSII) catalyzes water oxidation. During this process, oxygen is released from the OEC, while substrate waters are delivered to the OEC and protons are passed from the OEC to the lumen through water channels known as the narrow or the O4 channel, broad or the Cl1 channel, and large or the O1 channel. Protein residues lining the surfaces of these channels play a critical role in stabilizing the hydrogen-bonding networks that assist in the process. We carried out an occupancy analysis to better understand the structural and possible substrate water dynamics in full PSII monomer molecular dynamics (MD) trajectories in both the S1 and S2 states. We find that the equilibrated positions of water molecules derived from MD-derived electron density maps largely match the experimentally observed positions in crystallography. Furthermore, the occupancy reduction in MD simulations of some water molecules inside the single-filed narrow channel also correlates well with the crystallographic data during a structural transition when the S1 state of the OEC advances to the S2 state. The overall reduced occupancies of water molecules are the source of their "vacancy-hopping" dynamic nature inside these channels, unlike water molecules inside an ice lattice where all water molecules have a fixed unit occupancy. We propose on the basis of findings in our structural and molecular dynamics analysis that the water molecule occupying a pocket formed by D1-D61, D1-S169, and O4 of the OEC could be the last steppingstone to enter into the OEC and that the broad channel may be favored for proton transfer.
Collapse
Affiliation(s)
- Divya Kaur
- Department of Chemistry, Brock University, 500 Glenridge Avenue, St. Catharines L2S 3A1, Ontario, Canada
| | - Krystle Reiss
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Jimin Wang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520-8114, United States
| | - Victor S Batista
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - Gary W Brudvig
- Department of Chemistry, Yale University, New Haven, Connecticut 06520-8107, United States
| | - M R Gunner
- Department of Physics, City College of New York New York, New York 10031, United States
| |
Collapse
|
44
|
Xiao Z, Zha J, Yang X, Huang T, Huang S, Liu Q, Wang X, Zhong J, Zheng J, Liang R, Deng Z, Zhang J, Lin S, Dai S. A three-level regulatory mechanism of the aldo-keto reductase subfamily AKR12D. Nat Commun 2024; 15:2128. [PMID: 38459030 PMCID: PMC10923870 DOI: 10.1038/s41467-024-46363-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/23/2024] [Indexed: 03/10/2024] Open
Abstract
Modulation of protein function through allosteric regulation is central in biology, but biomacromolecular systems involving multiple subunits and ligands may exhibit complex regulatory mechanisms at different levels, which remain poorly understood. Here, we discover an aldo-keto reductase termed AKRtyl and present its three-level regulatory mechanism. Specifically, by combining steady-state and transient kinetics, X-ray crystallography and molecular dynamics simulation, we demonstrate that AKRtyl exhibits a positive synergy mediated by an unusual Monod-Wyman-Changeux (MWC) paradigm of allosteric regulation at low concentrations of the cofactor NADPH, but an inhibitory effect at high concentrations is observed. While the substrate tylosin binds at a remote allosteric site with positive cooperativity. We further reveal that these regulatory mechanisms are conserved in AKR12D subfamily, and that substrate cooperativity is common in AKRs across three kingdoms of life. This work provides an intriguing example for understanding complex allosteric regulatory networks.
Collapse
Affiliation(s)
- Zhihong Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jinyin Zha
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xu Yang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Tingting Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shuxin Huang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qi Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xiaozheng Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jie Zhong
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianting Zheng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Rubing Liang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Zixin Deng
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jian Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shuangjun Lin
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
- Haihe Laboratory of Synthetic Biology, Tianjin, 300308, China.
- Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Shaobo Dai
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory on Metabolic & Developmental Sciences, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|
45
|
Afonine PV, Adams PD, Sobolev OV, Urzhumtsev AG. Accounting for nonuniformity of bulk-solvent: A mosaic model. Protein Sci 2024; 33:e4909. [PMID: 38358136 PMCID: PMC10868464 DOI: 10.1002/pro.4909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
A flat mask-based model is almost universally used in macromolecular crystallography to account for disordered (bulk) solvent. This model assumes any voxel of the crystal unit cell that is not occupied by the atomic model is occupied by the solvent. The properties of this solvent are assumed to be exactly the same across the whole volume of the unit cell. While this is a reasonable approximation in practice, there are a number of scenarios where this model becomes suboptimal. In this work, we enumerate several of these scenarios and describe a new generalized approach to modeling the bulk-solvent which we refer to as mosaic bulk-solvent model. The mosaic bulk-solvent model allows nonuniform features of the solvent in the crystal to be accounted for in a computationally efficient way. It is implemented in the computational crystallography toolbox and the Phenix software.
Collapse
Affiliation(s)
- Pavel V. Afonine
- Molecular Biophysics & Integrated Bioimaging DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Paul D. Adams
- Molecular Biophysics & Integrated Bioimaging DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
- Department of BioengineeringUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Oleg V. Sobolev
- Molecular Biophysics & Integrated Bioimaging DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Alexandre G. Urzhumtsev
- Centre for Integrative BiologyInstitut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS–INSERM‐UdSIllkirchFrance
- Université de Lorraine, Faculté des Sciences et TechnologiesVandoeuvre‐les‐NancyFrance
| |
Collapse
|
46
|
Galchenkova M, Tolstikova A, Klopprogge B, Sprenger J, Oberthuer D, Brehm W, White TA, Barty A, Chapman HN, Yefanov O. Data reduction in protein serial crystallography. IUCRJ 2024; 11:190-201. [PMID: 38327201 PMCID: PMC10916297 DOI: 10.1107/s205225252400054x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/15/2024] [Indexed: 02/09/2024]
Abstract
Serial crystallography (SX) has become an established technique for protein structure determination, especially when dealing with small or radiation-sensitive crystals and investigating fast or irreversible protein dynamics. The advent of newly developed multi-megapixel X-ray area detectors, capable of capturing over 1000 images per second, has brought about substantial benefits. However, this advancement also entails a notable increase in the volume of collected data. Today, up to 2 PB of data per experiment could be easily obtained under efficient operating conditions. The combined costs associated with storing data from multiple experiments provide a compelling incentive to develop strategies that effectively reduce the amount of data stored on disk while maintaining the quality of scientific outcomes. Lossless data-compression methods are designed to preserve the information content of the data but often struggle to achieve a high compression ratio when applied to experimental data that contain noise. Conversely, lossy compression methods offer the potential to greatly reduce the data volume. Nonetheless, it is vital to thoroughly assess the impact of data quality and scientific outcomes when employing lossy compression, as it inherently involves discarding information. The evaluation of lossy compression effects on data requires proper data quality metrics. In our research, we assess various approaches for both lossless and lossy compression techniques applied to SX data, and equally importantly, we describe metrics suitable for evaluating SX data quality.
Collapse
Affiliation(s)
- Marina Galchenkova
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | | | - Bjarne Klopprogge
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Janina Sprenger
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Dominik Oberthuer
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Wolfgang Brehm
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Thomas A. White
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Anton Barty
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Henry N. Chapman
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
- Department of Physics, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
- Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Oleksandr Yefanov
- Center for Free-Electron Laser Science CFEL, Deutsche Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| |
Collapse
|
47
|
Ikushiro H, Honda T, Murai Y, Murakami T, Takahashi A, Sawai T, Goto H, Ikushiro SI, Miyahara I, Hirabayashi Y, Kamiya N, Monde K, Yano T. Racemization of the substrate and product by serine palmitoyltransferase from Sphingobacterium multivorum yields two enantiomers of the product from d-serine. J Biol Chem 2024; 300:105728. [PMID: 38325740 PMCID: PMC10912632 DOI: 10.1016/j.jbc.2024.105728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Serine palmitoyltransferase (SPT) catalyzes the pyridoxal-5'-phosphate (PLP)-dependent decarboxylative condensation of l-serine and palmitoyl-CoA to form 3-ketodihydrosphingosine (KDS). Although SPT was shown to synthesize corresponding products from amino acids other than l-serine, it is still arguable whether SPT catalyzes the reaction with d-serine, which is a question of biological importance. Using high substrate and enzyme concentrations, KDS was detected after the incubation of SPT from Sphingobacterium multivorum with d-serine and palmitoyl-CoA. Furthermore, the KDS comprised equal amounts of 2S and 2R isomers. 1H-NMR study showed a slow hydrogen-deuterium exchange at Cα of serine mediated by SPT. We further confirmed that SPT catalyzed the racemization of serine. The rate of the KDS formation from d-serine was comparable to those for the α-hydrogen exchange and the racemization reaction. The structure of the d-serine-soaked crystal (1.65 Å resolution) showed a distinct electron density of the PLP-l-serine aldimine, interpreted as the racemized product trapped in the active site. The structure of the α-methyl-d-serine-soaked crystal (1.70 Å resolution) showed the PLP-α-methyl-d-serine aldimine, mimicking the d-serine-SPT complex prior to racemization. Based on these enzymological and structural analyses, the synthesis of KDS from d-serine was explained as the result of the slow racemization to l-serine, followed by the reaction with palmitoyl-CoA, and SPT would not catalyze the direct condensation between d-serine and palmitoyl-CoA. It was also shown that the S. multivorum SPT catalyzed the racemization of the product KDS, which would explain the presence of (2R)-KDS in the reaction products.
Collapse
Affiliation(s)
- Hiroko Ikushiro
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan.
| | - Takumi Honda
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Yuta Murai
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan; Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan; Division of Applied Bioscience, Graduate School of Agriculture, Hokkaido University, Sapporo, Hokkaido, Japan.
| | - Taiki Murakami
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Aya Takahashi
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Taiki Sawai
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Haruna Goto
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shin-Ichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan
| | - Ikuko Miyahara
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Yoshio Hirabayashi
- RIKEN Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan; Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Nobuo Kamiya
- Research Center for Artificial Photosynthesis, Osaka Metropolitan University, Osaka, Japan
| | - Kenji Monde
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan; Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takato Yano
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan.
| |
Collapse
|
48
|
Kleywegt GJ, Adams PD, Butcher SJ, Lawson CL, Rohou A, Rosenthal PB, Subramaniam S, Topf M, Abbott S, Baldwin PR, Berrisford JM, Bricogne G, Choudhary P, Croll TI, Danev R, Ganesan SJ, Grant T, Gutmanas A, Henderson R, Heymann JB, Huiskonen JT, Istrate A, Kato T, Lander GC, Lok SM, Ludtke SJ, Murshudov GN, Pye R, Pintilie GD, Richardson JS, Sachse C, Salih O, Scheres SHW, Schroeder GF, Sorzano COS, Stagg SM, Wang Z, Warshamanage R, Westbrook JD, Winn MD, Young JY, Burley SK, Hoch JC, Kurisu G, Morris K, Patwardhan A, Velankar S. Community recommendations on cryoEM data archiving and validation. IUCRJ 2024; 11:140-151. [PMID: 38358351 PMCID: PMC10916293 DOI: 10.1107/s2052252524001246] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
In January 2020, a workshop was held at EMBL-EBI (Hinxton, UK) to discuss data requirements for the deposition and validation of cryoEM structures, with a focus on single-particle analysis. The meeting was attended by 47 experts in data processing, model building and refinement, validation, and archiving of such structures. This report describes the workshop's motivation and history, the topics discussed, and the resulting consensus recommendations. Some challenges for future methods-development efforts in this area are also highlighted, as is the implementation to date of some of the recommendations.
Collapse
Affiliation(s)
| | - Paul D. Adams
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- University of California, Berkeley, CA, USA
| | | | | | | | | | | | - Maya Topf
- Birkbeck, University of London, London, United Kingdom
| | | | | | | | | | | | | | | | - Sai J. Ganesan
- University of California at San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Ryan Pye
- EMBL-EBI, Cambridge, United Kingdom
| | | | | | | | | | | | | | | | | | - Zhe Wang
- EMBL-EBI, Cambridge, United Kingdom
| | | | | | - Martyn D. Winn
- Science and Technology Facilities Council, Research Complex at Harwell, Oxon, United Kingdom
| | - Jasmine Y. Young
- RCSB Protein Data Bank, The State University of New Jersey, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Jos S, Poulose R, Kambaru A, Gogoi H, Dalavaikodihalli Nanjaiah N, Padmanabhan B, Mehta B, Padavattan S. Tau-S214 Phosphorylation Inhibits Fyn Kinase Interaction and Increases the Decay Time of NMDAR-mediated Current. J Mol Biol 2024; 436:168445. [PMID: 38218365 DOI: 10.1016/j.jmb.2024.168445] [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: 08/17/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Fyn kinase SH3 domain interaction with PXXP motif in the Tau protein is implicated in AD pathology and is central to NMDAR function. Among seven PXXP motifs localized in proline-rich domain of Tau protein, tandem 5th and 6th PXXP motifs are critical to Fyn-SH3 domain interaction. Here, we report the crystal structure of Fyn-SH3 -Tau (207-221) peptide consisting of 5th and 6th PXXP motif complex to 1.01 Å resolution. Among five AD-specific phosphorylation sites encompassing the 5th and 6th PXXP motifs, only S214 residue showed interaction with SH3 domain. Biophysical studies showed that Tau (207-221) with S214-phosphorylation (pS214) inhibits its interaction with Fyn-SH3 domain. The individual administration of Tau (207-221) with/without pS214 peptides to a single neuron increased the decay time of evoked NMDA current response. Recordings of spontaneous NMDA EPSCs at +40 mV indicate an increase in frequency and amplitude of events for the Tau (207-221) peptide. Conversely, the Tau (207-221) with pS214 peptide exhibited a noteworthy amplitude increase alongside a prolonged decay time. These outcomes underscore the distinctive modalities of action associated with each peptide in the study. Overall, this study provides insights into how Tau (207-221) with/without pS214 affects the molecular framework of NMDAR signaling, indicating its involvement in Tau-related pathogenesis.
Collapse
Affiliation(s)
- Sneha Jos
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Roshni Poulose
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Archanalakshmi Kambaru
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Hemanga Gogoi
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | | | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India
| | - Bhupesh Mehta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| | - Sivaraman Padavattan
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Bangalore 560029, India.
| |
Collapse
|
50
|
Zecchin P, Pecqueur L, Oltmanns J, Velours C, Schünemann V, Fontecave M, Golinelli‐Pimpaneau B. Structure-based insights into the mechanism of [4Fe-4S]-dependent sulfur insertase LarE. Protein Sci 2024; 33:e4874. [PMID: 38100250 PMCID: PMC10806937 DOI: 10.1002/pro.4874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Several essential cellular metabolites, such as enzyme cofactors, contain sulfur atoms and their biosynthesis requires specific thiolation enzymes. LarE is an ATP-dependent sulfur insertase, which catalyzes the sequential conversion of the two carboxylate groups of the precursor of the lactate racemase cofactor into thiocarboxylates. Two types of LarE enzymes are known, one that uses a catalytic cysteine as a sacrificial sulfur donor, and the other one that uses a [4Fe-4S] cluster as a cofactor. Only the crystal structure of LarE from Lactobacillus plantarum (LpLarE) from the first class has been solved. We report here the crystal structure of LarE from Methanococcus maripaludis (MmLarE), belonging to the second class, in the cluster-free (apo-) and cluster-bound (holo-) forms. The structure of holo-MmLarE shows that the [4Fe-4S] cluster is chelated by three cysteines only, leaving an open coordination site on one Fe atom. Moreover, the fourth nonprotein-bonded iron atom was able to bind an anionic ligand such as a phosphate group or a chloride ion. Together with the spectroscopic analysis of holo-MmLarE and the previously reported biochemical investigations of holo-LarE from Thermotoga maritima, these crystal structures support the hypothesis of a reaction mechanism, in which the [4Fe-4S] cluster binds a hydrogenosulfide ligand in place of the chloride anion, thus generating a [4Fe-5S] intermediate, and transfers it to the substrate, as in the case of [4Fe-4S]-dependent tRNA thiolation enzymes.
Collapse
Affiliation(s)
- Paolo Zecchin
- Laboratoire de Chimie des Processus Biologiques, Collège de France, CNRS UMR 8829Sorbonne UniversitéParis cedex 05France
| | - Ludovic Pecqueur
- Laboratoire de Chimie des Processus Biologiques, Collège de France, CNRS UMR 8829Sorbonne UniversitéParis cedex 05France
| | - Jonathan Oltmanns
- Universität of Kaiserslautern‐LandauDepartment of PhysicsKaiserslauternGermany
| | - Christophe Velours
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRSUniversité Paris‐SaclayGif‐sur‐Yvette cedexFrance
- Present address:
Fundamental Microbiology and Pathogenicity LaboratoryUMR 5234 CNRS‐University of Bordeaux, SFR TransBioMedBordeauxFrance
| | - Volker Schünemann
- Universität of Kaiserslautern‐LandauDepartment of PhysicsKaiserslauternGermany
| | - Marc Fontecave
- Laboratoire de Chimie des Processus Biologiques, Collège de France, CNRS UMR 8829Sorbonne UniversitéParis cedex 05France
| | - Béatrice Golinelli‐Pimpaneau
- Laboratoire de Chimie des Processus Biologiques, Collège de France, CNRS UMR 8829Sorbonne UniversitéParis cedex 05France
| |
Collapse
|