151
|
Li X, Guo W, Wen Y, Meng C, Zhang Q, Chen H, Zhao X, Wu B. Structural basis for the RNA binding properties of mouse IGF2BP3. Structure 2025; 33:771-785.e3. [PMID: 39986276 DOI: 10.1016/j.str.2025.01.022] [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: 07/19/2024] [Revised: 01/11/2025] [Accepted: 01/28/2025] [Indexed: 02/24/2025]
Abstract
IGF2BP family proteins (IGF2BPs) contain six tandem RNA-binding domains (RBDs), resulting in highly complex RNA binding properties. Dissecting how IGF2BPs recognize their RNA targets is essential for understanding their regulatory roles in gene expression. Here, we have determined the crystal structures of mouse IGF2BP3 constructs complexed with different RNA substrates. Our structures reveal that the IGF2BP3-RRM12 domains can recognize CA-rich elements up to 5-nt in length, mainly through RRM1. We also captured the antiparallel RNA-binding mode of the IGF2BP3-KH12 domains, with five nucleotides bound by KH1 and two nucleotides bound by KH2. Furthermore, our structural and biochemical studies suggest that the IGF2BP3-KH12 domains could recognize the "zipcode" RNA element within the β-actin mRNA. Finally, we analyzed the similarities and differences of the RNA-binding properties between the KH12 and KH34. Our studies provide structural insights into RNA target recognition by mouse IGF2BP3.
Collapse
Affiliation(s)
- Xiaojia Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Wenting Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yan Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Chunyan Meng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qingrong Zhang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, China
| | - Haitao Chen
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, China
| | - Xiaomiao Zhao
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Baixing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China.
| |
Collapse
|
152
|
Rong L, An J, Chen X, Wang C, Wu J, Wang P, Zheng Y, Wang X, Chai X, Li W, Hu Z, Lu D, Chen GE, Ouyang M, Grimm B, Zhang L, Xu X. LTD coordinates chlorophyll biosynthesis and LIGHT-HARVESTING CHLOROPHYLL A/B-BINDING PROTEIN transport. THE PLANT CELL 2025; 37:koaf068. [PMID: 40138376 PMCID: PMC11979457 DOI: 10.1093/plcell/koaf068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025]
Abstract
Chlorophyll biosynthesis must be tightly coupled to light-harvesting chlorophyll a/b-binding protein (LHCP) biogenesis, as free chlorophyll and its precursors are phototoxic. However, precisely how these 2 processes are coordinated in Arabidopsis (Arabidopsis thaliana) remains elusive. Our previous studies demonstrated the role of LHCP TRANSLOCATION DEFECT (LTD) in delivering LHCPs to the chloroplast via the signal recognition particle-dependent pathway. Here, we show that LTD interacts with and stabilizes the chlorophyll biosynthesis enzymes Mg-protoporphyrin methyltransferase and Mg-protoporphyrin monomethylester (MgPME) cyclase, maintaining their activity. We also demonstrate the direct binding of LTD to MgPME, and through crystal structure analysis, we show that the groove of the LTD dimer is critical for MgPME binding. Thus, we propose that LTD transfers MgPME from Mg-protoporphyrin methyltransferase to the MgPME cyclase. These results elucidate a role for LTD in synchronizing chlorophyll biosynthesis with LHCP transport to ensure the correct insertion of chlorophylls into LHCPs.
Collapse
Affiliation(s)
- Liwei Rong
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing 100093, China
| | - Junhang An
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Xinyue Chen
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Chao Wang
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Jianghao Wu
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Peng Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong 999077, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
- Institute of Biology/Plant Physiology, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Yongxing Zheng
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Xin Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Xin Chai
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Wei Li
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Zhubing Hu
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Dandan Lu
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Guangyu E Chen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Min Ouyang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Bernhard Grimm
- Institute of Biology/Plant Physiology, Humboldt-Universität zu Berlin, Berlin 10115, Germany
| | - Lixin Zhang
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| | - Xiumei Xu
- The Zhongzhou Laboratory for Integrative Biology, State Key Laboratory of Crop Stress Adaptation and Improvement, Henan Key Laboratory of Synthetic Biology and Biomanufacturing, School of Life Sciences, Henan University, Kaifeng 475004, China
- Sanya Institute, Henan University, Sanya 572025, China
| |
Collapse
|
153
|
Pistofidis A, Schmeing TM. Protein ligation for the assembly and study of nonribosomal peptide synthetase megaenzymes. RSC Chem Biol 2025; 6:590-603. [PMID: 39957992 PMCID: PMC11824870 DOI: 10.1039/d4cb00306c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Nonribosomal peptide synthetases (NRPSs) are biosynthetic enzymes found in bacteria and fungi, that synthesize a plethora of pharmaceutically relevant compounds. NRPSs consist of repeating sets of functional domains called modules, and each module is responsible for the incorporation of a single amino acid to the growing peptidyl intermediate. The synthetic logic of an NRPS resembles an assembly line, with growing biosynthesis intermediates covalently attached to the prosthetic 4'-phosphopantetheine (ppant) moieties of T (thiolation or transfer) domains for shuttling within and between modules. Therefore, NRPSs must have each T domain phosphopantetheinylated to be functional, and host organisms encode ppant transferases that affix ppant to T domains. Ppant transferases can be promiscuous with respect to the T domain substrate and with respect to chemical modifications of the ppant thiol, which has been a useful characteristic for study of megaenzymes and other systems. However, defined studies of multimodular megaenzymes, where different analogs are required to be affixed to different T domains within the same multimodular protein, are hindered by this promiscuity. Study of NRPS peptide bond formation, for which two T domains simultaneously deliver substrates to the condensation domain, is a prime example where one would want two T domains bearing different acyl/peptidyl groups. Here, we report a strategy where two NRPS modules that are normally part of the same protein are expressed as separate constructs, modified separately with different acyl-ppants, and then ligated together by sortase A of Staphylococcus aureus or asparaginyl endopeptidase 1 of Oldenlandia affinis (OaAEP1). We assessed various reaction conditions to optimize the ligation reactions and maximize the yield of the complex of interest. Finally, we apply this method in large scale and show it allows the complex built by OaAEP1-mediated ligation to be characterized by X-ray crystallography.
Collapse
Affiliation(s)
- Angelos Pistofidis
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University Montréal QC H3G 0B1 Canada
| | - T Martin Schmeing
- Department of Biochemistry and Centre de Recherche en Biologie Structurale, McGill University Montréal QC H3G 0B1 Canada
| |
Collapse
|
154
|
Mortensen S, Burnim A, Dufault-Thompson K, Lipka AE, Jiang X, Sondermann H. Ancestral proteins trace the emergence of substrate specificity and oligomerization within bacterial DEDDy dinucleases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.30.646021. [PMID: 40236078 PMCID: PMC11996385 DOI: 10.1101/2025.03.30.646021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Nucleases play a crucial role in bacterial physiology, influencing processes such as DNA repair, genome maintenance, and host-pathogen interactions. We recently identified a class of nucleases, diDNases, which are encoded on mobile genetic elements and homologous to the house-keeping nanoRNase C (NrnC). Despite their shared structural fold, diDNases and NrnC orthologs exhibit differences. DiDNases form dimers and preferably cleave DNA dinucleotides, whereas NrnC homologs assemble into octamers that do not discriminate between RNA or DNA dinucleotides. Here, we investigate the evolutionary divergence of these enzymes using ancestral sequence reconstruction. Our results show that both diDNases and NrnC orthologs originated from a dimeric ancestor with intermediate substrate preferences. Structural analyses of ancestral and extant dinucleases provide a molecular rational for how gradual changes in conformation gave rise to substrate preferences, oligomeric state, and catalytic efficiency of these related, yet distinct enzyme clades. These findings provide insights into how small structural modifications enable large-scale changes in molecular assembly and functional specialization harnessing a conserved protein fold. In addition, the preference of the early ancestors for DNA dinucleotides and preservation of this activity in all extant enzymes strongly argues for a biological function of DNA dinucleotides.
Collapse
|
155
|
Zhang G, Huang P, Yuan H, Li E, Chi X, Sun H, Han J, Fang T, Dong Y, Li J, Wang Y, Li J, Chiu S, Yu C. Nasal delivery of secretory IgA confers enhanced neutralizing activity against Omicron variants compared to its IgG counterpart. Mol Ther 2025; 33:1687-1700. [PMID: 40025736 PMCID: PMC11997491 DOI: 10.1016/j.ymthe.2025.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/28/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its multiple variants continue to spread worldwide, causing respiratory symptoms primarily through mucosal infection. The mucosa serves as the primary barrier against viral entry, in which secretory immunoglobulin A (sIgA) plays a critical role in preventing infection. Here, we engineered and characterized a neutralizing monoclonal antibody, ZW2G10, in IgG, monomeric, dimeric, secretory IgA1, and IgA2 formats. All seven forms of the ZW2G10 antibody showed similar thermal stability. sIgA, especially sIgA1, displayed enhanced neutralizing activity against Omicron-lineage BA.2.75, BA.2.76 and BA.4/5 pseudoviruses compared to IgG. Nasal administration of sIgA1 conferred robust protection against the BA.2.76 pseudovirus in ACE2 transgenic mice, and its protective efficacy was superior to that of IgG. The crystal structure of Omicron receptor binding domain (RBD) and ZW2G10 antibody fragment (Fab) complex revealed that ZW2G10 had no clashes with ACE2. Thus, nasal administration of sIgA may serve as a promising tool for the prevention and treatment of Omicron infection.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/chemistry
- Immunoglobulin A, Secretory/administration & dosage
- Immunoglobulin A, Secretory/immunology
- Immunoglobulin A, Secretory/chemistry
- Mice
- COVID-19/immunology
- COVID-19/virology
- COVID-19/prevention & control
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/chemistry
- Administration, Intranasal
- Antibodies, Viral/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/chemistry
- Angiotensin-Converting Enzyme 2/genetics
- Mice, Transgenic
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/administration & dosage
Collapse
Affiliation(s)
- Guanying Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Ping Huang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Hongyu Yuan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Entao Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Xiangyang Chi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Hancong Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Jin Han
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Yunzhu Dong
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Jie Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yaoxing Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China.
| | - Sandra Chiu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China.
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China.
| |
Collapse
|
156
|
Möller C, Terholsen H, Schmöker O, Lê TLA, Wesche J, Schmiade P, Eppendorfer E, Rimkus N, Girbardt B, Böttcher D, Palm GJ, Hoppen J, Lammers M, Greinacher A, Aurich K, Bornscheuer UT. Identification and Protein Engineering of Galactosidases for the Conversion of Blood Type B to Blood Type O. Chembiochem 2025; 26:e202500072. [PMID: 40013737 PMCID: PMC12002104 DOI: 10.1002/cbic.202500072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 02/28/2025]
Abstract
The supply of blood products such as red blood cells poses a challenge due to rising demand and declining donor numbers. Careful matching of blood products of different types is required. Only type O of the blood types A, B, AB and O can be received by any patient without transfusion incompatibilities. Therefore, O-type blood can be considered "universal blood" and is especially needed in emergency situations. In this study, we focused on the conversion of the B antigen by enzymatic deglycosylation to generate the H antigen determining O-type blood. For this, we characterized several previously unstudied α-1,3-galactosidases belonging to the GH110 family. Our findings revealed that the α-1,3-galactosidase from Pedobacter panaciterrae (PpaGal) exhibits superior efficiency compared to previously described galactosidases. We further increased the activity of PpaGal by 2.5-fold using site-directed mutagenesis. Moreover, we solved two crystal structures of PpaGal, one in the apo-state and another in complex with d-galactose. The combination of our mutagenesis study with the solved crystal structures provides valuable information to guide further optimization of PpaGal or other B antigen converting enzymes paving the way for the easier production of universal blood from B-type blood.
Collapse
Affiliation(s)
- Christina Möller
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Henrik Terholsen
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Ole Schmöker
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Thi Linh Anne Lê
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Jan Wesche
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Paula Schmiade
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Esther Eppendorfer
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Niklas Rimkus
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Britta Girbardt
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Dominique Böttcher
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| | - Gottfried J. Palm
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Jens Hoppen
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Michael Lammers
- Institute of BiochemistryDepartment of Synthetic and Structural BiochemistryFelix-Hausdorff-Strasse 217489GreifswaldGermany
| | - Andreas Greinacher
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Konstanze Aurich
- Institute of Transfusion MedicineUniversity Medicine GreifswaldSauerbruchstrasse17475GreifswaldGermany
| | - Uwe T. Bornscheuer
- Institute of Biochemistry, Department of Biotechnology and Enzyme CatalysisUniversity GreifswaldFelix-Hausdorff-Straße 417487GreifswaldGermany
| |
Collapse
|
157
|
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
|
158
|
Pierce CT, Tan P, Greenberg LR, Walsh ME, Shi K, Nguyen AH, Meixner EL, Sarak S, Aihara H, Evans RL, Kazlauskas RJ. Crystal structures of forty- and seventy-one-substitution variants of hydroxynitrile lyase from rubber tree. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.30.646168. [PMID: 40236237 PMCID: PMC11996369 DOI: 10.1101/2025.03.30.646168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The α/β-hydrolase fold family contains mostly esterases but includes other enzymes such as hydroxynitrile lyase from Hevea brasiliensis (rubber tree, Hb HNL). Hb HNL shares 44% sequence identity and a Ser-His-Asp catalytic triad with esterase SABP2 (salicylic acid binding protein 2 from Nicotiana tabacum (tobacco)). To identify how large a region within Hb HNL influences the positions of the catalytic residues, we created variants where increasingly large regions surrounding the substrate-binding site had identical amino acid sequences to those in SABP2. Variant HNL40 contains 40 mutations (two inserted amino acid residues, 38 substitutions), shares 59% sequence identity with SABP2, and is identical in sequence to SABP2 within 10 Å of the substrate-binding site. Variant HNL71 contains 31 additional substitutions for a total of 71 changes (two insertions, 69 substitutions) and shares 71% sequence identity with SABP2. The sequences within 14 Å of the substrate-binding site are identical in SABP2 and HNL71. The crystal structures of HNL40 and HNL71 show that the positions of main chain Cɑ atoms move from their positions in Hb HNL to more closely match those in SABP2 (RMSD = 0.51 Å over 235 Cɑ atoms for HNL40, 0.41 Å over 219 Cɑ atoms for HNL71) and even more closely in the region within 10 Å of the substrate-binding site (RMSD = 0.38 Å over 58 Cɑ atoms for HNL40, 0.28 Å over 53 Cɑ atoms for HNL71). The pattern of tunnels in HNL40 and HNL71 are similar to each other and intermediate between the pattern in Hb HNL and SABP2. Synopsis Variants HNL40 and HNL71 of hydroxynitrile lyase from Hevea brasiliensis contain 40 and 71 mutations, respectively, to make regions surrounding the substrate-binding site identical in sequence to esterase SABP2. X-ray structures reveal increasing similarities to SABP2 in HNL40 and HNL71. PDB reference: hydroxynitrile lyase from Hevea brasiliensis with forty mutations, 8SNI, hydroxynitrile lyase from Hevea brasiliensis with seventy-one mutations, 9CLR.
Collapse
|
159
|
Chandit C, Hengphasatporn K, Donsuy P, Shigeta Y, Wangkanont K. Structure and catalytic activity of a dihydrofolate reductase-like enzyme from Leptospira interrogans. Int J Biol Macromol 2025; 298:139931. [PMID: 39824401 DOI: 10.1016/j.ijbiomac.2025.139931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
A dihydrofolate reductase (DHFR)-like enzyme from Leptospira interrogans (LiDHFRL) was cloned and the recombinant protein was characterized. Sequence alignment suggested that the enzyme lacked the conserved catalytic residues found in DHFR. Indeed, LiDHFRL did not catalyze the reduction of dihydrofolate by either NADH or NADPH. X-ray crystallography revealed that LiDHFRL bound NADP(H) tightly, but its active site architecture was vastly different from that of Escherichia coli DHFR (EcDHFR) and other DHFRLs. Interestingly, vanillin could serve as a substrate for LiDHFRL, demonstrating that LiDHFRL is a functional enzyme. A putative vanillin binding mode was proposed.
Collapse
Affiliation(s)
- Charin Chandit
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence in Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kowit Hengphasatporn
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Pattraporn Donsuy
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence in Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Kittikhun Wangkanont
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence in Molecular Crop, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
160
|
Upadhyay S, Bhardwaj M, Kumar SP, Khan S, Kumar A, Hassan MI. Impact of Cancer-Associated PKM2 Mutations on Enzyme Activity and Allosteric Regulation: Structural and Functional Insights into Metabolic Reprogramming. Biochemistry 2025; 64:1463-1475. [PMID: 40080100 DOI: 10.1021/acs.biochem.5c00009] [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: 03/15/2025]
Abstract
Mammalian pyruvate kinase M2 (PKM2) is a key regulator of glycolysis and is highly expressed in proliferative tissues including tumors. Mutations in PKM2 have been identified in various cancers, but their effects on enzyme activity and regulation are not fully understood. This study investigates the structural and functional effects of cancer-associated PKM2 mutations on enzyme kinetics, allosteric regulation, and oligomerization. Using computational modeling, X-ray crystallography, and biochemical assays, we demonstrated how these mutations impact PKM2 activity, substrate binding, and allosteric activation via fructose-1,6-bisphosphate (FBP), contributing to altered enzyme function. In this study, we characterized four cancer-associated PKM2 mutations (P403A, C474S, R516C, and L144P) using computational, structural, and biochemical approaches. Computational modeling revealed disruptions in allosteric signaling pathways, particularly affecting the communication between regulatory sites and the active site. X-ray crystallography demonstrated local conformational changes in the hinge and FBP-binding regions, leading to a shift from the active tetrameric state to a less active dimeric state, particularly in the C474S and R516C mutants. The mutants exhibited reduced maximal velocity, reduced substrate affinity, and altered activation by the allosteric activator fructose-1,6-bisphosphate (FBP). Under alkaline pH conditions, mimicking the tumor microenvironment, these mutations further destabilized the PKM2 oligomeric state, favoring the formation of lower-order species. Our findings suggest that PKM2 is highly sensitive to mutations, and these alterations may contribute to metabolic reprogramming in cancer cells by impairing its enzymatic regulation.
Collapse
Affiliation(s)
- Saurabh Upadhyay
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
| | - Mohit Bhardwaj
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
| | - Sivakumar Prasanth Kumar
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Shumayila Khan
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi 110016, India
- International Health Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India
| | - Ashwani Kumar
- Macromolecular Crystallography Section, Beamline Development & Application Section, Bhabha Atomic Research Center, Trombay, Mumbai 400085, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
161
|
Liu J, Wu C, Liu Y, Chen Q, Ding Y, Lin Z, Pan L, Xiao K, Li J, Liu Z, Liu W. Structural insights into the dual Ca 2+-sensor-mediated activation of the PPEF phosphatase family. Nat Commun 2025; 16:3120. [PMID: 40169586 PMCID: PMC11962071 DOI: 10.1038/s41467-025-58261-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Abstract
Serine/threonine-protein phosphatases with EF-hands (PPEFs) are a family of highly conserved proteins implicated in cancer and neuronal degeneration. The initially characterized member, Drosophila melanogaster retinal degeneration C (RDGC) contains a calmodulin (CaM)-interacting extended-IQ motif and a Ca2+-binding EF-like/EF-hand tandem. However, the molecular regulation of PPEF is poorly understood. In this study, we use cryogenic-electron microscopy to delineate the structures of the RDGC/CaM holoenzyme. In the absence of Ca2+, CaM and the EF-like/EF-hand tandem allow the extended-IQ motif to block substrate access to the catalytic sites, constituting an auto-inhibitory mechanism. Upon Ca2+ binding, CaM and the EF-like/EF-hand tandem drive drastic conformational changes in the extended-IQ motif to unlock the catalytic sites. This dual Ca2+-sensor-mediated activation is evolutionarily conserved in mammals. This study provides mechanistic insight into the molecular activation of PPEFs, paving the way for the development of therapeutic strategies for PPEF-related human diseases.
Collapse
Affiliation(s)
- Jia Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China
| | - Cang Wu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Yuyang Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China
| | - Qiangou Chen
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China
| | - Yuzhen Ding
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China
| | - Zhiqiao Lin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Lifeng Pan
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Kang Xiao
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen, China
| | - Jianchao Li
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, Guangdong, China.
- Institute of Geriatric Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China.
| |
Collapse
|
162
|
Baral J, Song D, Edwards TE, Dranow DM, Lorimer DD, Staker BL, Myler P, Smith CL. The crystal structures of apo and tryptophan-bound tryptophanyl-tRNA synthetase from Neisseria gonorrhoeae. Acta Crystallogr F Struct Biol Commun 2025; 81:130-137. [PMID: 40023769 PMCID: PMC11970124 DOI: 10.1107/s2053230x25001311] [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: 11/06/2024] [Accepted: 02/13/2025] [Indexed: 03/04/2025] Open
Abstract
Neisseria gonorrhoeae, the causative agent of the human disease gonorrhea, is the second most common sexually transmitted pathogen in the United States. Gonorrhea has a significantly high morbidity rate due to the ability of N. gonorrhoeae to rapidly develop antibiotic resistance. In this paper, crystal structures of tryptophanyl-tRNA synthetase (TrpRS) from N. gonorrhoeae (NgTrpRS) were determined in both its apo form and in complex with tryptophan. The structures reveal conserved HIGH and KMSKS motifs critical for ATP binding and catalysis, and highlight conformational changes in the active site upon tryptophan binding, including a methionine flip and the rearrangement of hydrogen-bonding residues. Structural alignments with human TrpRS isoforms demonstrate significant differences between the bacterial and human cytosolic forms, particularly in their active sites. While NgTrpRS and human mitochondrial TrpRS share conserved catalytic residues that are essential for binding tryptophan and indolmycin, the cytosolic TrpRS contains substitutions that introduce steric hindrance, limiting the binding of indolmycin. These results provide insight for the development of inhibitors targeting bacterial TrpRS without affecting the human mitochondrial or cytosolic isoforms, contributing to efforts to combat antibiotic-resistant N. gonorrhoeae infections.
Collapse
Affiliation(s)
- Jessika Baral
- Department of BiologyWashington University in St LouisSt LouisMO63130USA
| | - David Song
- Department of BiologyWashington University in St LouisSt LouisMO63130USA
| | | | | | | | - Bart L. Staker
- Center for Infectious Disease Research307 Westlake Avenue North, Suite 500SeattleWA98109USA
| | - Peter Myler
- Center for Infectious Disease Research307 Westlake Avenue North, Suite 500SeattleWA98109USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Craig L. Smith
- Department of BiologyWashington University in St LouisSt LouisMO63130USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| |
Collapse
|
163
|
Youle RL, Lista MJ, Bouton C, Kunzelmann S, Wilson H, Cottee MA, Purkiss AG, Morris ER, Neil SJD, Taylor IA, Swanson CM. Structural and functional characterization of the extended-diKH domain from the antiviral endoribonuclease KHNYN. J Biol Chem 2025; 301:108336. [PMID: 39984050 PMCID: PMC11997328 DOI: 10.1016/j.jbc.2025.108336] [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: 12/23/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
Zinc finger antiviral protein (ZAP) binds CpG dinucleotides in viral RNA and targets them for decay. ZAP interacts with several cofactors to form the ZAP antiviral system, including KHNYN, a multidomain endoribonuclease required for ZAP-mediated RNA decay. However, it is unclear how the individual domains in KHNYN contribute to its activity. Here, we demonstrate that the KHNYN amino-terminal extended-diKH (ex-diKH) domain is required for antiviral activity and present its crystal structure. The structure belongs to a rare group of KH-containing domains, characterized by a noncanonical arrangement between two type 1 KH modules, with an additional helical bundle. N4BP1 is a KHNYN paralog with an ex-diKH domain that functionally complements the KHNYN ex-diKH domain. Interestingly, the ex-diKH domain structure is present in N4BP1-like proteins in lancelets, which are basal chordates, indicating that it is evolutionarily ancient. While many KH domains demonstrate RNA binding activity, biolayer interferometry and electrophoretic mobility shift assays indicate that the KHNYN ex-diKH domain does not bind RNA. Furthermore, residues required for canonical KH domains to bind RNA are not required for KHNYN antiviral activity. By contrast, an inter-KH domain cleft in KHNYN is a potential protein-protein interaction site, and mutations that eliminate arginine salt bridges at the edge of this cleft decrease KHNYN antiviral activity. This suggests that this domain could be a binding site for an unknown KHNYN cofactor.
Collapse
Affiliation(s)
- Rebecca L Youle
- Department of Infectious Diseases, King's College London, London, United Kingdom; Macromolecular Structure Laboratory, The Francis Crick Institute, London, United Kingdom
| | - María José Lista
- Department of Infectious Diseases, King's College London, London, United Kingdom
| | - Clement Bouton
- Department of Infectious Diseases, King's College London, London, United Kingdom
| | - Simone Kunzelmann
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Harry Wilson
- Department of Infectious Diseases, King's College London, London, United Kingdom
| | - Matthew A Cottee
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Andrew G Purkiss
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Elizabeth R Morris
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Stuart J D Neil
- Department of Infectious Diseases, King's College London, London, United Kingdom.
| | - Ian A Taylor
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, United Kingdom.
| | - Chad M Swanson
- Department of Infectious Diseases, King's College London, London, United Kingdom.
| |
Collapse
|
164
|
Akanmori NN, Junop MS, Gupta RS, Park J. Conformational flexibility of human ribokinase captured in seven crystal structures. Int J Biol Macromol 2025; 299:140109. [PMID: 39837438 DOI: 10.1016/j.ijbiomac.2025.140109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
d-ribose is a critical sugar substrate involved in the biosynthesis of nucleotides, amino acids, and cofactors, with its phosphorylation to ribose-5-phosphate by ribokinase (RK) constituting the initial step in its metabolism. RK is conserved across all domains of life, and its activity is significantly enhanced by monovalent metal (M+) ions, particularly K+, although the precise mechanism of this activation remains unclear. In this study, we present several crystal structures of human RK in both unliganded and substrate-bound states, offering detailed insights into its substrate binding process, reaction mechanism, and conformational changes throughout the catalytic cycle. Notably, bound ATP exhibited significant conformational flexibility in its triphosphate moiety, a feature shared with other RK homologues, suggesting that achieving a catalytically productive triphosphate configuration plays a key role in regulating enzyme activity. We also identified a unique conformational change in the M+ ion binding loop of human RK, specifically the flipping of the Gly306-Thr307 peptide plane, likely influenced by the ionic radius of the bound ion. These findings provide new insights into the RK reaction mechanism and its activation by M+ ions, paving the way for future investigations into the allosteric regulation of human RK and related sugar kinase enzymes.
Collapse
Affiliation(s)
- Naomi N Akanmori
- Department of Biochemistry, Memorial University of Newfoundland, 45 Arctic Avenue, St. John's, Newfoundland and Labrador, Canada
| | - Murray S Junop
- Department of Biochemistry, Western University, 1151 Richmond Street, London, Ontario, Canada
| | - Radhey S Gupta
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, Canada
| | - Jaeok Park
- Department of Biochemistry, Memorial University of Newfoundland, 45 Arctic Avenue, St. John's, Newfoundland and Labrador, Canada.
| |
Collapse
|
165
|
Qu Y, Tai W, Ma E, Jiang Q, Fan M, Xiao W, Tian C, Liu Y, Liu J, Wang X, Ge J, Cheng G. Generation and characterization of neutralizing antibodies against M1R and B6R proteins of monkeypox virus. Nat Commun 2025; 16:3100. [PMID: 40164599 PMCID: PMC11958656 DOI: 10.1038/s41467-025-58180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
The global outbreak of monkeypox virus (MPXV), combined with the termination of smallpox vaccination and the lack of specific antiviral treatments, raises increasing concerns. The surface proteins M1R and B6R of MPXV are crucial for virus transmission and serve as key targets for vaccine development. In this study, a panel of human antibodies targeting M1R and B6R is isolated from a human antibody library using phage display technology. Among these antibodies, A138 against M1R and B026 against B6R show the most potent broad-spectrum neutralizing activities against MPXV and Vaccinia virus (VACV). When used in combination, A138 and B026 exhibit complementary neutralizing activity against both viruses in vitro. X-ray crystallography reveales that A138 binds to the loop regions of M1R, similar to the vulnerable epitope of 7D11 on VACV L1R. By contrast, A129 targets a more cryptic epitope, primarily comprising the β-strands of M1R. Moreover, prophylactic and therapeutic administration of A138 or B026 alone provides partial protection, while combining these two antibodies results in enhanced protection against VACV in male C57BL/6 mice. This study demonstrates of a dual-targeting strategy using two different components of the virion for the prevention and treatment of MPXV infection.
Collapse
Grants
- National Key Research and Development Plan of China (2021YFC2302405 to G.C.), Shenzhen Medical Research Fund (B2404002 to G.C.), the National Natural Science Foundation of China (32188101 to G.C.), the Shenzhen San-Ming Project for Prevention and Research on Vector-borne Diseases (SZSM202211023 to G.C.), the Yunnan Provincial Science and Technology Project at Southwest United Graduate School (202302AO370010 to G.C.), the New Cornerstone Science Foundation through the New Cornerstone Investigator Program, and the XPLORER PRIZE.
- Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2023-RC180-01)
- Shenzhen Medical Research Fund (E24010010, E24010014 to W.T.,), the National Natural Science Foundation of China (82271872, 82341046, 32100755 to W.T.)
- Shenzhen Medical Research Fund (B2301009, E24010012 to Y.L.), the National Natural Science Foundation of China (82241082, 32270182 to Y.L.;)
- National Natural Science Foundation of China (National Science Foundation of China)82372254
Collapse
Affiliation(s)
- Yuanyuan Qu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Enhao Ma
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China
- Southwest United Graduate School, Kunming, 650092, China
| | - Qiwei Jiang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, State Key Laboratory of Pathogen and Biosecurity, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun, China
| | - Miao Fan
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 102629, China
| | - Wangcheng Xiao
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Yang Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Jianying Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, China
| | - Xinquan Wang
- The Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Collaborative Innovation Center for Biotherapy, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jiwan Ge
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 102629, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, China.
- Southwest United Graduate School, Kunming, 650092, China.
| |
Collapse
|
166
|
Becht DC, Selvam K, Lachance C, Côté V, Li K, Nguyen MC, Pareek A, Shi X, Wen H, Blanco MA, Côté J, Kutateladze TG. A multivalent engagement of ENL with MOZ. Nat Struct Mol Biol 2025; 32:709-718. [PMID: 39794553 DOI: 10.1038/s41594-024-01455-8] [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: 02/26/2024] [Accepted: 11/19/2024] [Indexed: 01/13/2025]
Abstract
The epigenetic cofactor ENL (eleven-nineteen-leukemia) and the acetyltransferase MOZ (monocytic leukemia zinc finger) have vital roles in transcriptional regulation and are implicated in aggressive forms of leukemia. Here, we describe the mechanistic basis for the intertwined association of ENL and MOZ. Genomic analysis shows that ENL and MOZ co-occupy active promoters and that MOZ recruits ENL to its gene targets. Structural studies reveal a multivalent assembly of ENL at the intrinsically disordered region (IDR) of MOZ. While the extraterminal (ET) domain of ENL recognizes the canonical ET-binding motif in IDR, the YEATS domains of ENL and homologous AF9 bind to a set of acetylation sites in the MOZ IDR that are generated by the acetyltransferase CBP (CREB-binding protein). Our findings suggest a multifaceted acetylation-dependent and independent coupling of ENL, MOZ and CBP/p300, which may contribute to leukemogenic activities of the ENL-MOZ assembly and chromosomal translocations of ENL, MOZ and CBP/p300.
Collapse
Affiliation(s)
- Dustin C Becht
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Karthik Selvam
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Catherine Lachance
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Valérie Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Kuai Li
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Minh Chau Nguyen
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Akshay Pareek
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Hong Wen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - M Andres Blanco
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, USA
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of CHU de Québec-Université Laval Research, Laval University Cancer Research Center, Québec City, Québec, Canada
| | - Tatiana G Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
167
|
Ho NAT, Given FM, Stanborough T, Klein M, Allison TM, Bulloch EMM, Jiao W, Johnston JM. Apparent Reversal of Allosteric Response in Mycobacterium tuberculosis MenD Reveals Links to Half-of-Sites Reactivity. Chembiochem 2025; 26:e202400943. [PMID: 39945237 DOI: 10.1002/cbic.202400943] [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: 11/19/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
Redox-active molecules play critical roles in various biological functions, including cellular respiration. In bacterial electron transport chains, menaquinones serve as key electron carriers. The first committed enzyme in the menaquinone biosynthesis pathway of Mycobacterium tuberculosis (Mtb), MenD, is allosterically inhibited by 1,4-dihydroxy-2-naphthoic acid (DHNA), the first redox-active metabolite in the pathway. Structural asymmetries in Mtb-MenD suggest that this enzyme operates via a half-of-sites mechanism for catalysis. Here, we investigate the interplay between its catalytic and allosteric mechanisms. Using molecular dynamics (MD) simulations, mutagenesis, kinetic and binding assays, and structural analyses, we identified and characterised mutants of two residues, D141 and D306, involved in stabilising asymmetric conformations associated with allostery. These mutations had complex effects on Mtb-MenD's reaction kinetics, with the D306 mutants showing an apparent reversal of the allosteric response to DHNA. Our findings indicate that asymmetric active site conformations may facilitate optimal binding of cofactors and substrates, while the transition between alternating active site conformations is essential for the catalytic cycle. DHNA binding stabilises asymmetry in the tetramer, likely promoting the binding of cofactors, substrates, or reaction intermediates. However, DHNA interferes with the transition between alternating conformations, ultimately impairing turnover and catalytic cycling in Mtb-MenD.
Collapse
Affiliation(s)
- Ngoc Anh Thu Ho
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Fiona M Given
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Tamsyn Stanborough
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
| | - Michelle Klein
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Timothy M Allison
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Esther M M Bulloch
- School of Biological Sciences, University of Auckland, Private Bag, 92019, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Wanting Jiao
- Ferrier Research Institute, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| | - Jodie M Johnston
- Biomolecular Interaction Centre and School of Physical and Chemical Sciences, University of Canterbury, Christchurch, 8041, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag, 92019, Auckland, New Zealand
| |
Collapse
|
168
|
Furihata H, Zhu Z, Nishida K, Sakuraba Y, Tsuji A, Yamashita H, Nosaki S, Tachibana R, Yamagami A, Ikeda Y, Abe M, Sawasaki T, Nakano T, Yanagisawa S, Tanokura M, Miyakawa T. Structural insights into CDF1 accumulation on the CONSTANS promoter via a plant-specific DNA-binding domain. NATURE PLANTS 2025; 11:836-848. [PMID: 40263610 DOI: 10.1038/s41477-025-01946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 02/21/2025] [Indexed: 04/24/2025]
Abstract
DNA-binding with one-finger (Dof) proteins are a family of plant-specific transcription factors distinguished by the highly conserved Dof DNA-binding domain. Various members play crucial roles in diverse plant biological processes. However, it remains unclear how the Dof domain recognizes a restricted set of promoters for gene regulation by binding to just four nucleotides, AAAG/CTTT. Here we present the crystal structure of the Dof domain of CYCLING DOF FACTOR 1 (CDF1), a well-characterized Dof protein acting as a transcriptional repressor by binding to the CONSTANS promoter to regulate photoperiodic flowering, in complex with DNA containing two cis elements. The data reveal that the Dof domain exhibits a unique zinc ribbon fold that includes a three-stranded antiparallel β-sheet and a carboxy-terminal loop, enabling DNA recognition accompanied by directional expansion of the major groove. These features facilitate binding to contiguous target cis elements in a proper arrangement to effectively regulate gene expression.
Collapse
Affiliation(s)
- Hirotake Furihata
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Zhangliang Zhu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kaisei Nishida
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yasuhito Sakuraba
- Agro-Biotechnology Research Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Akihiro Tsuji
- Graduate School of Engineering Science, The University of Osaka, Osaka, Japan
| | - Hayato Yamashita
- Graduate School of Engineering Science, The University of Osaka, Osaka, Japan
| | - Shohei Nosaki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Ryo Tachibana
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Ayumi Yamagami
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Yoshiki Ikeda
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| | - Masayuki Abe
- Graduate School of Engineering Science, The University of Osaka, Osaka, Japan
| | | | - Takeshi Nakano
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Shuichi Yanagisawa
- Agro-Biotechnology Research Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
169
|
Shen YH, Cheng WL, Wang X, Dai HE, Wang M, Liu L. Crystal Structure of a Thioredoxin-like Ferredoxin Encoded Within a Cobalamin Biosynthetic Operon of Rhodobacter capsulatus. Protein J 2025; 44:192-200. [PMID: 39924633 DOI: 10.1007/s10930-025-10254-z] [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: 02/01/2025] [Indexed: 02/11/2025]
Abstract
Thioredoxin-like ferredoxin is a small homodimeric protein containing a [2Fe-2S] cluster in each monomer. It is only found in bacteria but its physiological function remains largely unknown. The cobalamin biosynthetic operon in the genome of the purple phototroph Rhodobacter capsulatus encodes a putative ferredoxin dubbed as CfrX. To characterize this protein, we cloned, expressed, purified, and crystalized the recombinant CfrX in the iron-sulfur cluster-bound state, and solved the structure at 2.1-Å resolution. Adopting a typical thioredoxin-like ferredoxin fold, a CfrX monomer binds one [2Fe-2S] cluster through four Cys residues located on two protruding loops. Unexpectedly, CfrX dimerizes in a previously unreported manner. With the structural information, we ascertained CfrX as a thioredoxin-like ferredoxin. While the precise function of CfrX in cobalamin biosynthesis is elusive, a link between CfrX and aerobic cobaltochelatase should exist due to the gene clustering pattern. We also discussed the possible relationship among CfrX, CobW, and CobNST with respect to the [2Fe-2S] cluster.
Collapse
Affiliation(s)
- Yan-Hao Shen
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Wen-Long Cheng
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Xiao Wang
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Huai-En Dai
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Mingzhu Wang
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Lin Liu
- School of Life Sciences, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China.
| |
Collapse
|
170
|
Qiu W, Trinh TKH, Catalano C, Mehta A, Desai UR, Kellogg GE, Hendrickson WA, Guo Y. Cholesterol-dependent enzyme activity of human TSPO1. Proc Natl Acad Sci U S A 2025; 122:e2323045122. [PMID: 40146857 PMCID: PMC12002307 DOI: 10.1073/pnas.2323045122] [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: 12/31/2023] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
The amino acid sequence of the tryptophan-rich sensory proteins (TSPO) is substantially conserved throughout all kingdoms of life. Human mitochondrial TSPO1 (HsTSPO1) binds to porphyrins and steroids, although its interactions with these molecules remains unknown. HsTSPO1 is associated with numerous physiological and pathological disorders, but the underlying molecular mechanisms are unknown. Here, we disclose the finding of human mitochondrial TSPO as a cholesterol-dependent protoporphyrin IX oxygenase. The results of our biochemical characterization are consistent with structural data and evolutionary analysis. The dependence of HsTSPO1 activity on cholesterol may be the result of the coevolution of this membrane protein with the membrane system. Our study provides a molecular foundation for comprehending the various roles played by mitochondrial TSPO in normal physiological and pathological situations.
Collapse
Affiliation(s)
- Weihua Qiu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Thi Kim Hoang Trinh
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Claudio Catalano
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Akul Mehta
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Umesh R. Desai
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Glen E. Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| | - Wayne A. Hendrickson
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY10032
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY10032
- New York Structural Biology Center, New York, NY10027
| | - Youzhong Guo
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA23298-0540
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA23298-0133
| |
Collapse
|
171
|
Chen J, Chen Y, Huang L, Lin X, Chen H, Xiang W, Liu L. Trans-nuclease activity of Cas9 activated by DNA or RNA target binding. Nat Biotechnol 2025; 43:558-568. [PMID: 38811761 DOI: 10.1038/s41587-024-02255-7] [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: 09/18/2023] [Accepted: 04/18/2024] [Indexed: 05/31/2024]
Abstract
Type V and type VI CRISPR-Cas systems have been shown to cleave nonspecific single-stranded DNA (ssDNA) or single-stranded RNA (ssRNA) in trans, but this has not been observed in type II CRISPR-Cas systems using single guide RNA. We show here that the type II CRISPR-Cas9 systems directed by CRISPR RNA and trans-activating CRISPR RNA dual RNAs show RuvC domain-dependent trans-cleavage activity for both ssDNA and ssRNA substrates. Cas9 possesses sequence preferences for trans-cleavage substrates, preferring to cleave T- or C-rich ssDNA substrates. We find that the trans-cleavage activity of Cas9 can be activated by target ssDNA, double-stranded DNA and ssRNA. The crystal structure of Cas9 in complex with guide RNA and target RNA provides a structural basis for the binding of target RNA to activate Cas9. Based on the trans-cleavage activity of Cas9 and nucleic acid amplification technology, we develop the nucleic acid detection platforms DNA-activated Cas9 detection and RNA-activated Cas9 detection, which are capable of detecting DNA and RNA samples with high sensitivity and specificity.
Collapse
Affiliation(s)
- Jiyun Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Ying Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Linglong Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xiaofeng Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Hong Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Wenwen Xiang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Liang Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
172
|
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
|
173
|
Zheng S, Shi X, Lin J, Yang Y, Xin Y, Bai X, Zhu H, Chen H, Wu J, Zheng X, Lin L, Huang Z, Yang S, Hu F, Liu W. Structural basis for the fast maturation of pcStar, a photoconvertible fluorescent protein. Acta Crystallogr D Struct Biol 2025; 81:181-195. [PMID: 40094266 DOI: 10.1107/s2059798325002141] [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: 11/11/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Green-to-red photoconvertible fluorescent proteins (PCFPs) serve as key players in single-molecule localization super-resolution imaging. As an early engineered variant, mEos3.2 has limited applications, mostly due to its slow maturation rate. The recent advent of a novel variant, pcStar, obtained by the simple mutation of only three amino acids (D28E/L93M/N166G) in mEos3.2, exhibits significantly accelerated maturation and enhanced fluorescent brightness. This improvement represents an important advance in the field of biofluorescence by enabling early detection with reliable signals, essential for labelling dynamic biological processes. However, the mechanism underlying the significant improvement in fluorescent performance from mEos3.2 to pcStar remains elusive, preventing the rational design of more robust variants through mutagenesis. In this study, we determined the crystal structures of mEos3.2 and pcStar in their green states at atomic resolution and performed molecular-dynamics simulations to reveal significant divergences between the two proteins. Our structural and computational analyses revealed crucial features that are distinctively present in pcStar, including the presence of an extra solvent molecule, high conformational stability and enhanced interactions of the chromophore with its surroundings, tighter tertiary-structure packing and dynamic central-helical deformation. Resulting from the triple mutations, all of these structural features are likely to establish a mechanistic link to the greatly improved fluorescent performance of pcStar. The data described here not only provide a good example illustrating how distant amino-acid substitutions can affect the structure and bioactivity of a protein, but also give rise to strategic considerations for the future engineering of more widely applicable PCFPs.
Collapse
Affiliation(s)
- Shuping Zheng
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Xiangrui Shi
- Institute of Immunology, PLA, Army Medical University, Chongqing, People's Republic of China
| | - Junjin Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou, People's Republic of China
| | - Yiwei Yang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, People's Republic of China
| | - Yiting Xin
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Xinru Bai
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Huachen Zhu
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Hui Chen
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jiasen Wu
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Xiaowei Zheng
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ling Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou, People's Republic of China
| | - Zhihong Huang
- Public Technology Service Center, Fujian Medical University, Fuzhou, People's Republic of China
| | - Sheng Yang
- Fujian Key Laboratory of Toxicant and Drug Toxicology, Medical College, Ningde Normal University, Ningde, People's Republic of China
| | - Fen Hu
- Key Laboratory of Gastrointestinal Cancer, Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People's Republic of China
| | - Wei Liu
- Institute of Immunology, PLA, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
174
|
Wijaya AJ, Dolan SK, Kohlstedt M, Gläser L, Brear P, Geddis S, Wittmann C, Spring DR, Welch M. The 2-methylcitrate cycle and the glyoxylate shunt in Pseudomonas aeruginosa are linked through enzymatic redundancy. J Biol Chem 2025; 301:108355. [PMID: 40015638 PMCID: PMC11982470 DOI: 10.1016/j.jbc.2025.108355] [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: 12/20/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
The 2-methylcitrate cycle and the glyoxylate cycle are central metabolic pathways in Pseudomonas aeruginosa, enabling the organism to utilize organic acids such as propionate and acetate during infection. Here, we show that these cycles are linked through enzymatic redundancy, with isocitrate lyase (AceA) exhibiting secondary 2-methylisocitrate lyase activity. Furthermore, we use a combination of structural analyses, enzyme kinetics, metabolomics, and targeted mutation of PrpBPa to demonstrate that whereas loss of PrpB function impairs growth on propionate, the promiscuous 2-methylisocitrate lyase activity of AceA compensates for this by mitigating the accumulation of toxic 2-methylcitrate cycle intermediates. Our findings suggest that simultaneous inhibition of PrpB and AceA could present a robust antimicrobial strategy to target P. aeruginosa in propionate-rich environments, such as the cystic fibrosis airways. Our results emphasize the importance of understanding pathway interconnections in the development of novel antimicrobial agents.
Collapse
Affiliation(s)
- Andre J Wijaya
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Stephen K Dolan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom; Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, USA.
| | - Michael Kohlstedt
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - Lars Gläser
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Geddis
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Wittmann
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - David R Spring
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
175
|
Jäger M, Mortenson DE, Ardejani MS, Kline GM, Dendle MT, Yan NL, Powers ET, Gruebele M, Kelly JW. Lysine carbamoylation during urea denaturation remodels the energy landscape of human transthyretin dissociation linked to unfolding. Protein Sci 2025; 34:e70009. [PMID: 40130761 PMCID: PMC11934213 DOI: 10.1002/pro.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 03/26/2025]
Abstract
Chemical denaturants such as urea have become indispensable in modern protein science for measuring the energetics of protein folding and assembly. Denaturants bind to and preferentially stabilize denatured states, folding transition states, and folding intermediates over the native state, allowing experimental access to free energies of folding and insights into folding mechanisms. However, too little attention is paid to the established chemical instability of aqueous urea, that is, its decomposition into the reactive electrophile ammonium cyanate or isocyanic acid depending on the solution pH. Protein carbamoylation by cyanate/isocyanic acid can change the dissociation and/or unfolding free energy landscape of the protein under study with time. This problem is exemplified using the human blood protein transthyretin (TTR), a kinetically stable transporter of thyroid hormone and holo-retinol binding protein. The dissociation, misfolding, and aggregation of TTR are associated with a prominent human amyloid disease. We demonstrate that modification of TTR by cyanate reshapes the energy landscape of TTR tetramer dissociation and unfolding on multiple time scales. Like certain halide anions and the more chemically inert thiocyanate anion, cyanate binds weakly and non-covalently to the thyroid hormone binding interface in the TTR tetramer. The close proximity of the bound cyanate ion to the pKa-perturbed lysine 15 ε-amino side chain nucleophile in the thyroid hormone binding sites of TTR favors carbamoylation of this nitrogen. Lysine 15 ε-amino carbamoylation substantially slows down TTR tetramer dissociation mediated by urea denaturation, thus introducing kinetic heterogeneity early in the unfolding reaction. Slower carbamoylation of the subpopulation of other, less pKa-perturbed lysine ε-amino groups hastens tetramer unfolding, leading to non-exponential, sigmoidal unfolding trajectories. We thus demonstrate that lysine carbamoylation in urea solutions can strongly alter protein unfolding energetics and the mechanism of unfolding.
Collapse
Affiliation(s)
- Marcus Jäger
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| | | | | | - Gabriel M. Kline
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| | - Maria T. Dendle
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| | - Nicholas L. Yan
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| | - Evan T. Powers
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| | - Martin Gruebele
- Departments of Chemistry and Physics, Center for Biophysics and Quantitative Biology, and Carle‐Illinois College of MedicineUniversity of Illinois Urbana‐ChampaignChampaignIllinoisUSA
| | - Jeffery W. Kelly
- Department of Chemistry and the Skaggs Institute of Chemical BiologyScripps ResearchLa JollaCaliforniaUSA
| |
Collapse
|
176
|
Davies AM, Trentham D, Sutton BJ, Brown PR. Structure and activity of a phosphinothricin N-acetyltransferase (PSPTO_3321) from Pseudomonas syringae pv. tomato DC3000. Biochem Biophys Res Commun 2025; 755:151539. [PMID: 40054337 DOI: 10.1016/j.bbrc.2025.151539] [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: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Phosphinothricin inhibits plant glutamine synthetase and is used as a herbicide. Streptomyces hygroscopicus and Streptomyces viridochromogenes, which produce phosphinothricin naturally, encode acetyltransferases that confer phosphinothricin resistance. In the Pseudomonas genome database, a number of proteins have been annotated as phosphinothricin acetyltransferases and putative phosphinothricin acetyltransferases. One such protein is PSPTO_3321 from P. syringae, a strain that causes tomato speck. Here, we reveal that PSPTO_3321 from P. syringae, termed syr_pat, is a phosphinothricin acetyltransferase, and also retains a lower level of activity against the structurally similar substrate methionine sulfoximine. We solved a 1.6 Å resolution crystal structure of syr_pat alone and a 2.5 Å resolution structure for a complex with L-phosphinothricin. We also characterised active site mutants, providing insights into substrate specificity. Our work now provides a basis for further study of the reaction mechanism.
Collapse
Affiliation(s)
- Anna M Davies
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, London, SE1 1UL, United Kingdom
| | - David Trentham
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, London, SE1 1UL, United Kingdom
| | - Brian J Sutton
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, London, SE1 1UL, United Kingdom
| | - Paul R Brown
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, London, SE1 1UL, United Kingdom.
| |
Collapse
|
177
|
Fang B, Bai H, Zhang J, Wang L, Li P, Ge Y, Yang H, Wang H, Peng B, Hu W, Ma H, Chen X, Fu L, Li L. Albumins constrainting the conformation of mitochondria-targeted photosensitizers for tumor-specific photodynamic therapy. Biomaterials 2025; 315:122914. [PMID: 39461059 DOI: 10.1016/j.biomaterials.2024.122914] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Tumor ablation Preclinical organelle-targeted phototherapies have effectively achieved tumor photoablation for regenerative biomedical applications in cancer therapies. However, engineering effective phototherapy drugs with precise tumor-localization targeting and organelle direction remains challenging. Herein, we report a albumins constrainting mitochondrial-targeted photosensitizer nanoparticles (PSs@BSAs) for tumor-specific photodynamic therapy. X-ray crystallography elucidates the two-stage assembly mechanism of PSs@BSAs. Femtosecond transient absorption spectroscopy and quantum mechanical calculations reveal the implications of conformational dynamics at the excited state. PSs@BSAs can efficiently disable mitochondrial activity, and further disrupt tumor angiogenesis based on the photodynamic effect. This triggers a metabolic and oxidative stress crisis to facilitate photoablation of solid tumor and antitumor metastasis. The study fully elucidates the interdisciplinary issues of chemistry, physics, and biological interfaces, thereby opening new horizons to inspire the engineering of organelle-targeted tumor-specific photosensitizers for biomedical applications.
Collapse
Affiliation(s)
- Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China; State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Limin Wang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - PanPan Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yihao Ge
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Hui Yang
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Hui Wang
- Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361102, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Wenbo Hu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Huili Ma
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xi Chen
- Key Laboratory of Synthetic and Natural Functional Molecule of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China.
| | - Li Fu
- State Key Laboratory of Solidification Processing, School of Materials Science and Engineering, Northwestern Polytechnical University, Xi'an, 710072, China.
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, China; Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361102, China; Future Display Institute in Xiamen, Xiamen, 361005, China.
| |
Collapse
|
178
|
Nguyen CL, Tramell AR, Norman JO, Abendroth J, Barrett KF, Craig JK, Edwards TE, Lorimer DD, Van Voorhis WC, McLaughlin KJ. Structural characterization of dUTPase from Legionella pneumophila. Acta Crystallogr F Struct Biol Commun 2025; 81:155-162. [PMID: 40091853 PMCID: PMC11970128 DOI: 10.1107/s2053230x25001815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Cellular deoxyuridine 5'-triphosphate nucleotidohydrolases (dUTPases) catalyze the hydrolysis of deoxyuridine triphosphate (dUTP) to deoxyuridine monophosphate (dUMP) and pyrophosphate (PPi). dUTPase is an essential metabolic enzyme which maintains the homeostatic dTTP:dUTP ratio. As DNA polymerases are unable to distinguish between thymine and uracil during replication, the dTTP:dUTP ratio is essential for preventing the misincorporation of uracil into DNA. In the absence of dUTPase regulation of the dTTP:dUTP ratio, many DNA double-strand breaks are induced by DNA-repair enzymes, which may ultimately lead to cell death. Legionnaires' disease is a rare but severe respiratory infection caused primarily by Legionella pneumophila serogroup 1. Increased characterization of the L. pneumophila proteome is of interest for the development of new treatments. Many DNA metabolism proteins have yet to be characterized in L. pneumophila, including dUTPase. Here, we present analysis of two crystal structures of L. pneumophila dUTPase in its apo and dUMP-bound states, determined to 1.80 and 1.95 Å resolution, respectively. The structures were solved by the Seattle Structural Genomics Center for Infectious Disease (SSGCID) as part of their mission to determine structures of proteins and other molecules with an important biological role in human pathogens.
Collapse
Affiliation(s)
- Chi L. Nguyen
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Abigail R. Tramell
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Jordan O. Norman
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Jan Abendroth
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Beryllium Discovery Corporation, 7869 Day Road West, Bainbridge Island, WA98110, USA
| | - Kayleigh F. Barrett
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious DiseasesUniversity of Washington School of MedicineSeattleWA98195USA
| | - Justin K. Craig
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious DiseasesUniversity of Washington School of MedicineSeattleWA98195USA
| | - Thomas E. Edwards
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Beryllium Discovery Corporation, 7869 Day Road West, Bainbridge Island, WA98110, USA
| | - Donald D. Lorimer
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Beryllium Discovery Corporation, 7869 Day Road West, Bainbridge Island, WA98110, USA
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious DiseasesUniversity of Washington School of MedicineSeattleWA98195USA
| | - Krystle J. McLaughlin
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
- Department of ChemistryVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| |
Collapse
|
179
|
Li F, Zhang M, Liu C, Cheng J, Yang Y, Peng X, Li Z, Cai W, Yu H, Wu J, Guo Y, Geng H, Fa Y, Zhang Y, Wu D, Yin Y. De novo discovery of a molecular glue-like macrocyclic peptide that induces MCL1 homodimerization. Proc Natl Acad Sci U S A 2025; 122:e2426006122. [PMID: 40131955 PMCID: PMC12002256 DOI: 10.1073/pnas.2426006122] [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: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Macrocyclic peptides have emerged as promising drug candidates, filling the gap between small molecules and large biomolecules in drug discovery. The antiapoptotic protein myeloid cell leukemia 1 (MCL1) is crucial for numerous cancers, yet it presents challenges for selective targeting by traditional inhibitors. In this study, we identified a macrocyclic peptide, 5L1, that strongly binds to MCL1, with a dissociation constant (KD) of 7.1 nM. This peptide shows the potential to specifically inhibit the function of MCL1, and demonstrates effective antitumor activity against several blood tumor cell lines with the half maximal inhibitory concentration (IC50) values for cell-penetrating peptide-conjugated 5L1 in the range of 0.6 to 3 μM. Structural analysis revealed that it functions similarly to molecular glue, capable of binding to two MCL1 molecules simultaneously and inducing their homodimerization. This unique mechanism of action distinguishes it from traditional small-molecule MCL1 inhibitors, underscoring the potential of macrocyclic peptides functioning as molecular glues. Moreover, it inspires the development of highly selective inhibitors targeting MCL1 and other related targets with this glue-like mechanism.
Collapse
Affiliation(s)
- Fengwei Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao266061, China
| | - Mengmeng Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Chao Liu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Jie Cheng
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Yawen Yang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Xiangda Peng
- Shanghai Zelixir Biotech Company Ltd., Shanghai200030, China
| | - Zhifeng Li
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Wenfeng Cai
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Haipeng Yu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Junjie Wu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Yuyu Guo
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | | | - Yun Fa
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao266061, China
| | - Youming Zhang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Dalei Wu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
| | - Yizhen Yin
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao266237, China
- Shandong Research Institute of Industrial Technology, Jinan250101, China
| |
Collapse
|
180
|
Caspy I, Tang S, Bellini D, Gorrec F. A generic cross-seeding approach to protein crystallization. J Appl Crystallogr 2025; 58:383-391. [PMID: 40170961 PMCID: PMC11957411 DOI: 10.1107/s1600576725000457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/17/2025] [Indexed: 04/03/2025] Open
Abstract
Obtaining diffraction-quality crystals is often the rate-limiting step during structure determination of biological macromolecules by X-ray crystallography. To address this problem, we investigated a cross-seeding approach with a mixture integrating a heterogeneous set of protein crystal fragments to be used as generic seeds. The fragments are nanometre-sized templates chosen to promote crystal nucleation of protein samples unrelated to the proteins forming the seeds. An atypical crystal form of the human serine hydrolase retinoblastoma binding protein 9 was obtained by adding the mixture to the protein sample before performing standard crystallization assays. The structure was solved by X-ray crystallography at 1.4 Å resolution. Follow-up experiments showed that crystal fragments of α-amylase were critical components in this particular result. The limitations and future applications of our experimental developments are discussed.
Collapse
Affiliation(s)
- Ido Caspy
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, CambridgeCB2 0QH, United Kingdom
| | - Shan Tang
- Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001People’s Republic of China
| | - Dom Bellini
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, CambridgeCB2 0QH, United Kingdom
| | - Fabrice Gorrec
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, CambridgeCB2 0QH, United Kingdom
| |
Collapse
|
181
|
Doherty EE, Adler BA, Yoon PH, Hsieh K, Loi K, Armbuster EG, Lahiri A, Bolling CS, Wilcox XE, Akkati A, Iavarone AT, Pogliano J, Doudna JA. A miniature CRISPR-Cas10 enzyme confers immunity by an inverse signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.646030. [PMID: 40196485 PMCID: PMC11974785 DOI: 10.1101/2025.03.28.646030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Microbial and viral co-evolution has created immunity mechanisms involving oligonucleotide signaling that share mechanistic features with human anti-viral systems 1 . In these pathways, including CBASS and type III CRISPR systems in bacteria and cGAS-STING in humans, oligonucleotide synthesis occurs upon detection of virus or foreign genetic material in the cell, triggering the antiviral response 2-4 . In a surprising inversion of this process, we show here that the CRISPR-related enzyme mCpol synthesizes cyclic oligonucleotides constitutively as part of an active mechanism that maintains cell health. Cell-based experiments demonstrated that the absence or loss of mCpol-produced cyclic oligonucleotides triggers cell death, preventing spread of viruses that attempt immune evasion by depleting host cyclic nucleotides. Structural and mechanistic investigation revealed mCpol to be a di-adenylate cyclase whose product, c-di-AMP, prevents toxic oligomerization of the effector protein 2TMβ. Analysis of cells by fluorescence microscopy showed that lack of mCpol allows 2TMβ-mediated cell death due to inner membrane collapse. These findings unveil a powerful new defense strategy against virus-mediated immune suppression, expanding our understanding of oligonucleotides in cell health and disease. These results raise the possibility of similar protective roles for cyclic oligonucleotides in other organisms including humans.
Collapse
|
182
|
Nesa ML, Mandal SK, Toelzer C, Humer D, Moody PCE, Berger I, Spadiut O, Raven EL. Crystal structure of ferric recombinant horseradish peroxidase. J Biol Inorg Chem 2025; 30:221-227. [PMID: 40053124 PMCID: PMC11965164 DOI: 10.1007/s00775-025-02103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/04/2025] [Indexed: 04/03/2025]
Abstract
Horseradish peroxidase (HRP), isolated from horseradish roots, is heavily glycosylated, making it difficult to crystallize. In this work, we produced recombinant HRP in E. coli and obtained an X-ray structure of the ferric enzyme at 1.63 Å resolution. The structure shows that the recombinant HRP contains four disulphide bonds and two calcium ions, which are highly conserved in class III peroxidase enzymes. The heme active site contains histidine residues at the proximal (His 170) and distal (His 42) positions, and an active site arginine (Arg 38). Surprisingly, an ethylene glycol molecule was identified in the active site, forming hydrogen bonds with His 42 and Arg 38 at the δ-heme edge. The high yields obtained from the recombinant expression system, and the successful crystallization of the enzyme pave the way for new structural studies in the future.
Collapse
Affiliation(s)
| | | | | | - Diana Humer
- Institute of Chemical, Environmental and Bioscience Engineering, Research Division Biochemical Engineering, TU Wien, Vienna, Austria
| | - Peter C E Moody
- Leicester Institute for Structural & Chemical Biology, Department Molecular & Cell Biology, Henry Wellcome Building, University of Leicester, Leicester, LE1 9HN, UK
| | - Imre Berger
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Oliver Spadiut
- Institute of Chemical, Environmental and Bioscience Engineering, Research Division Biochemical Engineering, TU Wien, Vienna, Austria
| | - Emma L Raven
- School of Chemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
183
|
Pederick JL, Kumar A, Pukala TL, Bruning JB. Functional and structural characterization of Staphylococcus aureus N-acetylglucosamine 1-phosphate uridyltransferase (GlmU) reveals a redox-sensitive acetyltransferase activity. Protein Sci 2025; 34:e70111. [PMID: 40143772 PMCID: PMC11947611 DOI: 10.1002/pro.70111] [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: 12/02/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025]
Abstract
The bifunctional enzyme N-acetylglucosamine 1-phosphate uridyltransferase (GlmU) is a promising antibiotic drug target, as it facilitates the biosynthesis of uridine 5'-diphospho-N-acetylglucosamine, an essential precursor of cell wall constituents. We identified that Staphylococcus aureus GlmU (SaGlmU), which was previously targeted for inhibitor development, possesses a dual-cysteine variation (C379/C404) within the acetyltransferase active site. Enzyme assays performed under reducing and non-reducing conditions revealed that the acetyltransferase activity of SaGlmU is redox-sensitive, displaying ~15-fold lower turnover and ~3-fold higher KM value for the acetyl CoA substrate under non-reducing conditions. This sensitivity was absent in a C379A SaGlmU mutant. Analysis of SaGlmU by mass spectrometry, x-ray crystallography, and in silico modeling support that C379 and C404 act as a reversible, redox-sensitive switch by forming a disulfide under non-reducing conditions that impedes acetyl CoA recognition and turnover. Therefore, we recommend that future in vitro screening and characterization of SaGlmU inhibitors consider both reducing and non-reducing conditions.
Collapse
Affiliation(s)
- Jordan L Pederick
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Akhil Kumar
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Tara L Pukala
- Department of Chemistry, School of Physical Sciences, North Terrace Campus, The University of Adelaide, Adelaide, South Australia, Australia
| | - John B Bruning
- Institute for Photonics and Advanced Sensing (IPAS), School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
184
|
Torres Robles J, Stiegler AL, Boggon TJ, Turk BE. Cancer hotspot mutations rewire ERK2 specificity by selective exclusion of docking interactions. J Biol Chem 2025; 301:108348. [PMID: 40015635 PMCID: PMC11982978 DOI: 10.1016/j.jbc.2025.108348] [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: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
The protein kinase ERK2 is recurrently mutated in human squamous cell carcinomas and other tumors. ERK2 mutations cluster in an essential docking recruitment site that interacts with short linear motifs found within intrinsically disordered regions of ERK substrates and regulators. Cancer-associated mutations do not disrupt ERK2 docking interactions altogether but selectively inhibit some interactions while sparing others. However, the full scope of disrupted or maintained interactions remains unknown, limiting our understanding of how these mutations contribute to cancer. We recently defined the docking interactome of wild-type ERK2 by screening a yeast two-hybrid library of proteomic short linear motifs. Here, we apply this approach to the two most recurrent cancer-associated mutants. We find that most sequences binding to WT ERK2 also interact with both mutant forms. Analysis of differentially interacting sequences revealed that ERK2 mutants selectively lose the ability to bind sequences conforming to a specific motif. We solved the co-crystal structure of ERK2 in complex with a peptide fragment of ISG20, a screening hit that binds exclusively to the WT kinase. This structure demonstrated the mechanism by which cancer hotspot mutations at Glu81, Arg135, Asp321, and Glu322 selectively impact peptide binding. Finally, we found that cancer-associated ERK2 mutations had decreased activity in phosphorylating GEF-H1/ARHGEF2, a known ERK substrate harboring a WT-selective docking motif. Collectively, our studies provide a structural rationale for how a broad set of interactions are disrupted by ERK2 hotspot mutations, suggesting mechanisms for pathway rewiring in cancers harboring these mutations.
Collapse
Affiliation(s)
- Jaylissa Torres Robles
- Department of Chemistry, Yale University, New Haven, Connecticut, USA; Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Amy L Stiegler
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Titus J Boggon
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Benjamin E Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
185
|
Earl CP, Cobbaut M, Barros-Carvalho A, Ivanova ME, Briggs DC, Morais-de-Sá E, Parker PJ, McDonald NQ. Capture, mutual inhibition and release mechanism for aPKC-Par6 and its multisite polarity substrate Lgl. Nat Struct Mol Biol 2025; 32:729-739. [PMID: 39762628 PMCID: PMC11996676 DOI: 10.1038/s41594-024-01425-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/15/2024] [Indexed: 02/23/2025]
Abstract
The mutually antagonistic relationship of atypical protein kinase C (aPKC) and partitioning-defective protein 6 (Par6) with the substrate lethal (2) giant larvae (Lgl) is essential for regulating polarity across many cell types. Although aPKC-Par6 phosphorylates Lgl at three serine sites to exclude it from the apical domain, aPKC-Par6 and Lgl paradoxically form a stable kinase-substrate complex, with conflicting roles proposed for Par6. We report the structure of human aPKCι-Par6α bound to full-length Llgl1, captured through an aPKCι docking site and a Par6PDZ contact. This complex traps a phospho-S663 Llgl1 intermediate bridging between aPKC and Par6, impeding phosphorylation progression. Thus, aPKCι is effectively inhibited by Llgl1pS663 while Llgl1 is captured by aPKCι-Par6. Mutational disruption of the Lgl-aPKC interaction impedes complex assembly and Lgl phosphorylation, whereas disrupting the Lgl-Par6PDZ contact promotes complex dissociation and Lgl phosphorylation. We demonstrate a Par6PDZ-regulated substrate capture-and-release model requiring binding by active Cdc42 and the apical partner Crumbs to drive complex disassembly. Our results suggest a mechanism for mutual regulation and spatial control of aPKC-Par6 and Lgl activities.
Collapse
Affiliation(s)
- Christopher P Earl
- Signalling and Structural Biology Laboratory, Francis Crick Institute, London, UK
| | - Mathias Cobbaut
- Signalling and Structural Biology Laboratory, Francis Crick Institute, London, UK.
- Protein Phosphorylation Laboratory, Francis Crick Institute, London, UK.
| | - André Barros-Carvalho
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Marina E Ivanova
- Signalling and Structural Biology Laboratory, Francis Crick Institute, London, UK
- Imperial College, London, UK
| | - David C Briggs
- Signalling and Structural Biology Laboratory, Francis Crick Institute, London, UK
| | - Eurico Morais-de-Sá
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Peter J Parker
- Protein Phosphorylation Laboratory, Francis Crick Institute, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, London, UK
| | - Neil Q McDonald
- Signalling and Structural Biology Laboratory, Francis Crick Institute, London, UK.
- Institute of Structural and Molecular Biology, School of Natural Sciences, Birkbeck College, London, UK.
| |
Collapse
|
186
|
Davies AM, Bui TTT, Pacheco‐Gómez R, Vester SK, Beavil AJ, Gould HJ, Sutton BJ, McDonnell JM. The Crystal Structure of Human IgD-Fc Reveals Unexpected Differences With Other Antibody Isotypes. Proteins 2025; 93:786-800. [PMID: 39582378 PMCID: PMC11878202 DOI: 10.1002/prot.26771] [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: 08/05/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
Of the five human antibody isotypes, the function of IgD is the least well-understood, although various studies point to a role for IgD in mucosal immunity. IgD is also the least well structurally characterized isotype. Until recently, when crystal structures were reported for the IgD Fab, the only structural information available was a model for intact IgD based on solution scattering data. We now report the crystal structure of human IgD-Fc solved at 3.0 Å resolution. Although similar in overall architecture to other human isotypes, IgD-Fc displays markedly different orientations of the Cδ3 domains in the Cδ3 domain dimer and the lowest interface area of all the human isotypes. The nature of the residues that form the dimer interface also differs from those conserved in the other isotypes. By contrast, the interface between the Cδ2 and Cδ3 domains in each chain is the largest among the human isotypes. This interface is characterized by two binding pockets, not seen in other isotypes, and points to a potential role for the Cδ2/Cδ3 interface in stabilizing the IgD-Fc homodimer. We investigated the thermal stability of IgD-Fc, alone and in the context of an intact IgD antibody, and found that IgD-Fc unfolds in a single transition. Human IgD-Fc clearly has unique structural features not seen in the other human isotypes, and comparison with other mammalian IgD sequences suggests that these unique features might be widely conserved.
Collapse
Affiliation(s)
- Anna M. Davies
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Tam T. T. Bui
- Centre for Biomolecular SpectroscopyKing's College LondonLondonUK
| | | | - Susan K. Vester
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Andrew J. Beavil
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Hannah J. Gould
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - Brian J. Sutton
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| | - James M. McDonnell
- Randall Centre for Cell and Molecular BiophysicsKing's College LondonLondonUK
| |
Collapse
|
187
|
Zhang J, Zhou Y, Duan Q, Xu X, Wang X, Wang J, Liu L. Characterization of Arabidopsis thaliana FRATAXIN HOMOLOG in heme catabolism. Biochimie 2025; 231:110-115. [PMID: 39722354 DOI: 10.1016/j.biochi.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Frataxin plays vital roles in various iron related processes. Arabidopsis thaliana FRATAXIN HOMOLOG (AtFH) is the first identified plant frataxin and has been found to regulate the last step of heme biosynthesis. Here, we report the involvement of AtFH in heme catabolism by regulating the activity of heme oxygenase. AtFH forms a homodimer, and its crystal structure shows the dimeric interactions. A structural comparison with known frataxin structures suggests the iron binding sites, and the site for heme oxygenase activity is possibly located in a region containing Glu78. The results indicate a previously uncharacterized role of plant frataxin in heme catabolism.
Collapse
Affiliation(s)
- Jing Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Yingying Zhou
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Qianyi Duan
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Xinhe Xu
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Xiao Wang
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China
| | - Jia Wang
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China.
| | - Lin Liu
- School of Life Sciences, Anhui University, Hefei, Anhui, 230601, China.
| |
Collapse
|
188
|
Jangra M, Travin DY, Aleksandrova EV, Kaur M, Darwish L, Koteva K, Klepacki D, Wang W, Tiffany M, Sokaribo A, Coombes BK, Vázquez-Laslop N, Polikanov YS, Mankin AS, Wright GD. A broad-spectrum lasso peptide antibiotic targeting the bacterial ribosome. Nature 2025; 640:1022-1030. [PMID: 40140562 DOI: 10.1038/s41586-025-08723-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/30/2025] [Indexed: 03/28/2025]
Abstract
Lasso peptides (biologically active molecules with a distinct structurally constrained knotted fold) are natural products that belong to the class of ribosomally synthesized and post-translationally modified peptides1-3. Lasso peptides act on several bacterial targets4,5, but none have been reported to inhibit the ribosome, one of the main targets of antibiotics in the bacterial cell6,7. Here we report the identification and characterization of the lasso peptide antibiotic lariocidin and its internally cyclized derivative lariocidin B, produced by Paenibacillus sp. M2, which has broad-spectrum activity against a range of bacterial pathogens. We show that lariocidins inhibit bacterial growth by binding to the ribosome and interfering with protein synthesis. Structural, genetic and biochemical data show that lariocidins bind at a unique site in the small ribosomal subunit, where they interact with the 16S ribosomal RNA and aminoacyl-tRNA, inhibiting translocation and inducing miscoding. Lariocidin is unaffected by common resistance mechanisms, has a low propensity for generating spontaneous resistance, shows no toxicity to human cells, and has potent in vivo activity in a mouse model of Acinetobacter baumannii infection. Our identification of ribosome-targeting lasso peptides uncovers new routes towards the discovery of alternative protein-synthesis inhibitors and offers a novel chemical scaffold for the development of much-needed antibacterial drugs.
Collapse
MESH Headings
- Ribosomes/drug effects
- Ribosomes/metabolism
- Ribosomes/chemistry
- Anti-Bacterial Agents/pharmacology
- Anti-Bacterial Agents/chemistry
- Anti-Bacterial Agents/metabolism
- Animals
- Mice
- Protein Biosynthesis/drug effects
- Humans
- Paenibacillus/metabolism
- Paenibacillus/chemistry
- RNA, Ribosomal, 16S/metabolism
- RNA, Ribosomal, 16S/chemistry
- Peptides, Cyclic/pharmacology
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/metabolism
- Female
- Models, Molecular
- RNA, Transfer, Amino Acyl/metabolism
- RNA, Transfer, Amino Acyl/chemistry
- Bacteria/drug effects
- Bacteria/growth & development
- Peptides/pharmacology
- Peptides/chemistry
- Microbial Sensitivity Tests
Collapse
Affiliation(s)
- Manoj Jangra
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Dmitrii Y Travin
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Elena V Aleksandrova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Manpreet Kaur
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lena Darwish
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kalinka Koteva
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Dorota Klepacki
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Wenliang Wang
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Maya Tiffany
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Akosiererem Sokaribo
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Brian K Coombes
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Nora Vázquez-Laslop
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Yury S Polikanov
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Alexander S Mankin
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Gerard D Wright
- David Braley Centre for Antibiotics Discovery, McMaster University, Hamilton, Ontario, Canada.
- M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
189
|
Nguyen CL, Fan W, Fisher S, Matthews K, Norman JO, Abendroth J, Barrett KF, Craig JK, Edwards TE, Lorimer DD, McLaughlin KJ. Structures of Legionella pneumophila serogroup 1 peptide deformylase bound to nickel(II) and actinonin. Acta Crystallogr F Struct Biol Commun 2025; 81:163-170. [PMID: 40091854 PMCID: PMC11970127 DOI: 10.1107/s2053230x25001876] [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: 12/03/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Legionella pneumophila serogroup 1 is the primary causative agent of Legionnaires' disease, a rare but severe respiratory infection. While the fatality rate of Legionnaires' disease is low in the general population, it is more pronounced in vulnerable communities such as the immunocompromised. Thus, the development of new antimicrobials is of interest for use when existing antibiotics may not be applicable. Peptide deformylases (PDFs) have been under continued investigation as targets for novel antimicrobial compounds. PDF plays an essential role in protein synthesis, removing the N-terminal formyl group from new polypeptides, and is required for growth in most bacteria. Here, we report two crystal structures of L. pneumophila serogroup 1 PDF (LpPDF) bound to either Ni2+, an active state, or inhibited by actinonin and Zn2+; the structures were determined to 1.5 and 1.65 Å resolution, respectively, and were solved by the Seattle Structural Genomics Center for Infectious Disease (SSGCID). The SSGCID is charged with determining structures of biologically important proteins and molecules from human pathogens. As actinonin is an antimicrobial natural product that has been used as a reference compound in drug development, these structures will help support the ongoing drug-development process.
Collapse
Affiliation(s)
- Chi L. Nguyen
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - William Fan
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Sean Fisher
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Krystal Matthews
- Chemistry DepartmentVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Jordan O. Norman
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| | - Jan Abendroth
- UCB Biosciences, 7869 Day Road West, Bainbridge Island, WA98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Kayleigh F. Barrett
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious DiseasesUniversity of Washington School of MedicineSeattleWA98195USA
| | - Justin K. Craig
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
- Center for Emerging and Re-emerging Infectious Diseases (CERID), Department of Medicine, Division of Allergy and Infectious DiseasesUniversity of Washington School of MedicineSeattleWA98195USA
| | - Thomas E. Edwards
- UCB Biosciences, 7869 Day Road West, Bainbridge Island, WA98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Donald D. Lorimer
- UCB Biosciences, 7869 Day Road West, Bainbridge Island, WA98110, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Krystle J. McLaughlin
- Biochemistry ProgramVassar College124 Raymond AvenuePoughkeepsieNY12604USA
- Chemistry DepartmentVassar College124 Raymond AvenuePoughkeepsieNY12604USA
| |
Collapse
|
190
|
Appelt EA, Thoden JB, Gehrke SA, Bachmeier HD, Rayment I, Shusta EV, Holden HM. The High-Resolution Structure of a Variable Lymphocyte Receptor From Petromyzon marinus Capable of Binding to the Brain Extracellular Matrix. Proteins 2025; 93:801-811. [PMID: 39601379 PMCID: PMC11878203 DOI: 10.1002/prot.26768] [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: 09/05/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Variable lymphocyte receptors (VLRs) are antigen receptors derived from the adaptive immune system of jawless vertebrates such as lamprey ( Petromyzon marinus ). First discovered in 2004, VLRs have been the subject of numerous biochemical and structural investigations. Due to their unique antigen binding properties, VLRs have been leveraged as possible drug delivery agents. One such VLR, previously identified and referred to as P1C10, was shown to bind to the brain extracellular matrix. Here, we present the high-resolution X-ray crystal structure of this VLR determined to 1.3 Å resolution. The fold is dominated by a six-stranded mixed β-sheet which provides a concave surface for possible antigen binding. Electron density corresponding to a 4-(2-hydroxyethyl)piperazine-1-propanesulfonic acid buffer molecule (HEPPS) was found in this region. By comparing the P1C10 molecular architecture and its buffer binding residues with those of other VLRs previously reported, it was possible to illustrate how this unique class of proteins can accommodate diverse binding partners. Additionally, we provide an analysis of the experimentally determined structure compared to the models generated by the commonly used AlphaFold and iTASSER structure prediction software packages.
Collapse
Affiliation(s)
- Elizabeth A. Appelt
- Department of Chemical and Biological EngineeringUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - James B. Thoden
- Department of BiochemistryUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Seth A. Gehrke
- Department of Chemical and Biological EngineeringUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Hannah D. Bachmeier
- Department of Chemical and Biological EngineeringUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Ivan Rayment
- Department of BiochemistryUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Eric V. Shusta
- Department of Chemical and Biological EngineeringUniversity of Wisconsin–MadisonMadisonWisconsinUSA
- Department of Neurological SurgeryUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Hazel M. Holden
- Department of BiochemistryUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| |
Collapse
|
191
|
Wang X, Zhou N, Gao XJ, Zhu Z, Sun M, Wang Q, Cao H, Wu X, Zhou C, Zheng Q, Yuan Y, Liu Y, Chen L, Jiang J, Bu P, Gao L. Selective G6PDH inactivation for Helicobacter pylori eradication with transformed polysulfide. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1158-1173. [PMID: 39821832 DOI: 10.1007/s11427-024-2775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/07/2024] [Indexed: 01/19/2025]
Abstract
Alternative treatment for the highly prevalent Helicobacter pylori infection is imperative due to rising antibiotic resistance. We unexpectedly discovered that the anti-H. pylori component in garlic is hydrogen polysulfide (H2Sn, n⩾2), not organic polysulfides. Studies on the mechanism of action (MoA) show that H2Sn specifically inactivates H. pylori glucose-6-phosphate dehydrogenase (G6PDH) by interfering with electron transfer from glucose-6-phosphate (G6P) to nicotinamide adenine dinucleotide phosphate (NADP+). However, low H2Sn yield makes garlic derivatives hard to be a reliable donor of H2Sn to treat H. pylori infection. To address this challenge, we established a polysulfide transformation process from garlic organosulfur compounds into Fe3S4 that generates H2Sn with a 25-58 times increase in yield. Through chitosan encapsulation, we designed a gastric-adaptive H2Sn microreactor (GAPSR) that eradicates H. pylori with 250 times higher efficiency under gastric conditions. A single GAPSR achieves more rapid H. pylori eradication than combined antibiotics therapy without disturbing the gut microbiota. These findings indicate a distinct MoA transformation mediated by polysulfide as an alternative candidate to treat H. pylori infection.
Collapse
Affiliation(s)
- Xiaonan Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ning Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuejiao J Gao
- College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, 330022, China
| | - Zijing Zhu
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Minmin Sun
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian Wang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Haolin Cao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuetong Wu
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Caiyu Zhou
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingkang Zheng
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ye Yuan
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yuan Liu
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Chen
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Jiang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Pengcheng Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- Nanozyme Laboratory in Zhongyuan, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China.
| |
Collapse
|
192
|
Tresco BIC, Wu KJY, Ramkissoon A, Aleksandrova EV, Purdy M, See DNY, Liu RY, Polikanov YS, Myers AG. Discovery of a fluorinated macrobicyclic antibiotic through chemical synthesis. Nat Chem 2025; 17:582-589. [PMID: 40055575 DOI: 10.1038/s41557-025-01738-7] [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: 08/07/2024] [Accepted: 01/14/2025] [Indexed: 04/04/2025]
Abstract
The emergence of bacterial antimicrobial resistance threatens to undermine the utility of antibiotic therapy in medicine. This threat can be addressed, in part, by reinventing existing antibiotic classes using chemical synthesis. Here we present the discovery of BT-33, a fluorinated macrobicyclic oxepanoprolinamide antibiotic with broad-spectrum activity against multidrug-resistant bacterial pathogens. Structure-activity relationships within the macrobicyclic substructure reveal structural features that are essential to the enhanced potency of BT-33 as well as its increased metabolic stability relative to its predecessors clindamycin, iboxamycin and cresomycin. Using X-ray crystallography, we determine the structure of BT-33 in complex with the bacterial ribosome revealing that its fluorine atom makes an additional van der Waals contact with nucleobase G2505. Through variable-temperature 1H NMR experiments, density functional theory calculations and vibrational circular dichroism spectroscopy, we compare macrobicyclic homologues of BT-33 and a C7 desmethyl analogue and find that the C7 methyl group of BT-33 rigidifies the macrocyclic ring in a conformation that is highly preorganized for ribosomal binding.
Collapse
Affiliation(s)
- Ben I C Tresco
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Kelvin J Y Wu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Antonio Ramkissoon
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Elena V Aleksandrova
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael Purdy
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Dominic N Y See
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Richard Y Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA.
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Andrew G Myers
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
193
|
Mavila AM, Vargas JA, Condori E, Suclupe Farro EG, Furtado AA, López JM, Gonzalez SL, Pereira HD, Marapara JL, Paredes RR, Cobos M, Castro JC, Garratt RC, Leonardo DA. Phylogenetic analysis and structural studies of heteromeric acetyl-CoA carboxylase from the oleaginous Amazonian microalgae Ankistrodesmus sp.: Insights into the BC and BCCP subunits. J Struct Biol 2025; 217:108200. [PMID: 40174731 DOI: 10.1016/j.jsb.2025.108200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Acetyl-CoA carboxylase (ACC) is an essential enzyme in fatty acid biosynthesis that catalyzes the formation of malonyl-CoA from acetyl-CoA. While structural studies on ACC components have largely focused on prokaryotes and higher plants, the assembly and molecular adaptations of ACC in microalgae remain underexplored. This study aimed to fill this gap by providing the first structural and evolutionary characterization of both biotin carboxylase (BC) and biotin carboxyl carrier protein (BCCP) from a microalga (Ankistrodesmus sp.). Phylogenetic analysis revealed distinct evolutionary trajectories for BC and BCCP, with BC forming a chlorophyte-specific clade closely related to other oleaginous species, while BCCP displayed two distinct isoforms within green algae, resulting from gene duplication. The crystallographic structure of BC was solved in its apo (1.75 Å) and ADP-Mg2+-bound (1.90 Å) states, revealing conserved conformational changes associated with cofactor binding. BCCP from Ankistrodesmus sp. displayed a unique QLGTF/H motif instead of the canonical AMKXM biotinylation motif, suggesting loss of biotinylation capacity. However, the presence of three additional lysines in the protruding thumb loop, with Lys95 as a candidate for biotin attachment, indicates potential compensatory adaptations. SEC-MALS and pull-down assays confirmed the formation of a stable 1:1 BC-BCCP complex, and circular dichroism showed increased thermal stability of the complex, supporting its structural stability. This study highlights unique structural adaptations in Ankistrodesmus sp. ACC, emphasizing the evolutionary plasticity of BC and BCCP. These insights provide a foundation for future investigations into ACC regulation in photosynthetic organisms and offer potential biotechnological applications for optimizing lipid production in microalgae.
Collapse
Affiliation(s)
- Andry Mercedes Mavila
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
| | - Jhon Antoni Vargas
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Eloy Condori
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Erick Giancarlo Suclupe Farro
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Adriano Alves Furtado
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Josué Manuel López
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru; Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Silvia Lucila Gonzalez
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
| | - Humberto D'Muniz Pereira
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Jorge Luis Marapara
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru; Departamento Académico de Ciencias Biomédicas y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria de Zungarococha, Iquitos 1600, Perú
| | - Roger Ruiz Paredes
- Facultad de Industrias Alimentarias, Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria de Zungarococha, Iquitos 1600, Peru
| | - Marianela Cobos
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru; Departamento Académico de Ciencias Biomédicas y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria de Zungarococha, Iquitos 1600, Perú
| | - Juan C Castro
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru; Departamento Académico de Ciencias Biomédicas y Biotecnología, Facultad de Ciencias Biológicas, Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria de Zungarococha, Iquitos 1600, Perú
| | - Richard Charles Garratt
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil
| | - Diego Antonio Leonardo
- Instituto de Física de São Carlos, Universidade de São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-723, Brazil.
| |
Collapse
|
194
|
Pinto Torres JE, Claes M, Hendrickx R, Yuan M, Smiejkowska N, Van Wielendaele P, Hacisuleyman A, De Winter H, Muyldermans S, Michels PAM, Walkinshaw MD, Versées W, Caljon G, Magez S, Sterckx YGJ. Allosteric inhibition of trypanosomatid pyruvate kinases by a camelid single-domain antibody. eLife 2025; 13:RP100066. [PMID: 40163365 PMCID: PMC11957543 DOI: 10.7554/elife.100066] [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] [Indexed: 04/02/2025] Open
Abstract
African trypanosomes are the causative agents of neglected tropical diseases affecting both humans and livestock. Disease control is highly challenging due to an increasing number of drug treatment failures. African trypanosomes are extracellular, blood-borne parasites that mainly rely on glycolysis for their energy metabolism within the mammalian host. Trypanosomal glycolytic enzymes are therefore of interest for the development of trypanocidal drugs. Here, we report the serendipitous discovery of a camelid single-domain antibody (sdAb aka Nanobody) that selectively inhibits the enzymatic activity of trypanosomatid (but not host) pyruvate kinases through an allosteric mechanism. By combining enzyme kinetics, biophysics, structural biology, and transgenic parasite survival assays, we provide a proof-of-principle that the sdAb-mediated enzyme inhibition negatively impacts parasite fitness and growth.
Collapse
Affiliation(s)
- Joar Esteban Pinto Torres
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
| | - Mathieu Claes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Rik Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Meng Yuan
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Natalia Smiejkowska
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Pieter Van Wielendaele
- Laboratory of Medical Biochemistry (LMB) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Aysima Hacisuleyman
- Department of Computational Biology, University of LausanneLausanneSwitzerland
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of AntwerpWilrijkBelgium
| | - Serge Muyldermans
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
| | - Paul AM Michels
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Malcolm D Walkinshaw
- School of Biological Sciences, The University of EdinburghEdinburghUnited Kingdom
| | - Wim Versées
- VIB-VUB Center for Structural Biology, VIBBrusselsBelgium
- Structural Biology Brussels, Vrije Universiteit BrusselBrusselsBelgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| | - Stefan Magez
- Laboratory for Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB)BrusselBelgium
- Center for Biomedical Research, Ghent University Global CampusIncheonRepublic of Korea
- Department for Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Yann G-J Sterckx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH) and the Infla-Med Centre of Excellence, University of AntwerpWilrijkBelgium
| |
Collapse
|
195
|
Aggarwal A, Negrean A, Chen Y, Iyer R, Reep D, Liu A, Palutla A, Xie ME, MacLennan BJ, Hagihara KM, Kinsey LW, Sun JL, Yao P, Zheng J, Tsang A, Tsegaye G, Zhang Y, Patel RH, Arthur BJ, Hiblot J, Leippe P, Tarnawski M, Marvin JS, Vevea JD, Turaga SC, Tebo AG, Carandini M, Federico Rossi L, Kleinfeld D, Konnerth A, Svoboda K, Turner GC, Hasseman J, Podgorski K. Glutamate indicators with increased sensitivity and tailored deactivation rates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.643984. [PMID: 40196590 PMCID: PMC11974752 DOI: 10.1101/2025.03.20.643984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Identifying the input-output operations of neurons requires measurements of synaptic transmission simultaneously at many of a neuron's thousands of inputs in the intact brain. To facilitate this goal, we engineered and screened 3365 variants of the fluorescent protein glutamate indicator iGluSnFR3 in neuron culture, and selected variants in the mouse visual cortex. Two variants have high sensitivity, fast activation (< 2 ms) and deactivation times tailored for recording large populations of synapses (iGluSnFR4s, 153 ms) or rapid dynamics (iGluSnFR4f, 26 ms). By imaging action-potential evoked signals on axons and visually-evoked signals on dendritic spines, we show that iGluSnFR4s/4f primarily detect local synaptic glutamate with single-vesicle sensitivity. The indicators detect a wide range of naturalistic synaptic transmission, including in the vibrissal cortex layer 4 and in hippocampal CA1 dendrites. iGluSnFR4 increases the sensitivity and scale (4s) or speed (4f) of tracking information flow in neural networks in vivo.
Collapse
Affiliation(s)
- Abhi Aggarwal
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- University of Calgary Cumming School of Medicine and Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - Adrian Negrean
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
| | - Yang Chen
- Institute of Neuroscience and Munich Cluster for Systems Neurology, Technical University of Munich, Munich, Germany
| | - Rishyashring Iyer
- Department of Physics, University of California, San Diego, La Jolla, California, USA
| | - Daniel Reep
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Anyi Liu
- University College London, Gower St, London, United Kingdom
| | - Anirudh Palutla
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Michael E. Xie
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
- Johns Hopkins University, Baltimore, MD, USA
| | | | | | - Lucas W. Kinsey
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Julianna L. Sun
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Pantong Yao
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Jihong Zheng
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Arthur Tsang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Getahun Tsegaye
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Yonghai Zhang
- Institute of Neuroscience and Munich Cluster for Systems Neurology, Technical University of Munich, Munich, Germany
| | - Ronak H. Patel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Benjamin J. Arthur
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Julien Hiblot
- Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Philipp Leippe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Wien, Austria
| | | | - Jonathan S. Marvin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Jason D. Vevea
- Neuronal Cell Biology Division, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Srinivas C. Turaga
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Alison G. Tebo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | | | - L. Federico Rossi
- University College London, Gower St, London, United Kingdom
- Center for Neuroscience and Cognitive Systems, Italian Institute of Technology, Rovereto, Italy
| | - David Kleinfeld
- Department of Physics, University of California, San Diego, La Jolla, California, USA
- Department of Neurobiology, University of California, San Diego, La Jolla, California, USA
| | - Arthur Konnerth
- Institute of Neuroscience and Munich Cluster for Systems Neurology, Technical University of Munich, Munich, Germany
| | - Karel Svoboda
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Glenn C. Turner
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Jeremy Hasseman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
- The GENIE Project Team
| | - Kaspar Podgorski
- Allen Institute for Neural Dynamics, Seattle, Washington, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| |
Collapse
|
196
|
Yun MKD, Subramanian C, Miller K, Jackson P, Radka CD, Rock CO. Isoleucine binding and regulation of Escherichia coli and Staphylococcus aureus threonine dehydratase (IlvA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641827. [PMID: 40093177 PMCID: PMC11908243 DOI: 10.1101/2025.03.06.641827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In Staphylococcus aureus, the branched-chain amino acid biosynthetic pathway provides essential intermediates for membrane biosynthesis. Threonine deaminase (IlvA) is the first enzyme in the pathway, and isoleucine feedback-regulates the enzyme in Escherichia coli. These studies on E. coli IlvA (EcIlvA) introduced the concept of allosteric regulation. To investigate the regulation of S. aureus IlvA (SaIlvA), we first conducted additional studies on EcIlvA. The previously determined crystal structure of EcIlvA revealed a tetrameric assembly of protomers, each with catalytic and regulatory domains, but the structural basis of isoleucine regulation was not characterized. Here, we present the crystal structure of the EcIlvA regulatory domain bound to isoleucine, which reveals the isoleucine binding site and conformational changes that initiate at Phe352 and propagate 23 Angstrom across the domain. This suggests an allosteric pathway that extends to the active site of the adjacent protomer, mediating regulation across the protomer-protomer interface. The EcIlvA(F352A) mutant binds isoleucine but is feedback-resistant due to the absence of the initiating Phe352. In contrast, SaIlvA is not feedback-regulated by isoleucine and does not bind it. The structure of the SaIlvA regulatory domain reveals a different organization that lacks the isoleucine binding site. Other potential allosteric inhibitors of SaIlvA, including phospholipid intermediates, do not affect enzyme activity. We propose that the absence of feedback inhibition in SaIlvA is due to its role in membrane biosynthesis. These findings enhance our understanding of IlvA's allosteric regulation and offer opportunities for engineering feedback-resistant IlvA variants for biotechnological use.
Collapse
|
197
|
Prakash D, Mitra S, Sony S, Murphy M, Andi B, Ashley L, Prasad P, Chakraborty S. Controlling outer-sphere solvent reorganization energy to turn on or off the function of artificial metalloenzymes. Nat Commun 2025; 16:3048. [PMID: 40155633 PMCID: PMC11953277 DOI: 10.1038/s41467-025-57904-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025] Open
Abstract
Metalloenzymes play essential roles in biology. However, unraveling how outer-sphere interactions can be predictably controlled to influence their functions remains a significant challenge. Inspired by Cu enzymes, we demonstrate how variations in the primary, secondary, and outer coordination-sphere interactions of de novo designed artificial copper proteins (ArCuPs) within trimeric (3SCC) and tetrameric (4SCC) self-assemblies-featuring a trigonal Cu(His)3 and a square pyramidal Cu(His)4(OH2) coordination-influence their catalytic and electron transfer properties. While 3SCC electrocatalyzes C-H oxidation, 4SCC does not. CuI-3SCC reacts more rapidly with H2O2 than O2, whereas 4SCC is less active. Electron transfer, reorganization energies, and extended H2O-mediated hydrogen bonding patterns provide insights into the observed reactivity differences. The inactivity of 4SCC is attributed to a significant solvent reorganization energy barrier mediated by a specific His---Glu hydrogen bond. When this hydrogen bond is disrupted, the solvent reorganization energy is reduced, and C-H peroxidation activity is restored.
Collapse
Affiliation(s)
- Divyansh Prakash
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Northwestern University, Evanston, IL, USA
| | - Suchitra Mitra
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Weill Cornell Medicine, New York, NY, USA
| | - Simran Sony
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Morgan Murphy
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Babak Andi
- Center for BioMolecular Structure, National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, NY, USA
| | - Landon Ashley
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
| | - Pallavi Prasad
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Saumen Chakraborty
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS, USA.
| |
Collapse
|
198
|
Nicolas WJ, Shiriaeva A, Martynowycz MW, Grey AC, Ruma YN, Donaldson PJ, Gonen T. Structure of the lens MP20 mediated adhesive junction. Nat Commun 2025; 16:2977. [PMID: 40140346 PMCID: PMC11947226 DOI: 10.1038/s41467-025-57903-6] [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: 05/21/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Human lens fiber membrane intrinsic protein MP20 is the second most abundant membrane protein of the human eye lens. Despite decades of effort its structure and function remained elusive. Here, we determined the MicroED structure of full-length human MP20 in lipidic-cubic phase to a resolution of 3.5 Å. MP20 forms tetramers each of which contain 4 transmembrane α-helices that are packed against one another forming a helical bundle. We find that each MP20 tetramer formed adhesive interactions with an opposing tetramer in a head-to-head fashion. Investigation of MP20 localization in human lenses indicate that in young fiber cells MP20 is initially localized to the cytoplasm in differentiating fiber cells but upon fiber cell maturation is inserted into the plasma membrane, correlating with the restriction of the diffusion of extracellular tracers into the lens. Together these results suggest that MP20 forms lens thin junctions in vivo, confirming its role as a structural protein in the human eye lens essential for its optical transparency.
Collapse
Affiliation(s)
- William J Nicolas
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA
| | - Anna Shiriaeva
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Michael W Martynowycz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Angus C Grey
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Yasmeen N Ruma
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA
| | - Paul J Donaldson
- Department of Physiology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Tamir Gonen
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Howard Hughes Medical Institute, University of California, Los Angeles, CA, USA.
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
199
|
Suckling RJ, Pamukcu C, Simmons RA, Fonseca D, Grant E, Harrison R, Shaikh SA, Khanolkar RC, Ghadbane H, Creese A, Hock M, Gligoris TG, Lepore M, Karuppiah V, Salio M. Molecular basis underpinning MR1 allomorph recognition by an MR1-restricted T cell receptor. Front Immunol 2025; 16:1547664. [PMID: 40207221 PMCID: PMC11979126 DOI: 10.3389/fimmu.2025.1547664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction The MHC-class-I-related molecule MR1 presents small metabolites of microbial and self-origin to T cells bearing semi-invariant or variant T cell receptors. One such T cell receptor, MC.7.G5, was previously shown to confer broad MR1-restricted reactivity to tumor cells but not normal cells, sparking interest in the development of non-MHC-restricted immunotherapy approaches. Methods/Results Here we provide cellular, biophysical, and crystallographic evidence that the MC.7.G5 TCR does not have pan-cancer specificity but is restricted to a rare allomorph of MR1, bearing the R9H mutation. Discussion Our results underscore the importance of in-depth characterization of MR1-reactive TCRs against targets expressing the full repertoire of MR1 allomorphs.
Collapse
|
200
|
Powell T, Karuppiah V, Shaikh SA, Pengelly R, Mai N, Barnbrook K, Sharma A, Harper S, Ebner M, Creese AJ. Determining T-cell receptor binding orientation and Peptide-HLA interactions using cross-linking mass spectrometry. J Biol Chem 2025; 301:108445. [PMID: 40154610 PMCID: PMC12019010 DOI: 10.1016/j.jbc.2025.108445] [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: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
T cell receptors (TCRs) recognize specific peptides presented by human leukocyte antigens (HLAs) on the surface of antigen-presenting cells and are involved in fighting pathogens and cancer surveillance. Canonical docking orientation of TCRs to their target peptide-HLAs (pHLAs) is essential for T cell activation, with reverse binding TCRs lacking functionality. TCR binding geometry and molecular interaction footprint with pHLAs are typically obtained by determining the crystal structure. Here, we describe the use of a cross-linking tandem mass spectrometry (XL-MS/MS) method to decipher the binding orientation of several TCRs to their target pHLAs. Cross-linking sites were localized to specific residues and their molecular interactions showed differentiation between TCRs binding in canonical or reverse orientations. Structural prediction and crystal structure determination of two TCR-pHLA complexes validated these findings. The XL-MS/MS method described herein offers a faster and simpler approach for elucidating TCR-pHLA binding orientation and interactions.
Collapse
Affiliation(s)
| | | | | | | | - Nicole Mai
- Immunocore Limited, Abingdon, United Kingdom
| | | | - Amit Sharma
- Immunocore Limited, Abingdon, United Kingdom
| | | | | | | |
Collapse
|