1
|
Capuano A, Cocca E, Montone AMI, D'Urso G, Del Prete S, Agrillo B, Marino C, Palmieri G, Casapullo A. Disclose ATP-synthase as a protein target of the antimicrobial peptide RiLK1 in Escherichia coli: An alternative receptor-mediated bactericidal mechanism. Food Chem 2025; 479:143838. [PMID: 40096810 DOI: 10.1016/j.foodchem.2025.143838] [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: 07/17/2024] [Revised: 02/18/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
In this study, we investigated the mechanism of action of the 10-aminoacid RiLK1 peptide against Escherichia coli (strain ATCC 25922), both in vitro and in contaminated meat matrices. Therefore, a mass spectrometry-based functional proteomics platform was employed to identify specific molecular targets of RiLK1 in a membrane protein-enriched E. coli lysate and to obtain information on their interaction mechanism. This target deconvolution approach combines MS-limited proteolysis techniques, like Drug Affinity Responsive Target Stability (DARTS) and targeted-limited Proteolysis coupled with Mass Spectrometry (t-LiP-MS). The b and δ subunits of the multimeric enzymatic complex ATP synthase, the smallest known biological nanomotor found in all cells, were identified as the relevant RiLK1 membrane protein targets. Extensive molecular docking and biochemical analyses validated and improved the suggested interaction profile. These unique findings could rationally explain the relevant RiLK1 bactericidal effects against E. coli strains, suggesting its potential application in food safety and preservation.
Collapse
Affiliation(s)
- Alessandra Capuano
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 80084 Fisciano, Italy
| | - Ennio Cocca
- Institute of Biosciences and BioResources, National Research Council (IBBR-CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | | | - Gilda D'Urso
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 80084 Fisciano, Italy
| | - Sonia Del Prete
- Institute of Biosciences and BioResources, National Research Council (IBBR-CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Bruna Agrillo
- Institute of Biosciences and BioResources, National Research Council (IBBR-CNR), Via Pietro Castellino 111, 80131 Naples, Italy
| | - Carmen Marino
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 80084 Fisciano, Italy
| | - Gianna Palmieri
- Institute of Biosciences and BioResources, National Research Council (IBBR-CNR), Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 80084 Fisciano, Italy.
| |
Collapse
|
2
|
Li Y, Gao X, Li Y, Wang Y, Yang C, Song H, Chen Y, Yan S, Li P, Gu Q. Nisin Self-assembles to Interfere with Cellular Endocytosis for Ribosome-Mediated Anti-inflammatory Efficiency. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40403150 DOI: 10.1021/acs.jafc.5c02087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Nisin, derived from Streptococcus lactis, is the only approved bacteriocin for food preservation. With wide application in multitudinous food, progressively increasing studies have focused on its anti-inflammatory activity, while its structure-effect relationship and molecular mechanism remain unclear. In this study, the anti-inflammatory efficiency and structural differences of two typical types of nisin, A and Z, were first compared based on a mouse model. The self-assembly behavior of nisin was uncovered, and the characteristics of the self-assembled nanoscale nisin were explored. Based on the transcriptomic analysis, the anti-inflammatory mechanisms of the ribosomal pathway activation of both nisin A and nisin Z were further analyzed, and the upregulation of atp7b and entpd4 triggered by A and Z, respectively, and the consequent adenosine production levels elucidated the therapeutic differences. This study deepens the understanding of the structural and molecular mechanisms underlying the anti-inflammatory activity of nisin and provides an innovative horizon for the future application of nisin.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Xin Gao
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Yapeng Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Yadi Wang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Chen Yang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Hongdi Song
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Yunle Chen
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Shihai Yan
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbiology and Nutrition of Zhejiang Province, Hangzhou 310018, Zhejiang, People's Republic of China
| |
Collapse
|
3
|
Otomo A, Hui Zhu LG, Okuni Y, Yamamoto M, Iino R. ATP synthesis of Enterococcus hirae V-ATPase driven by sodium motive force. J Biol Chem 2025; 301:108422. [PMID: 40118453 PMCID: PMC12018189 DOI: 10.1016/j.jbc.2025.108422] [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: 12/27/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
V-ATPases generally function as ion pumps driven by ATP hydrolysis in the cell, but their capability of ATP synthesis remains largely unexplored. Here we show ATP synthesis of Na+-transporting Enterococcus hirae V-ATPase (EhVoV1) driven by the electrochemical potential gradient of Na+ across the membrane (sodium motive force, smf). We reconstituted EhVoV1 into liposome and performed a luciferin/luciferase-based assay to analyze ATP synthesis quantitatively. Our result demonstrates that EhVoV1 synthesizes ATP with a rate of 4.7 s-1 under high smf (269.3 mV). The Michaelis constants for ADP (21 μM) and inorganic phosphate (2.1 mM) in ATP synthesis reaction were comparable to those for ATP synthases, suggesting similar substrate affinities among rotary ATPases regardless of their physiological functions. Both components of smf, Na+ concentration gradient across the membrane (ΔpNa) and membrane potential (Δψ), contributed to ATP synthesis, with ΔpNa showing a slightly larger impact. At the equilibrium points where smf and Gibbs free energy of ATP synthesis are balanced, EhVoV1 showed reversible reactions between ATP synthesis and hydrolysis. The obtained Na+/ATP ratio (3.2 ± 0.4) closely matched the value expected from the structural symmetry ratio between EhVo and EhV1 (10/3 = 3.3), indicating tight coupling between ATP synthesis/hydrolysis and Na+ transport. These results reveal the inherent functional reversibility of EhVoV1. We propose that the physiological function of EhVoV1in vivo is determined by relatively small smf against large Gibbs free energy of ATP synthesis, in addition to the absence of inhibitory mechanisms of ATP hydrolysis which are known for ATP synthases.
Collapse
Affiliation(s)
- Akihiro Otomo
- Institute for Molecular Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Kanazawa, Japan.
| | | | - Yasuko Okuni
- Institute for Molecular Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Mayuko Yamamoto
- Institute for Molecular Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Ryota Iino
- Institute for Molecular Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Kanazawa, Japan.
| |
Collapse
|
4
|
Zhang X, Wu J, Min Z, Wang J, Hong X, Pei X, Rao Z, Xu X. Structure of ATP synthase from an early photosynthetic bacterium Chloroflexus aurantiacus. Proc Natl Acad Sci U S A 2025; 122:e2425824122. [PMID: 40131952 PMCID: PMC12002316 DOI: 10.1073/pnas.2425824122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/28/2025] [Indexed: 03/27/2025] Open
Abstract
F-type ATP synthase (F1FO) catalyzes proton motive force-driven ATP synthesis in mitochondria, chloroplasts, and bacteria. Different from the mitochondrial and bacterial enzymes, F1FO from photosynthetic organisms have evolved diverse structural and mechanistic details to adapt to the light-dependent reactions. Although complete structure of chloroplast F1FO has been reported, no high-resolution structure of an F1FO from photosynthetic bacteria has been available. Here, we report cryo-EM structures of an intact and functionally competent F1FO from Chloroflexus aurantiacus (CaF1FO), a filamentous anoxygenic phototrophic bacterium from the earliest branch of photosynthetic organisms. The structures of CaF1FO in its ADP-free and ADP-bound forms for three rotational states reveal a previously unrecognized architecture of ATP synthases. A pair of peripheral stalks connect to the CaF1 head through a dimer of δ-subunits, and associate with two membrane-embedded a-subunits that are asymmetrically positioned outside and clamp CaFO's c10-ring. The two a-subunits constitute two proton inlets on the periplasmic side and two proton outlets on the cytoplasmic side, endowing CaF1FO with unique proton translocation pathways that allow more protons being translocated relative to single a-subunit F1FO. Our findings deepen understanding of the architecture and proton translocation mechanisms of F1FO synthases and suggest innovative strategies for modulating their activities by altering the number of a-subunit.
Collapse
Affiliation(s)
- Xin Zhang
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
- Photosynthesis Research Center, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Jingyi Wu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
- Photosynthesis Research Center, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Zhenzhen Min
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Jiamao Wang
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Xin Hong
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
- Photosynthesis Research Center, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Xinkai Pei
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
| | - Zihe Rao
- Laboratory of Structural Biology, School of Medicine, Tsinghua University, Beijing100084, China
| | - Xiaoling Xu
- Zhejiang Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou311121, China
- Photosynthesis Research Center, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou311121, China
| |
Collapse
|
5
|
Tan J, Zhang H, Liu Y, Hou Z, Wang D, Zhou J, Cao Y, Qian S, Zheng B, Nie J, Cui Y, Du Y, Huang K, Yang S, Chen D, Liu X. Interfering with proton and electron transfer enables antibacterial starvation therapy. SCIENCE ADVANCES 2025; 11:eadt3159. [PMID: 40106542 PMCID: PMC11922021 DOI: 10.1126/sciadv.adt3159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Implant-associated infections are urgently addressed; however, existing materials are difficult to kill bacteria without damaging cells. Here, we propose an innovative concept of selective antibacterial starvation therapy based on interfering with proton and electron transfer on the bacterial membrane. As a proof-of-principle demonstration, a special Schottky heterojunction film composed of gold and alkaline magnesium-iron mixed metal oxides (Au/MgFe-MMO) was constructed on the titanium implant. Once bacteria contacted this implant, the Au/MgFe-MMO film continuously captured the proton and electron participated in respiratory chain of bacteria to impede their energy metabolism, leading to the deficit of adenosine 5'-triphosphate. Prolonged exposure to this starvation state inhibited numerous biosynthesis processes and triggered severe oxidative stress in bacteria, ultimately leading to their death due to DNA and membrane damage. In addition, this heterojunction film was comfortable for mammalian cells, without inhibiting mitochondrial function. This proposed starvation antibacterial therapy gives a notable perspective in designing biosafe smart antibacterial biomaterials.
Collapse
Affiliation(s)
- Ji Tan
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Haifeng Zhang
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yisi Liu
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Zhenhao Hou
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Donghui Wang
- School of Health Science and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Junjie Zhou
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Yuanming Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
| | - Shi Qian
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Bowen Zheng
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - JingJun Nie
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | | | - Yun Du
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Kai Huang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Xuanyong Liu
- State Key Laboratory of Advanced Ceramics, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
6
|
Kubo S, Okada Y. The ATPase asymmetry: Novel computational insight into coupling diverse F O motors with tripartite F 1. Biophys J 2025; 124:891-900. [PMID: 38459696 PMCID: PMC11947463 DOI: 10.1016/j.bpj.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024] Open
Abstract
ATP synthase, a crucial enzyme for cellular bioenergetics, operates via the coordinated coupling of an FO motor, which presents variable symmetry, and a tripartite F1 motor. Despite extensive research, the understanding of their coupling dynamics, especially with non-10-fold symmetrical FO motors, remains incomplete. This study investigates the coupling patterns between eightfold and ninefold FO motors and the constant threefold F1 motor using coarse-grained molecular dynamics simulations. We unveil that in the case of a ninefold FO motor, a 3-3-3 motion is most likely to occur, whereas a 3-3-2 motion predominates with an eightfold FO motor. Furthermore, our findings propose a revised model for the coupling method, elucidating that the pathways' energy usage is primarily influenced by F1 rotation and conformational changes hindered by the b-subunits. Our results present a crucial step toward comprehending the energy landscape and mechanisms governing ATP synthase operation.
Collapse
Affiliation(s)
- Shintaroh Kubo
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Yasushi Okada
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan; Universal Biology Institute and International Research Center for Neurointelligence, The University of Tokyo, Tokyo, Japan; Laboratory for Cell Polarity Regulation, Center for Biosystems Dynamics Research (BDR), RIKEN, Osaka, Japan
| |
Collapse
|
7
|
Bruman SM, Zubareva VM, Shugaeva TE, Lapashina AS, Feniouk BA. Activation of Bacterial F-ATPase by LDAO: Deciphering the Molecular Mechanism. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:374-388. [PMID: 40367080 DOI: 10.1134/s0006297924602600] [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: 07/07/2024] [Revised: 12/06/2024] [Accepted: 01/29/2025] [Indexed: 05/16/2025]
Abstract
Proton FOF1 ATP synthase catalyzes the formation of ATP from ADP and inorganic phosphate coupled with transmembrane proton transfer using the energy of the protonmotive force (pmf). As pmf decreases, the direction of the reaction is reversed and the enzyme generates pmf, transferring protons across the membrane using the energy of ATP hydrolysis. ATPase activity of the enzyme can be suppressed by ADP in a non-competitive manner (ADP-inhibition), and in a number of bacteria, it can be inhibited by conformational changes in the regulatory C-terminal domain of the ε subunit. Lauryldimethylamine oxide (LDAO), a zwitterionic detergent, is known to attenuate both of these inhibitory mechanisms, significantly increasing the ATPase activity of the enzyme. For this reason, LDAO is sometimes used for semi-quantitative estimation of the enzyme's susceptibility to these regulatory mechanisms. However, the binding site of LDAO in ATP synthase remains unknown. The mechanism by which the detergent counteracts ADP-inhibition and the inhibition involving the ε subunit is also unclear. We performed molecular docking and predicted that LDAO binding might occur at the catalytic site of ATP synthase, whether empty or containing nucleotides. Molecular dynamics simulations showed that LDAO could affect the mobility of the loop in the β subunit (residues β404-415 in Escherichia coli ATP synthase) near the catalytic site. Mutagenesis of residue β409 in the E. coli enzyme and the corresponding β419 residue in the Bacillus subtilis ATP synthase revealed that the type of side chain of this residue indeed affects LDAO-dependent stimulation of ATPase activity. We also found that LDAO activates the enzyme more strongly in the presence of 100 mM sulfate compared to sulfate-free medium. This phenomenon is likely due to the enhancement of ADP-inhibition of the enzyme by sulfate.
Collapse
Affiliation(s)
- Sofya M Bruman
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Valeria M Zubareva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Tatiana E Shugaeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Anna S Lapashina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Boris A Feniouk
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
8
|
Sun Y, Tao H, Han H, Zou Y, Xue Y, Chen S, Tao F. Identification and expression analysis of P-type ATPase IIIA subfamily in Puccinia Striiformis f. sp. tritici. BMC Genomics 2025; 26:68. [PMID: 39856561 PMCID: PMC11759449 DOI: 10.1186/s12864-025-11219-3] [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: 09/29/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Puccinia striiformis f. sp. tritici (Pst) causes wheat stripe (yellow) rust disease, which is one of the most destructive diseases affecting wheat worldwide. ATPases, a class of membrane proteins, play an important role in material exchange and signal transduction both within and outside biological cells by transporting ions and phospholipids. In plant pathogens, P-type ATPases primarily participate in pathogen development and virulence regulation. However, the P-type ATPase of subfamily IIIA (PMA) has not yet been identified in Pst. To investigate the potential functions of the PMA gene family in Pst, we conducted a genome-wide bioinformatics analysis and examined the expression profiles of the PMA gene family. RESULTS Six PMA genes were identified in the genome of P. striiformis f. sp. tritici (CYR34 race). The PMA proteins encoded by these genes ranged in length from 811 to 960 amino acids (aa). Each of the six PMA genes contained a typical ATPase IIIA H superfamily domain and was distributed across four chromosomes. Thirty-six major cis-regulatory elements were detected within the PMA gene family members. Elements such as the CGTCA-motif and TGACG-motif play significant roles in responding to environmental stresses and hormone signals. Quantitative PCR analysis revealed that the expression of the PMA04 gene was generally higher at 9 °C under various temperature stresses. The PMA06 gene typically exhibited higher expression levels at 16 °C. During the infection of Pst, the expression levels of PMA04, PMA05, and PMA06 were elevated at 72 h post treatment. CONCLUSIONS Our results indicate that the PMA gene family in the CYR34 strain comprises six PMA genes, which are crucial for managing temperature stress and pathogen infection, and exhibit a distinctive splicing pattern. This study not only identifies a target and direction for the development of new, efficient, and environmentally friendly control agents for wheat stripe rust but also establishes a foundation for analyzing its pathogenic mechanisms.
Collapse
Affiliation(s)
- Yingjie Sun
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China
| | - Hong Tao
- Forest Seedling Service Station of Linxia Hui Autonomous Prefecture, Linxia, 731100, China
| | - Hong Han
- Academy of Agricultural Sciences of Linxia Hui Autonomous Prefecture, Linxia, 731100, China
| | - Yiping Zou
- College of Life Sciences, Shandong Normal University, Jinan, 250358, China
| | - Yingyu Xue
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China.
| | - Shiwen Chen
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China
| | - Fei Tao
- College of Plant Protection, Gansu Agricultural University, Lanzhou, 730070, China.
| |
Collapse
|
9
|
Shi S, Liu D, Wei C, Li J, Zhao C, Tian Y, Li X, Song R, Song B. A benzo[b]thiophene-derived inhibitor of virus particle assembly via targeting capsid protein residue Arg157. Int J Biol Macromol 2025; 287:138467. [PMID: 39657887 DOI: 10.1016/j.ijbiomac.2024.138467] [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: 07/15/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
As a biological macromolecule, the coat protein (CP) of potato virus Y (PVY) mediates the virus' primary pathogenic behaviors. It has been gradually realized that certain residues on the CP are crucial for functions such as virus particle movement and assembly. However, there are few reports of potential drugs successfully targeting these key residues with unique mechanisms of action. Here, we disclose the first new phytovirucide that acts on the key site Arg157 (R157) on the PVY CP. In this investigation, we developed a series of benzo[b]thiophene-based compounds, strategically introducing sulfonamide functionalities to enhance their antiviral performance. Through bio-screening, derivative C54 (EC50 = 69.2 μg/mL for inactive activity) emerged as notably more effective against PVY than the established antiviral agent ningnanmycin (EC50 = 79.6 μg/mL). Mechanistic studies revealed that C54 is an inhibitor of viral particle assembly by specifically binding to the CP residue R157, thereby disrupting its interaction with RNA. These results underscore the promise of C54 as a potent antiviral lead and provide a fresh perspective on the strategic design of inhibitors focusing on viral assembly processes.
Collapse
Affiliation(s)
- Shaojie Shi
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Deguo Liu
- College of Plant Protection, Shandong Agricultural University, NO.61 Daizong Street, Tai'an City, Shandong province 271018, China
| | - Chunle Wei
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Jianzhuan Li
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Chunni Zhao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Yanping Tian
- College of Plant Protection, Shandong Agricultural University, NO.61 Daizong Street, Tai'an City, Shandong province 271018, China.
| | - Xiangdong Li
- College of Plant Protection, Shandong Agricultural University, NO.61 Daizong Street, Tai'an City, Shandong province 271018, China
| | - Runjiang Song
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Baoan Song
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| |
Collapse
|
10
|
Zharova TV, Grivennikova VG. F o·F 1 ATP-synthase/ATPase of Paracoccus denitrificans: Mystery of Unidirectional Catalysis. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S86-S104. [PMID: 40164154 DOI: 10.1134/s000629792460399x] [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: 08/28/2024] [Revised: 09/24/2024] [Accepted: 10/16/2024] [Indexed: 04/02/2025]
Abstract
Fo·F1 ATP synthases/ATPases (Fo·F1) catalyze ATP synthesis by consuming energy of electrochemical potential of hydrogen ions (pmf), or ATP hydrolysis resulting in the pmf formation. It is generally accepted to consider Fo·F1 as a reversible chemomechanical-electrical molecular machine, however: (i) the mechanism of energy-dependent ATP synthesis is based only on the data on hydrolytic activity of the enzyme, (ii) Fo·F1 from a number of organisms effectively synthesize, but is unable to hydrolyze ATP, which indicates non-observance of the principle of microreversibility and requires development of a new hypotheses concerning the enzyme mechanism. Since 1980, the group of A. D. Vinogradov has been developing a concept according to which the elementary catalysis stages of ATP hydrolysis and ATP synthesis do not coincide, and there are two independently operating forms of Fo·F1 in the coupled membranes - pmf-generating ATPase and pmf-consuming ATP synthase. Fo·F1 of P. denitrificans as a natural model of an irreversibly functioning enzyme is a convenient object for experimental verification of the hypothesis of unidirectional energy conversion. The review considers modern concepts of the molecular mechanisms of regulation of Fo·F1 ATP synthase/ATPase of P. denitrificans and development of the hypothesis of two forms of Fo·F1.
Collapse
Affiliation(s)
- Tatiana V Zharova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - Vera G Grivennikova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
11
|
Gerle C, Jiko C, Nakano A, Yokoyama K, Gopalasingam CC, Shigematsu H, Abe K. Human F-ATP synthase as a drug target. Pharmacol Res 2024; 209:107423. [PMID: 39303772 DOI: 10.1016/j.phrs.2024.107423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Practical and conceptual barriers have kept human F-ATP synthase out of reach as a target for the treatment of human diseases. Although this situation has persisted for decades, it may change in the near future. In this review the principal functionalities of human F-ATP synthase--proton motive force / ATP interconversion, membrane bending and mitochondrial permeability transition--are surveyed in the context of their respective potential for pharmaceutical intervention. Further, the technical requirements necessary to allow drug designs that are effective at the multiple levels of functionality and modality of human F-ATP synthase are discussed. The structure-based development of gastric proton pump inhibitors is used to exemplify what might be feasible for human F-ATP synthase. And finally, four structural regions of the human F-ATP synthase are examined as potential sites for the development of structure based drug development.
Collapse
Affiliation(s)
- Christoph Gerle
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Kouto, 1-1-1, Sayo, Hyogo, Japan.
| | - Chimari Jiko
- Division of Radiation Life Science, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Atsuki Nakano
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto 603-8555, Japan
| | - Ken Yokoyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto 603-8555, Japan
| | - Chai C Gopalasingam
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Kouto, 1-1-1, Sayo, Hyogo, Japan
| | - Hideki Shigematsu
- Structural Biology Division, Japan Synchrotron Radiation Research Institute, SPring-8, Sayo, Hyogo, Japan
| | - Kazuhiro Abe
- Molecular Biochemistry Lab, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| |
Collapse
|
12
|
Martin FJO, Santiveri M, Hu H, Taylor NMI. Ion-driven rotary membrane motors: From structure to function. Curr Opin Struct Biol 2024; 88:102884. [PMID: 39053417 DOI: 10.1016/j.sbi.2024.102884] [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: 02/15/2024] [Revised: 05/16/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024]
Abstract
Ion-driven membrane motors, essential across all domains of life, convert a gradient of ions across a membrane into rotational energy, facilitating diverse biological processes including ATP synthesis, substrate transport, and bacterial locomotion. Herein, we highlight recent structural advances in the understanding of two classes of ion-driven membrane motors: rotary ATPases and 5:2 motors. The recent structure of the human F-type ATP synthase is emphasised along with the gained structural insight into clinically relevant mutations. Furthermore, we highlight the diverse roles of 5:2 motors and recent mechanistic understanding gained through the resolution of ions in the structure of a sodium-driven motor, combining insights into potential unifying mechanisms of ion selectivity and rotational torque generation in the context of their function as part of complex biological systems.
Collapse
Affiliation(s)
- Freddie J O Martin
- Structural Biology of Molecular Machines Group, Protein Structure & Function Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Mònica Santiveri
- Structural Biology of Molecular Machines Group, Protein Structure & Function Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Haidai Hu
- Structural Biology of Molecular Machines Group, Protein Structure & Function Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Nicholas M I Taylor
- Structural Biology of Molecular Machines Group, Protein Structure & Function Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
13
|
Shen J, Qiao L. Proteomic and metabolic analysis of Moorella thermoacetica-g-C 3N 4 nanocomposite system for artificial photosynthesis. Talanta 2024; 278:126479. [PMID: 38941811 DOI: 10.1016/j.talanta.2024.126479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Artificial photosynthesis by microbe-semiconductor biohybrid systems has been demonstrated as a valuable strategy in providing sustainable energy and in carbon fixation. However, most of the developed biohybrid systems for light harvesting employ heavy metal materials, especially cadmium sulfide (CdS), which normally cause environmental pollution and restrict the widespread of the systems. Herein, we constructed an environmentally friendly biohybirid system based on a typical acetogenic bacteria, Moorella thermoacetica, coupling with a carbon-based semiconductor, graphitic carbon nitride (g-C3N4), to realize light-driven carbon fixation. The proposed biohybrid system displayed outstanding acetate productivity with a quantum yield of 2.66 ± 0.43 %. Non-targeted proteomic analysis indicated that the physiological activity of the bacteria was improved, coupling with the non-toxic material. We further proposed the mechanisms of energy generation, electron transfer and CO2 fixation of the irradiated biohybrid system by proteomic and metabolomic characterization. With the photoelectron generated in g-C3N4 under illumination, CO2 is finally converted to acetate via the Wood-Ljungdahl pathway (WLP). Other associated pathways were also proved to be activated, providing extra energy or substrates for acetate production. The study reveals that the future focus of the development of biohybrid systems for light harvesting can be on the metal-free biocompatible material, which can activate the expression of the key enzymes involved in the electron transfer and carbon metabolism under light irradiation.
Collapse
Affiliation(s)
- Jiayuan Shen
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai, 200000, China
| | - Liang Qiao
- Department of Chemistry, and Minhang Hospital, Fudan University, Shanghai, 200000, China.
| |
Collapse
|
14
|
García-Martín J, García-Abad L, Santamaría RI, Díaz M. Functional connexion of bacterioferritin in antibiotic production and morphological differentiation in Streptomyces coelicolor. Microb Cell Fact 2024; 23:234. [PMID: 39182107 PMCID: PMC11344345 DOI: 10.1186/s12934-024-02510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Several two-component systems of Streptomyces coelicolor, a model organism used for studying antibiotic production in Streptomyces, affect the expression of the bfr (SCO2113) gene that encodes a bacterioferritin, a protein involved in iron storage. In this work, we have studied the effect of the deletion mutant ∆bfr in S. coelicolor. RESULTS The ∆bfr mutant exhibits a delay in morphological differentiation and produces a lesser amount of the two pigmented antibiotics (actinorhodin and undecylprodigiosin) compared to the wild type on complex media. The effect of iron in minimal medium was tested in the wild type and ∆bfr mutant. Consequently, we also observed different levels of production of the two pigmented antibiotics between the two strains, depending on the iron concentration and the medium (solid or liquid) used. Contrary to expectations, no differences in intracellular iron concentration were detected between the wild type and ∆bfr mutant. However, a higher level of reactive oxygen species in the ∆bfr mutant and a higher tolerance to oxidative stress were observed. Proteomic analysis showed no variation in iron response proteins, but there was a lower abundance of proteins related to actinorhodin and ribosomal proteins, as well as others related to secondary metabolite production and differentiation. Additionally, a higher abundance of proteins related to various types of stress, such as respiration and hypoxia among others, was also revealed. Data are available via ProteomeXchange with identifier PXD050869. CONCLUSION This bacterioferritin in S. coelicolor (Bfr) is a new element in the complex regulation of secondary metabolism in S. coelicolor and, additionally, iron acts as a signal to modulate the biosynthesis of active molecules. Our model proposes an interaction between Bfr and iron-containing regulatory proteins. Thus, identifying these interactions would provide new information for improving antibiotic production in Streptomyces.
Collapse
Affiliation(s)
- Javier García-Martín
- Departamento de Microbiología y Genética, Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca (USAL), C/ Zacarías González, nº 2, Salamanca, 37007, Spain
| | - Laura García-Abad
- Departamento de Microbiología y Genética, Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca (USAL), C/ Zacarías González, nº 2, Salamanca, 37007, Spain
| | - Ramón I Santamaría
- Departamento de Microbiología y Genética, Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca (USAL), C/ Zacarías González, nº 2, Salamanca, 37007, Spain.
| | - Margarita Díaz
- Departamento de Microbiología y Genética, Instituto de Biología Funcional y Genómica (IBFG), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca (USAL), C/ Zacarías González, nº 2, Salamanca, 37007, Spain.
| |
Collapse
|
15
|
Thitirungreangchai T, Roytrakul S, Aunpad R. Deciphering the Intracellular Action of the Antimicrobial Peptide A11 via an In-Depth Analysis of Its Effect on the Global Proteome of Acinetobacter baumannii. ACS Infect Dis 2024; 10:2795-2813. [PMID: 39075773 PMCID: PMC11320580 DOI: 10.1021/acsinfecdis.4c00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/31/2024]
Abstract
The potential antimicrobial activity and low propensity to induce the development of bacterial resistance have rendered antimicrobial peptides (AMPs) as novel and ideal candidate therapeutic agents for the treatment of infections caused by drug-resistant pathogenic bacteria. The targeting of bacterial membranes by AMPs has been typically considered their sole mode of action; however, increasing evidence supports the existence of multiple and complementary functions of AMPs that result in bacterial death. An in-depth characterization of their mechanism of action could facilitate further research and development of AMPs with higher potency. The current study employs biophysics and proteomics approaches to unveil the mechanisms underlying the antibacterial activity of A11, a potential candidate AMP, against Acinetobacter baumannii, a leading cause of hospital-acquired infections (HAIs) and consequently, a serious global threat. A11 peptide was found to induce membrane depolarization to a high extent, as revealed by flow cytometry and electron microscopy analyses. The prompt intracellular penetration of A11 peptide, observed using confocal microscopy, was found to occur concomitantly with a very low degree of membrane lysis, suggesting that its mode of action predominantly involves a nonlytic killing mechanism. Quantitative proteomics analysis employed for obtaining insights into the mechanisms underlying the antimicrobial activity of A11 peptide revealed that it disrupted energy metabolism, interfered with protein homeostasis, and inhibited fatty acid synthesis that is essential for cell membrane integrity; all these impacted the cellular functions of A. baumannii. A11 treatment also impacted signal transduction associated with the regulation of biofilm formation, hindered the stress response, and influenced DNA repair processes; these are all crucial survival mechanisms of A. baumannii. Additionally, robust antibacterial activity was exhibited by A11 peptide against multidrug-resistant (MDR) and extensively drug-resistant (XDR) clinical isolates of A. baumannii; moreover, A11 peptide exhibited synergy with levofloxacin and minocycline as well as low propensity for inducing resistance. Taken together, the findings emphasize the therapeutic potential of A11 peptide as an antibacterial agent against drug-resistant A. baumannii and underscore the need for further investigation.
Collapse
Affiliation(s)
- Thanit Thitirungreangchai
- Graduate
Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sittiruk Roytrakul
- Functional
Proteomics Technology Laboratory, National Center for Genetic Engineering
and Biotechnology, National Science and
Technology Development Agency, Khlong Luang, Pathum Thani 12120, Thailand
| | - Ratchaneewan Aunpad
- Graduate
Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Khlong Luang, Pathum Thani 12120, Thailand
| |
Collapse
|
16
|
Shiraliyev R, Orman MA. Metabolic disruption impairs ribosomal protein levels, resulting in enhanced aminoglycoside tolerance. eLife 2024; 13:RP94903. [PMID: 39093940 PMCID: PMC11296704 DOI: 10.7554/elife.94903] [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: 08/04/2024] Open
Abstract
Aminoglycoside antibiotics target ribosomes and are effective against a wide range of bacteria. Here, we demonstrated that knockout strains related to energy metabolism in Escherichia coli showed increased tolerance to aminoglycosides during the mid-exponential growth phase. Contrary to expectations, these mutations did not reduce the proton motive force or aminoglycoside uptake, as there were no significant changes in metabolic indicators or intracellular gentamicin levels between wild-type and mutant strains. Our comprehensive proteomics analysis unveiled a noteworthy upregulation of proteins linked to the tricarboxylic acid (TCA) cycle in the mutant strains during the mid-exponential growth phase, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research provides valuable insights into the mechanisms of aminoglycoside tolerance, paving the way for novel strategies to combat such cells.
Collapse
Affiliation(s)
- Rauf Shiraliyev
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of HoustonHoustonUnited States
| | - Mehmet A Orman
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of HoustonHoustonUnited States
| |
Collapse
|
17
|
Mishra A, Chakraborty S, Jaiswal TP, Bhattacharjee S, Kesarwani S, Mishra AK, Singh SS. Untangling the adaptive strategies of thermophilic bacterium Anoxybacillus rupiensis TPH1 under low temperature. Extremophiles 2024; 28:31. [PMID: 39020126 DOI: 10.1007/s00792-024-01346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/10/2024] [Indexed: 07/19/2024]
Abstract
The present study investigates the low temperature tolerance strategies of thermophilic bacterium Anoxybacillus rupiensis TPH1, which grows optimally at 55 °C , by subjecting it to a temperature down-shift of 10 °C (45 °C) for 4 and 6 h followed by studying its growth, morphophysiological, molecular and proteomic responses. Results suggested that although TPH1 experienced increased growth inhibition, ROS production, protein oxidation and membrane disruption after 4 h of incubation at 45 °C yet maintained its DNA integrity and cellular structure through the increased expression of DNA damage repair and cell envelop synthesizing proteins and also progressively alleviated growth inhibition by 20% within two hours i.e., 6 h, by inducing the expression of antioxidative enzymes, production of unsaturated fatty acids, capsular and released exopolysaccharides and forming biofilm along with chemotaxis proteins. Conclusively, the adaptation of Anoxybacillus rupiensis TPH1 to lower temperature is mainly mediated by the synthesis of large numbers of defense proteins and exopolysaccharide rich biofilm formation.
Collapse
Affiliation(s)
- Aditi Mishra
- Laboratory of Cyanobacterial Systematics and Stress Biology, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Sindhunath Chakraborty
- Laboratory of Microbial Genetics, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Tameshwar Prasad Jaiswal
- Laboratory of Cyanobacterial Systematics and Stress Biology, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Samujjal Bhattacharjee
- Laboratory of Microbial Genetics, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Shreya Kesarwani
- Laboratory of Cyanobacterial Systematics and Stress Biology, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Arun Kumar Mishra
- Laboratory of Microbial Genetics, Department of Botany, Banaras Hindu University, Varanasi, India
| | - Satya Shila Singh
- Laboratory of Cyanobacterial Systematics and Stress Biology, Department of Botany, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
18
|
Shiraliyev R, Orman MA. METABOLIC DISRUPTION IMPAIRS RIBOSOMAL PROTEIN LEVELS, RESULTING IN ENHANCED AMINOGLYCOSIDE TOLERANCE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.20.572673. [PMID: 38187583 PMCID: PMC10769322 DOI: 10.1101/2023.12.20.572673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Aminoglycoside antibiotics display broad-spectrum activity against Gram-negative and Grampositive bacteria by targeting their ribosomes. Herein, we have demonstrated that energy metabolism plays a crucial role in aminoglycoside tolerance, as knockout strains associated with the tricarboxylic acid cycle (TCA) and the electron transport chain (ETC) exhibited increased tolerance to aminoglycosides in the mid-exponential growth phase of Escherichia coli cells. Given that aminoglycoside uptake relies on the energy-driven electrochemical potential across the cytoplasmic membrane, our initial expectation was that these genetic perturbations would decrease the proton motive force (PMF), subsequently affecting the uptake of aminoglycosides. However, our results did not corroborate this assumption. We found no consistent metabolic changes, ATP levels, cytoplasmic pH variations, or membrane potential differences in the mutant strains compared to the wild type. Additionally, intracellular concentrations of fluorophore-labeled gentamicin remained similar across all strains. To uncover the mechanism responsible for the observed tolerance in mutant strains, we employed untargeted mass spectrometry to quantify the proteins within these mutants and subsequently compared them to their wild-type counterparts. Our comprehensive analysis, which encompassed protein-protein association networks and functional enrichment, unveiled a noteworthy upregulation of proteins linked to the TCA cycle in the mutant strains during the mid-exponential growth phase, suggesting that these strains compensate for the perturbation in their energy metabolism by increasing TCA cycle activity to maintain their membrane potential and ATP levels. Furthermore, our pathway enrichment analysis shed light on local network clusters displaying downregulation across all mutant strains, which were associated with both large and small ribosomal binding proteins, ribosome biogenesis, translation factor activity, and the biosynthesis of ribonucleoside monophosphates. These findings offer a plausible explanation for the observed tolerance of aminoglycosides in the mutant strains. Altogether, this research has the potential to uncover mechanisms behind aminoglycoside tolerance, paving the way for novel strategies to combat such cells.
Collapse
Affiliation(s)
- Rauf Shiraliyev
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204
| | - Mehmet A Orman
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, 77204
| |
Collapse
|
19
|
Kohram M, Sanderson AE, Loui A, Thompson PV, Vashistha H, Shomar A, Oltvai ZN, Salman H. Nonlethal deleterious mutation-induced stress accelerates bacterial aging. Proc Natl Acad Sci U S A 2024; 121:e2316271121. [PMID: 38709929 PMCID: PMC11098108 DOI: 10.1073/pnas.2316271121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/29/2024] [Indexed: 05/08/2024] Open
Abstract
Random mutagenesis, including when it leads to loss of gene function, is a key mechanism enabling microorganisms' long-term adaptation to new environments. However, loss-of-function mutations are often deleterious, triggering, in turn, cellular stress and complex homeostatic stress responses, called "allostasis," to promote cell survival. Here, we characterize the differential impacts of 65 nonlethal, deleterious single-gene deletions on Escherichia coli growth in three different growth environments. Further assessments of select mutants, namely, those bearing single adenosine triphosphate (ATP) synthase subunit deletions, reveal that mutants display reorganized transcriptome profiles that reflect both the environment and the specific gene deletion. We also find that ATP synthase α-subunit deleted (ΔatpA) cells exhibit elevated metabolic rates while having slower growth compared to wild-type (wt) E. coli cells. At the single-cell level, compared to wt cells, individual ΔatpA cells display near normal proliferation profiles but enter a postreplicative state earlier and exhibit a distinct senescence phenotype. These results highlight the complex interplay between genomic diversity, adaptation, and stress response and uncover an "aging cost" to individual bacterial cells for maintaining population-level resilience to environmental and genetic stress; they also suggest potential bacteriostatic antibiotic targets and -as select human genetic diseases display highly similar phenotypes, - a bacterial origin of some human diseases.
Collapse
Affiliation(s)
- Maryam Kohram
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA15260
| | - Amy E. Sanderson
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA15260
| | - Alicia Loui
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA15260
| | | | - Harsh Vashistha
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA15260
| | - Aseel Shomar
- Department of Chemical Engineering, Technion–Israel Institute of Technology, Haifa32000, Israel
| | - Zoltán N. Oltvai
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA15260
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA15260
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, NY14627
| | - Hanna Salman
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, PA15260
| |
Collapse
|
20
|
Ivontsin LA, Mashkovtseva EV, Nartsissov YR. Molecular Dynamics Simulations of the Mutated Proton-Transferring a-Subunit of E. coli F oF 1-ATP Synthase. Int J Mol Sci 2024; 25:5143. [PMID: 38791189 PMCID: PMC11121307 DOI: 10.3390/ijms25105143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
The membrane Fo factor of ATP synthase is highly sensitive to mutations in the proton half-channel leading to the functional blocking of the entire protein. To identify functionally important amino acids for the proton transport, we performed molecular dynamic simulations on the selected mutants of the membrane part of the bacterial FoF1-ATP synthase embedded in a native lipid bilayer: there were nine different mutations of a-subunit residues (aE219, aH245, aN214, aQ252) in the inlet half-channel. The structure proved to be stable to these mutations, although some of them (aH245Y and aQ252L) resulted in minor conformational changes. aH245 and aN214 were crucial for proton transport as they directly facilitated H+ transfer. The substitutions with nonpolar amino acids disrupted the transfer chain and water molecules or neighboring polar side chains could not replace them effectively. aE219 and aQ252 appeared not to be determinative for proton translocation, since an alternative pathway involving a chain of water molecules could compensate the ability of H+ transmembrane movement when they were substituted. Thus, mutations of conserved polar residues significantly affected hydration levels, leading to drastic changes in the occupancy and capacity of the structural water molecule clusters (W1-W3), up to their complete disappearance and consequently to the proton transfer chain disruption.
Collapse
Affiliation(s)
- Leonid A. Ivontsin
- Institute of Cytochemistry and Molecular Pharmacology, 24/14 6th Radialnaya Street, Moscow 115404, Russia;
| | - Elena V. Mashkovtseva
- Institute of Cytochemistry and Molecular Pharmacology, 24/14 6th Radialnaya Street, Moscow 115404, Russia;
| | - Yaroslav R. Nartsissov
- Institute of Cytochemistry and Molecular Pharmacology, 24/14 6th Radialnaya Street, Moscow 115404, Russia;
- Biomedical Research Group, BiDiPharma GmbH, 5 Bültbek, 22962 Siek, Germany
| |
Collapse
|
21
|
Hołyst R, Bubak G, Kalwarczyk T, Kwapiszewska K, Michalski J, Pilz M. Living Cell as a Self-Synchronized Chemical Reactor. J Phys Chem Lett 2024; 15:3559-3570. [PMID: 38526849 PMCID: PMC11000238 DOI: 10.1021/acs.jpclett.4c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/21/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Thermal fluctuations power all processes inside living cells. Therefore, these processes are inherently random. However, myriad multistep chemical reactions act in concerto inside a cell, finally leading to this chemical reactor's self-replication. We speculate that an underlying mechanism in nature must exist that allows all of these reactions to synchronize at multiple time and length scales, overcoming in this way the random nature of any single process in a cell. This Perspective discusses what type of research is needed to understand this undiscovered synchronization law.
Collapse
Affiliation(s)
- Robert Hołyst
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Grzegorz Bubak
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Tomasz Kalwarczyk
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Karina Kwapiszewska
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Jarosław Michalski
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Marta Pilz
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
22
|
Blanc FEC, Hummer G. Mechanism of proton-powered c-ring rotation in a mitochondrial ATP synthase. Proc Natl Acad Sci U S A 2024; 121:e2314199121. [PMID: 38451940 PMCID: PMC10945847 DOI: 10.1073/pnas.2314199121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/10/2024] [Indexed: 03/09/2024] Open
Abstract
Proton-powered c-ring rotation in mitochondrial ATP synthase is crucial to convert the transmembrane protonmotive force into torque to drive the synthesis of adenosine triphosphate (ATP). Capitalizing on recent cryo-EM structures, we aim at a structural and energetic understanding of how functional directional rotation is achieved. We performed multi-microsecond atomistic simulations to determine the free energy profiles along the c-ring rotation angle before and after the arrival of a new proton. Our results reveal that rotation proceeds by dynamic sliding of the ring over the a-subunit surface, during which interactions with conserved polar residues stabilize distinct intermediates. Ordered water chains line up for a Grotthuss-type proton transfer in one of these intermediates. After proton transfer, a high barrier prevents backward rotation and an overall drop in free energy favors forward rotation, ensuring the directionality of c-ring rotation required for the thermodynamically disfavored ATP synthesis. The essential arginine of the a-subunit stabilizes the rotated configuration through a salt bridge with the c-ring. Overall, we describe a complete mechanism for the rotation step of the ATP synthase rotor, thereby illuminating a process critical to all life at atomic resolution.
Collapse
Affiliation(s)
- Florian E. C. Blanc
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main60438, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main60438, Germany
- Institute for Biophysics, Goethe University Frankfurt, Frankfurt am Main60438, Germany
| |
Collapse
|
23
|
Zhang M, Luo X, Zhang B, Luo D, Huang L, Long Q. Unveiling OSCP as the potential therapeutic target for mitochondrial dysfunction-related diseases. Life Sci 2024; 336:122293. [PMID: 38030056 DOI: 10.1016/j.lfs.2023.122293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Mitochondria are important organelles in cells responsible for energy production and regulation. Mitochondrial dysfunction has been implicated in the pathogenesis of many diseases. Oligomycin sensitivity-conferring protein (OSCP), a component of the inner mitochondrial membrane, has been studied for a long time. OSCP is a component of the F1Fo-ATP synthase in mitochondria and is closely related to the regulation of the mitochondrial permeability transition pore (mPTP). Studies have shown that OSCP plays an important role in cardiovascular disease, neurological disorders, and tumor development. This review summarizes the localization, structure, function, and regulatory mechanisms of OSCP and outlines its role in cardiovascular disease, neurological disease, and tumor development. In addition, this article reviews the research on the interaction between OSCP and mPTP. Finally, the article suggests future research directions, including further exploration of the mechanism of action of OSCP, the interaction between OSCP and other proteins and signaling pathways, and the development of new treatment strategies for mitochondrial dysfunction. In conclusion, in-depth research on OSCP will help to elucidate its importance in cell function and disease and provide new ideas for the treatment and prevention of related diseases.
Collapse
Affiliation(s)
- Mingyue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xia Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Binzhi Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Lizhen Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qinqiang Long
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine (Institute of Chinese Medicine), Guangdong Pharmaceutical University, Guangzhou 510006, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
24
|
Abstract
Synthetic chemistry has traditionally relied on reactions between reactants of high chemical potential and transformations that proceed energetically downhill to either a global or local minimum (thermodynamic or kinetic control). Catalysts can be used to manipulate kinetic control, lowering activation energies to influence reaction outcomes. However, such chemistry is still constrained by the shape of one-dimensional reaction coordinates. Coupling synthesis to an orthogonal energy input can allow ratcheting of chemical reaction outcomes, reminiscent of the ways that molecular machines ratchet random thermal motion to bias conformational dynamics. This fundamentally distinct approach to synthesis allows multi-dimensional potential energy surfaces to be navigated, enabling reaction outcomes that cannot be achieved under conventional kinetic or thermodynamic control. In this Review, we discuss how ratcheted synthesis is ubiquitous throughout biology and consider how chemists might harness ratchet mechanisms to accelerate catalysis, drive chemical reactions uphill and programme complex reaction sequences.
Collapse
Affiliation(s)
- Stefan Borsley
- Department of Chemistry, University of Manchester, Manchester, UK
| | | | - David A Leigh
- Department of Chemistry, University of Manchester, Manchester, UK.
| | | |
Collapse
|
25
|
Zhang N, Shan W, Gao L, Kou SH, Lu C, Yang H, Peng B, Tam KY, Lee LTO, Zheng J. Repurposing the Hedgehog pathway inhibitor, BMS-833923, as a phosphatidylglycerol-selective membrane-disruptive colistin adjuvant against ESKAPE pathogens. Int J Antimicrob Agents 2023; 62:106888. [PMID: 37328075 DOI: 10.1016/j.ijantimicag.2023.106888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/18/2023]
Abstract
The rapid emergence and spread of multi-drug- or pan-drug-resistant bacterial pathogens, such as ESKAPE, pose a serious threat to global health. However, the development of novel antibiotics is hindered by difficulties in identifying new antibiotic targets and the rapid development of drug resistance. Drug repurposing is an effective alternative strategy for combating antibiotic resistance that both saves resources and extends the life of existing antibiotics in combination treatment regimens. Screening of a chemical compound library identified BMS-833923 (BMS), a smoothened antagonist that kills Gram-positive bacteria directly, and potentiates colistin to destroy various Gram-negative bacteria. BMS did not induce detectable antibiotic resistance in vitro, and showed effective activity against drug-resistant bacteria in vivo. Mechanistic studies revealed that BMS caused membrane disruption by targeting the membrane phospholipids phosphatidylglycerol and cardiolipin, promoting membrane dysfunction, metabolic disturbance, leakage of cellular components, and, ultimately, cell death. This study describes a potential strategy to enhance the efficacy of colistin and combat multi-drug-resistant ESKAPE pathogens.
Collapse
Affiliation(s)
- Nian Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Si Hoi Kou
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Chang Lu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Huilin Yang
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Bo Peng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo Tsz On Lee
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Ministry of Education Frontiers Science Centre for Precision Oncology, University of Macau, Taipa, Macau, China; Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
26
|
Courbon GM, Palme PR, Mann L, Richter A, Imming P, Rubinstein JL. Mechanism of mycobacterial ATP synthase inhibition by squaramides and second generation diarylquinolines. EMBO J 2023; 42:e113687. [PMID: 37377118 PMCID: PMC10390873 DOI: 10.15252/embj.2023113687] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Mycobacteria, such as Mycobacterium tuberculosis, depend on the activity of adenosine triphosphate (ATP) synthase for growth. The diarylquinoline bedaquiline (BDQ), a mycobacterial ATP synthase inhibitor, is an important medication for treatment of drug-resistant tuberculosis but suffers from off-target effects and is susceptible to resistance mutations. Consequently, both new and improved mycobacterial ATP synthase inhibitors are needed. We used electron cryomicroscopy and biochemical assays to study the interaction of Mycobacterium smegmatis ATP synthase with the second generation diarylquinoline TBAJ-876 and the squaramide inhibitor SQ31f. The aryl groups of TBAJ-876 improve binding compared with BDQ, while SQ31f, which blocks ATP synthesis ~10 times more potently than ATP hydrolysis, binds a previously unknown site in the enzyme's proton-conducting channel. Remarkably, BDQ, TBAJ-876, and SQ31f all induce similar conformational changes in ATP synthase, suggesting that the resulting conformation is particularly suited for drug binding. Further, high concentrations of the diarylquinolines uncouple the transmembrane proton motive force while for SQ31f they do not, which may explain why high concentrations of diarylquinolines, but not SQ31f, have been reported to kill mycobacteria.
Collapse
Affiliation(s)
- Gautier M Courbon
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoONCanada
- Department of Medical BiophysicsThe University of TorontoTorontoONCanada
| | - Paul R Palme
- Institut für PharmazieMartin‐Luther‐Universität Halle‐WittenbergHalle, SaaleGermany
| | - Lea Mann
- Institut für PharmazieMartin‐Luther‐Universität Halle‐WittenbergHalle, SaaleGermany
| | - Adrian Richter
- Institut für PharmazieMartin‐Luther‐Universität Halle‐WittenbergHalle, SaaleGermany
| | - Peter Imming
- Institut für PharmazieMartin‐Luther‐Universität Halle‐WittenbergHalle, SaaleGermany
| | - John L Rubinstein
- Molecular Medicine ProgramThe Hospital for Sick ChildrenTorontoONCanada
- Department of Medical BiophysicsThe University of TorontoTorontoONCanada
- Department of BiochemistryThe University of TorontoTorontoONCanada
| |
Collapse
|
27
|
Bernardi P, Gerle C, Halestrap AP, Jonas EA, Karch J, Mnatsakanyan N, Pavlov E, Sheu SS, Soukas AA. Identity, structure, and function of the mitochondrial permeability transition pore: controversies, consensus, recent advances, and future directions. Cell Death Differ 2023; 30:1869-1885. [PMID: 37460667 PMCID: PMC10406888 DOI: 10.1038/s41418-023-01187-0] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
The mitochondrial permeability transition (mPT) describes a Ca2+-dependent and cyclophilin D (CypD)-facilitated increase of inner mitochondrial membrane permeability that allows diffusion of molecules up to 1.5 kDa in size. It is mediated by a non-selective channel, the mitochondrial permeability transition pore (mPTP). Sustained mPTP opening causes mitochondrial swelling, which ruptures the outer mitochondrial membrane leading to subsequent apoptotic and necrotic cell death, and is implicated in a range of pathologies. However, transient mPTP opening at various sub-conductance states may contribute several physiological roles such as alterations in mitochondrial bioenergetics and rapid Ca2+ efflux. Since its discovery decades ago, intensive efforts have been made to identify the exact pore-forming structure of the mPT. Both the adenine nucleotide translocase (ANT) and, more recently, the mitochondrial F1FO (F)-ATP synthase dimers, monomers or c-subunit ring alone have been implicated. Here we share the insights of several key investigators with different perspectives who have pioneered mPT research. We critically assess proposed models for the molecular identity of the mPTP and the mechanisms underlying its opposing roles in the life and death of cells. We provide in-depth insights into current controversies, seeking to achieve a degree of consensus that will stimulate future innovative research into the nature and role of the mPTP.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Christoph Gerle
- Laboratory of Protein Crystallography, Institute for Protein Research, Osaka University, Suita, Japan
| | - Andrew P Halestrap
- School of Biochemistry and Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Jason Karch
- Department of Integrative Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Nelli Mnatsakanyan
- Department of Cellular and Molecular Physiology, College of Medicine, Penn State University, State College, PA, USA
| | - Evgeny Pavlov
- Department of Molecular Pathobiology, New York University, New York, NY, USA
| | - Shey-Shing Sheu
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Alexander A Soukas
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Kubo S, Niina T, Takada S. F O-F 1 coupling and symmetry mismatch in ATP synthase resolved in every F O rotation step. Biophys J 2023; 122:2898-2909. [PMID: 36171725 PMCID: PMC10397808 DOI: 10.1016/j.bpj.2022.09.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/18/2022] [Accepted: 09/26/2022] [Indexed: 11/19/2022] Open
Abstract
FOF1 ATP synthase, a ubiquitous enzyme that synthesizes most ATP in living cells, is composed of two rotary motors: a membrane-embedded proton-driven FO motor and a catalytic F1 motor. These motors share both central and peripheral stalks. Although both FO and F1 have pseudo-symmetric structures, their symmetries do not match. How symmetry mismatch is solved remains elusive because of the missing intermediate structures of the rotational steps. Here, for the case of Bacillus PS3 ATP synthases with three- and 10-fold symmetries in F1 and FO, respectively, we uncovered the mechanical couplings between FO and F1 at every 36° rotation step via molecular dynamics simulations and comparative studies of cryoelectron microscopy (cryo-EM) structures from three species. We found that the mismatch could be solved using several elements: 1) the F1 head partially rotates relative to the FO a subunit via elastic distortion of the b subunits, 2) the rotor is twisted, and 3) comparisons of cryo-EM structures further suggest that the c ring rotary angles can deviate from the symmetric ones. In addition, the F1 motor may have non-canonical structures, relieving stronger frustration. Thus, we provide new insights for solving the symmetry mismatch problem.
Collapse
Affiliation(s)
- Shintaroh Kubo
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan; Department of Anatomy and Cell Biology, McGill University, Montréal, Québec H3A 0C7, Canada.
| | - Toru Niina
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | - Shoji Takada
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
29
|
Nakano A, Kishikawa JI, Mitsuoka K, Yokoyama K. Mechanism of ATP hydrolysis dependent rotation of bacterial ATP synthase. Nat Commun 2023; 14:4090. [PMID: 37429854 DOI: 10.1038/s41467-023-39742-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
F1 domain of ATP synthase is a rotary ATPase complex in which rotation of central γ-subunit proceeds in 120° steps against a surrounding α3β3 fueled by ATP hydrolysis. How the ATP hydrolysis reactions occurring in three catalytic αβ dimers are coupled to mechanical rotation is a key outstanding question. Here we describe catalytic intermediates of the F1 domain in FoF1 synthase from Bacillus PS3 sp. during ATP mediated rotation captured using cryo-EM. The structures reveal that three catalytic events and the first 80° rotation occur simultaneously in F1 domain when nucleotides are bound at all the three catalytic αβ dimers. The remaining 40° rotation of the complete 120° step is driven by completion of ATP hydrolysis at αDβD, and proceeds through three sub-steps (83°, 91°, 101°, and 120°) with three associated conformational intermediates. All sub-steps except for one between 91° and 101° associated with phosphate release, occur independently of the chemical cycle, suggesting that the 40° rotation is largely driven by release of intramolecular strain accumulated by the 80° rotation. Together with our previous results, these findings provide the molecular basis of ATP driven rotation of ATP synthases.
Collapse
Affiliation(s)
- Atsuki Nakano
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan
| | - Jun-Ichi Kishikawa
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kaoru Mitsuoka
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Ken Yokoyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto, 603-8555, Japan.
| |
Collapse
|
30
|
Lai Y, Zhang Y, Zhou S, Xu J, Du Z, Feng Z, Yu L, Zhao Z, Wang W, Tang Y, Yang X, Guddat LW, Liu F, Gao Y, Rao Z, Gong H. Structure of the human ATP synthase. Mol Cell 2023:S1097-2765(23)00324-6. [PMID: 37244256 DOI: 10.1016/j.molcel.2023.04.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/06/2023] [Accepted: 04/28/2023] [Indexed: 05/29/2023]
Abstract
Biological energy currency ATP is produced by F1Fo-ATP synthase. However, the molecular mechanism for human ATP synthase action remains unknown. Here, we present snapshot images for three main rotational states and one substate of human ATP synthase using cryoelectron microscopy. These structures reveal that the release of ADP occurs when the β subunit of F1Fo-ATP synthase is in the open conformation, showing how ADP binding is coordinated during synthesis. The accommodation of the symmetry mismatch between F1 and Fo motors is resolved by the torsional flexing of the entire complex, especially the γ subunit, and the rotational substep of the c subunit. Water molecules are identified in the inlet and outlet half-channels, suggesting that the proton transfer in these two half-channels proceed via a Grotthus mechanism. Clinically relevant mutations are mapped to the structure, showing that they are mainly located at the subunit-subunit interfaces, thus causing instability of the complex.
Collapse
Affiliation(s)
- Yuezheng Lai
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuying Zhang
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shan Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jinxu Xu
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhanqiang Du
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ziyan Feng
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Long Yu
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ziqing Zhao
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiwei Wang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yanting Tang
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiuna Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Luke W Guddat
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Fengjiang Liu
- Innovative Center for Pathogen Research, Guangzhou Laboratory, Guangzhou 510005, China.
| | - Yan Gao
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Zihe Rao
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China; Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Innovative Center for Pathogen Research, Guangzhou Laboratory, Guangzhou 510005, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing 100101, China; Laboratory of Structural Biology, Tsinghua University, Beijing 100084, China.
| | - Hongri Gong
- State Key Laboratory of Medicinal Chemical Biology and Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute for Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
31
|
Kelam LM, Wani MA, Dhaked DK. An update on ATP synthase inhibitors: A unique target for drug development in M. tuberculosis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 180-181:87-104. [PMID: 37105260 DOI: 10.1016/j.pbiomolbio.2023.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
ATP synthase is a key protein in the oxidative phosphorylation process, as it aids in the effective production of ATP (Adenosine triphosphate) in all life's of kingdoms. ATP synthases have distinctive properties that contribute to efficient ATP synthesis. The ATP synthase of mycobacterium is of special relevance since it has been identified as a target for potential anti-TB molecules, especially Bedaquiline (BDQ). Better knowledge of how mycobacterial ATP synthase functions and its peculiar characteristics will aid in our understanding of bacterial energy metabolism adaptations. Furthermore, identifying and understanding the important distinctions between human ATP synthase and bacterial ATP synthase may provide insight into the design and development of inhibitors that target specific ATP synthase. In recent years, many potential candidates targeting the ATP synthase of mycobacterium have been developed. In this review, we discuss the druggable targets of the Electron transport chain (ETC) and recently identified potent inhibitors (including clinical molecules) from 2015 to 2022 of diverse classes that target ATP synthase of M. tuberculosis.
Collapse
Affiliation(s)
- Lakshmi Mounika Kelam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Mushtaq Ahmad Wani
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Devendra K Dhaked
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
32
|
Yokoyama K. Rotary mechanism of V/A-ATPases-how is ATP hydrolysis converted into a mechanical step rotation in rotary ATPases? Front Mol Biosci 2023; 10:1176114. [PMID: 37168257 PMCID: PMC10166205 DOI: 10.3389/fmolb.2023.1176114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
V/A-ATPase is a rotary molecular motor protein that produces ATP through the rotation of its central rotor. The soluble part of this protein, the V1 domain, rotates upon ATP hydrolysis. However, the mechanism by which ATP hydrolysis in the V1 domain couples with the mechanical rotation of the rotor is still unclear. Cryo-EM snapshot analysis of V/A-ATPase indicated that three independent and simultaneous catalytic events occurred at the three catalytic dimers (ABopen, ABsemi, and ABclosed), leading to a 120° rotation of the central rotor. Besides the closing motion caused by ATP bound to ABopen, the hydrolysis of ATP bound to ABsemi drives the 120° step. Our recent time-resolved cryo-EM snapshot analysis provides further evidence for this model. This review aimed to provide a comprehensive overview of the structure and function of V/A-ATPase from a thermophilic bacterium, one of the most well-studied rotary ATPases to date.
Collapse
Affiliation(s)
- Ken Yokoyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kyoto, Japan
| |
Collapse
|
33
|
Kamiyama Y, Parkin D, Takano M. Torque generation mechanism in F o motor of ATP synthase elucidated by free-energy and Coulomb-energy landscapes along the c-ring rotation. Biochem Biophys Res Commun 2023; 651:56-61. [PMID: 36791499 DOI: 10.1016/j.bbrc.2023.01.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Fo portion of ATP synthase is a proton-motive rotary motor. The Coulombic attraction between the conserved acidic residues in the c-ring and the arginine in the a-subunit (aR) was early proposed to drive the c-ring rotation relative to the a-subunit, and has been actually observed in our previous molecular dynamics simulation with full atomistic description of Fo embedded in the membrane. In this study, to quantify the driving force, we conducted the umbrella sampling (US) and obtained the free-energy landscape for the c-ring rotation. We first show that the free-energy gradient toward the ATP-synthesis direction appears in the deprotonated state of cE. Using the sampled snapshots that cover a wide range of the rotational angle, we further analyzed the rotational-angle dependence of the hydration and the protonation states and obtained the Coulomb-energy landscapes with a focus on the cE-aR interaction. The results indicate that both the Coulombic solvation energy of cE and the interaction energy between cE and aR contribute to the torque generation for the c-ring rotation.
Collapse
Affiliation(s)
- Yukinari Kamiyama
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo, 169-8555, Japan
| | - Dan Parkin
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo, 169-8555, Japan
| | - Mitsunori Takano
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo, 169-8555, Japan; Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo, 169-8555, Japan.
| |
Collapse
|
34
|
Mackieh R, Al-Bakkar N, Kfoury M, Roufayel R, Sabatier JM, Fajloun Z. Inhibitors of ATP Synthase as New Antibacterial Candidates. Antibiotics (Basel) 2023; 12:antibiotics12040650. [PMID: 37107012 PMCID: PMC10135114 DOI: 10.3390/antibiotics12040650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
ATP, the power of all cellular functions, is constantly used and produced by cells. The enzyme called ATP synthase is the energy factory in all cells, which produces ATP by adding inorganic phosphate (Pi) to ADP. It is found in the inner, thylakoid and plasma membranes of mitochondria, chloroplasts and bacteria, respectively. Bacterial ATP synthases have been the subject of multiple studies for decades, since they can be genetically manipulated. With the emergence of antibiotic resistance, many combinations of antibiotics with other compounds that enhance the effect of these antibiotics have been proposed as approaches to limit the spread of antibiotic-resistant bacteria. ATP synthase inhibitors, such as resveratrol, venturicidin A, bedaquiline, tomatidine, piceatannol, oligomycin A and N,N-dicyclohexylcarbodiimide were the starting point of these combinations. However, each of these inhibitors target ATP synthase differently, and their co-administration with antibiotics increases the susceptibility of pathogenic bacteria. After a brief description of the structure and function of ATP synthase, we aim in this review to highlight therapeutic applications of the major bacterial ATP synthase inhibitors, including animal’s venoms, and to emphasize their importance in decreasing the activity of this enzyme and subsequently eradicating resistant bacteria as ATP synthase is their source of energy.
Collapse
|
35
|
F1·Fo ATP Synthase/ATPase: Contemporary View on Unidirectional Catalysis. Int J Mol Sci 2023; 24:ijms24065417. [PMID: 36982498 PMCID: PMC10049701 DOI: 10.3390/ijms24065417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
F1·Fo-ATP synthases/ATPases (F1·Fo) are molecular machines that couple either ATP synthesis from ADP and phosphate or ATP hydrolysis to the consumption or production of a transmembrane electrochemical gradient of protons. Currently, in view of the spread of drug-resistant disease-causing strains, there is an increasing interest in F1·Fo as new targets for antimicrobial drugs, in particular, anti-tuberculosis drugs, and inhibitors of these membrane proteins are being considered in this capacity. However, the specific drug search is hampered by the complex mechanism of regulation of F1·Fo in bacteria, in particular, in mycobacteria: the enzyme efficiently synthesizes ATP, but is not capable of ATP hydrolysis. In this review, we consider the current state of the problem of “unidirectional” F1·Fo catalysis found in a wide range of bacterial F1·Fo and enzymes from other organisms, the understanding of which will be useful for developing a strategy for the search for new drugs that selectively disrupt the energy production of bacterial cells.
Collapse
|
36
|
Akutsu H. Strategies for elucidation of the structure and function of the large membrane protein complex, F oF 1-ATP synthase, by nuclear magnetic resonance. Biophys Chem 2023; 296:106988. [PMID: 36898347 DOI: 10.1016/j.bpc.2023.106988] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023]
Abstract
Nuclear magnetic resonance (NMR) investigation of large membrane proteins requires well-focused questions and critical techniques. Here, research strategies for FoF1-ATP synthase, a membrane-embedded molecular motor, are reviewed, focusing on the β-subunit of F1-ATPase and c-subunit ring of the enzyme. Segmental isotope-labeling provided 89% assignment of the main chain NMR signals of thermophilic Bacillus (T)F1β-monomer. Upon nucleotide binding to Lys164, Asp252 was shown to switch its hydrogen-bonding partner from Lys164 to Thr165, inducing an open-to-closed bend motion of TF1β-subunit. This drives the rotational catalysis. The c-ring structure determined by solid-state NMR showed that cGlu56 and cAsn23 of the active site took a hydrogen-bonded closed conformation in membranes. In 505 kDa TFoF1, the specifically isotope-labeled cGlu56 and cAsn23 provided well-resolved NMR signals, which revealed that 87% of the residue pairs took a deprotonated open conformation at the Foa-c subunit interface, whereas they were in the closed conformation in the lipid-enclosed region.
Collapse
Affiliation(s)
- Hideo Akutsu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Japan; Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan.
| |
Collapse
|
37
|
Jackson PJ, Hitchcock A, Brindley AA, Dickman MJ, Hunter CN. Absolute quantification of cellular levels of photosynthesis-related proteins in Synechocystis sp. PCC 6803. PHOTOSYNTHESIS RESEARCH 2023; 155:219-245. [PMID: 36542271 PMCID: PMC9958174 DOI: 10.1007/s11120-022-00990-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
Quantifying cellular components is a basic and important step for understanding how a cell works, how it responds to environmental changes, and for re-engineering cells to produce valuable metabolites and increased biomass. We quantified proteins in the model cyanobacterium Synechocystis sp. PCC 6803 given the general importance of cyanobacteria for global photosynthesis, for synthetic biology and biotechnology research, and their ancestral relationship to the chloroplasts of plants. Four mass spectrometry methods were used to quantify cellular components involved in the biosynthesis of chlorophyll, carotenoid and bilin pigments, membrane assembly, the light reactions of photosynthesis, fixation of carbon dioxide and nitrogen, and hydrogen and sulfur metabolism. Components of biosynthetic pathways, such as those for chlorophyll or for photosystem II assembly, range between 1000 and 10,000 copies per cell, but can be tenfold higher for CO2 fixation enzymes. The most abundant subunits are those for photosystem I, with around 100,000 copies per cell, approximately 2 to fivefold higher than for photosystem II and ATP synthase, and 5-20 fold more than for the cytochrome b6f complex. Disparities between numbers of pathway enzymes, between components of electron transfer chains, and between subunits within complexes indicate possible control points for biosynthetic processes, bioenergetic reactions and for the assembly of multisubunit complexes.
Collapse
Affiliation(s)
- Philip J Jackson
- Plants, Photosynthesis and Soil, School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S1 3JD, UK.
| | - Andrew Hitchcock
- Plants, Photosynthesis and Soil, School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Amanda A Brindley
- Plants, Photosynthesis and Soil, School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, S1 3JD, UK
| | - C Neil Hunter
- Plants, Photosynthesis and Soil, School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
38
|
Parkin D, Takano M. Coulombic Organization in Membrane-Embedded Rotary Motor of ATP Synthase. J Phys Chem B 2023; 127:1552-1562. [PMID: 36734508 DOI: 10.1021/acs.jpcb.2c07875] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The electrochemical potential difference of protons across the membrane is used to synthesize ATP through the proton-motive rotatory motion of the membrane-embedded region of ATP synthase called Fo. In this study, we illuminate the unsolved proton-motive rotary mechanism of Fo on the basis of atomistic simulation with full description of protein, lipid, and water molecules, and highlight the underlying Coulombic design. We first show that a water channel is spontaneously formed at the interfacial region between the rotor (c-ring) and the stator (a-subunit). The observed water channel is a full channel penetrating the membrane, but a Coulomb barrier by a strictly conserved arginine of the a-subunit dominates at the midpoint of the full channel, preventing proton leakage. Our molecular dynamics simulation further demonstrates that the Coulomb attraction between the arginine and the essential glutamic acid of the c-subunit drives the c-ring rotation. We finally illustrate that the charge-state changes of the glutamic acids, enabled by the electrochemical potential difference of proton and the thermal motion, can produce unidirectional rotation of the c-ring.
Collapse
Affiliation(s)
- Dan Parkin
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo169-8555, Japan
| | - Mitsunori Takano
- Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-Ku, Tokyo169-8555, Japan.,Department of Pure and Applied Physics, Waseda University, 3-8-1 Okubo, Shinjuku-Ku, Tokyo169-8555, Japan
| |
Collapse
|
39
|
Rogers JR, Nikolényi G, AlQuraishi M. Growing ecosystem of deep learning methods for modeling protein-protein interactions. Protein Eng Des Sel 2023; 36:gzad023. [PMID: 38102755 DOI: 10.1093/protein/gzad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
Numerous cellular functions rely on protein-protein interactions. Efforts to comprehensively characterize them remain challenged however by the diversity of molecular recognition mechanisms employed within the proteome. Deep learning has emerged as a promising approach for tackling this problem by exploiting both experimental data and basic biophysical knowledge about protein interactions. Here, we review the growing ecosystem of deep learning methods for modeling protein interactions, highlighting the diversity of these biophysically informed models and their respective trade-offs. We discuss recent successes in using representation learning to capture complex features pertinent to predicting protein interactions and interaction sites, geometric deep learning to reason over protein structures and predict complex structures, and generative modeling to design de novo protein assemblies. We also outline some of the outstanding challenges and promising new directions. Opportunities abound to discover novel interactions, elucidate their physical mechanisms, and engineer binders to modulate their functions using deep learning and, ultimately, unravel how protein interactions orchestrate complex cellular behaviors.
Collapse
Affiliation(s)
- Julia R Rogers
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Gergő Nikolényi
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | | |
Collapse
|
40
|
Changes within the central stalk of E. coli F 1F o ATP synthase observed after addition of ATP. Commun Biol 2023; 6:26. [PMID: 36631659 PMCID: PMC9834311 DOI: 10.1038/s42003-023-04414-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
F1Fo ATP synthase functions as a biological generator and makes a major contribution to cellular energy production. Proton flow generates rotation in the Fo motor that is transferred to the F1 motor to catalyze ATP production, with flexible F1/Fo coupling required for efficient catalysis. F1Fo ATP synthase can also operate in reverse, hydrolyzing ATP and pumping protons, and in bacteria this function can be regulated by an inhibitory ε subunit. Here we present cryo-EM data showing E. coli F1Fo ATP synthase in different rotational and inhibited sub-states, observed following incubation with 10 mM MgATP. Our structures demonstrate how structural transitions within the inhibitory ε subunit induce torsional movement in the central stalk, thereby enabling its rotation within the Fο motor. This highlights the importance of the central rotor for flexible coupling of the F1 and Fo motors and provides further insight into the regulatory mechanism mediated by subunit ε.
Collapse
|
41
|
Krah A, Vogelaar T, de Jong SI, Claridge JK, Bond PJ, McMillan DGG. ATP binding by an F 1F o ATP synthase ε subunit is pH dependent, suggesting a diversity of ε subunit functional regulation in bacteria. Front Mol Biosci 2023; 10:1059673. [PMID: 36923639 PMCID: PMC10010621 DOI: 10.3389/fmolb.2023.1059673] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/03/2023] [Indexed: 03/03/2023] Open
Abstract
It is a conjecture that the ε subunit regulates ATP hydrolytic function of the F1Fo ATP synthase in bacteria. This has been proposed by the ε subunit taking an extended conformation, with a terminal helix probing into the central architecture of the hexameric catalytic domain, preventing ATP hydrolysis. The ε subunit takes a contracted conformation when bound to ATP, thus would not interfere with catalysis. A recent crystallographic study has disputed this; the Caldalkalibacillus thermarum TA2.A1 F1Fo ATP synthase cannot natively hydrolyse ATP, yet studies have demonstrated that the loss of the ε subunit terminal helix results in an ATP synthase capable of ATP hydrolysis, supporting ε subunit function. Analysis of sequence and crystallographic data of the C. thermarum F1Fo ATP synthase revealed two unique histidine residues. Molecular dynamics simulations suggested that the protonation state of these residues may influence ATP binding site stability. Yet these residues lie outside the ATP/Mg2+ binding site of the ε subunit. We then probed the effect of pH on the ATP binding affinity of the ε subunit from the C. thermarum F1Fo ATP synthase at various physiologically relevant pH values. We show that binding affinity changes 5.9 fold between pH 7.0, where binding is weakest, to pH 8.5 where it is strongest. Since the C. thermarum cytoplasm is pH 8.0 when it grows optimally, this correlates to the ε subunit being down due to ATP/Mg2+ affinity, and not being involved in blocking ATP hydrolysis. Here, we have experimentally correlated that the pH of the bacterial cytoplasm is of critical importance for ε subunit ATP affinity regulated by second-shell residues thus the function of the ε subunit changes with growth conditions.
Collapse
Affiliation(s)
- Alexander Krah
- Korea Institute for Advanced Study, School of Computational Sciences, Seoul, South Korea.,Bioinformatics Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Timothy Vogelaar
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Sam I de Jong
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Jolyon K Claridge
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| | - Peter J Bond
- Bioinformatics Institute, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Duncan G G McMillan
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands.,School of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| |
Collapse
|
42
|
Mencía M. Acid digestion and symbiont: Proton sharing at the origin of mitochondriogenesis?: Proton production by a symbiotic bacterium may have been the origin of two hallmark eukaryotic features, acid digestion and mitochondria: Proton production by a symbiotic bacterium may have been the origin of two hallmark eukaryotic features, acid digestion and mitochondria. Bioessays 2023; 45:e2200136. [PMID: 36373631 DOI: 10.1002/bies.202200136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
The initial relationships between organisms leading to endosymbiosis and the first eukaryote are currently a topic of hot debate. Here, I present a theory that offers a gradual scenario in which the origins of phagocytosis and mitochondria are intertwined in such a way that the evolution of one would not be possible without the other. In this scenario, the premitochondrial bacterial symbiont became initially associated with a protophagocytic host on the basis of cooperation to kill prey with symbiont-produced toxins and reactive oxygen species (ROS). Subsequently, the cooperation was focused on the digestion stage, through the acidification of the protophagocytic cavities via exportation of protons produced by the aerobic respiration of the symbiont. The host gained an improved phagocytic capacity and the symbiont received organic compounds from prey. As the host gradually lost its membrane energetics to develop lysosomal digestion, respiration was centralized in the premitochondrial symbiont for energy production for the consortium.
Collapse
Affiliation(s)
- Mario Mencía
- Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (CBMSO) UAM-CSIC, Universidad Autónoma de Madrid, Madrid, 28409, Spain
| |
Collapse
|
43
|
Nesci S. Bacterial and mammalian F1FO-ATPase: Structural similarities and divergences to exploit in the battle against Mycobacterium tuberculosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023. [DOI: 10.1016/bs.ircmb.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
44
|
Otomo A, Iida T, Okuni Y, Ueno H, Murata T, Iino R. Direct observation of stepping rotation of V-ATPase reveals rigid component in coupling between V o and V 1 motors. Proc Natl Acad Sci U S A 2022; 119:e2210204119. [PMID: 36215468 PMCID: PMC9586324 DOI: 10.1073/pnas.2210204119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/13/2022] [Indexed: 11/30/2022] Open
Abstract
V-ATPases are rotary motor proteins that convert the chemical energy of ATP into the electrochemical potential of ions across cell membranes. V-ATPases consist of two rotary motors, Vo and V1, and Enterococcus hirae V-ATPase (EhVoV1) actively transports Na+ in Vo (EhVo) by using torque generated by ATP hydrolysis in V1 (EhV1). Here, we observed ATP-driven stepping rotation of detergent-solubilized EhVoV1 wild-type, aE634A, and BR350K mutants under various Na+ and ATP concentrations ([Na+] and [ATP], respectively) by using a 40-nm gold nanoparticle as a low-load probe. When [Na+] was low and [ATP] was high, under the condition that only Na+ binding to EhVo is rate limiting, wild-type and aE634A exhibited 10 pausing positions reflecting 10-fold symmetry of the EhVo rotor and almost no backward steps. Duration time before the forward steps was inversely proportional to [Na+], confirming that Na+ binding triggers the steps. When both [ATP] and [Na+] were low, under the condition that both Na+ and ATP bindings are rate limiting, aE634A exhibited 13 pausing positions reflecting 10- and 3-fold symmetries of EhVo and EhV1, respectively. The distribution of duration time before the forward step was fitted well by the sum of two exponential decay functions with distinct time constants. Furthermore, occasional backward steps smaller than 36° were observed. Small backward steps were also observed during three long ATP cleavage pauses of BR350K. These results indicate that EhVo and EhV1 do not share pausing positions, Na+ and ATP bindings occur at different angles, and the coupling between EhVo and EhV1 has a rigid component.
Collapse
Affiliation(s)
- Akihiro Otomo
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Functional Molecular Science, School of Physical Sciences, Graduate University for Advanced Studies, Hayama 240-0193, Japan
| | - Tatsuya Iida
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Functional Molecular Science, School of Physical Sciences, Graduate University for Advanced Studies, Hayama 240-0193, Japan
| | - Yasuko Okuni
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
| | - Hiroshi Ueno
- Department of Applied Chemistry, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Takeshi Murata
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Ryota Iino
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Functional Molecular Science, School of Physical Sciences, Graduate University for Advanced Studies, Hayama 240-0193, Japan
| |
Collapse
|
45
|
Iwamoto-Kihara A. Regulatory Mechanisms and Environmental Adaptation of the F-ATPase Family. Biol Pharm Bull 2022; 45:1412-1418. [PMID: 36184497 DOI: 10.1248/bpb.b22-00419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The F-type ATPase family of enzymes, including ATP synthases, are found ubiquitously in biological membranes. ATP synthesis from ADP and inorganic phosphate is driven by an electrochemical H+ gradient or H+ motive force, in which intramolecular rotation of F-type ATPase is generated with H+ transport across the membranes. Because this rotation is essential for energy coupling between catalysis and H+-transport, regulation of the rotation is important to adapt to environmental changes and maintain ATP concentration. Recently, a series of cryo-electron microscopy images provided detailed insights into the structure of the H+ pathway and the multiple subunit arrangement. However, the regulatory mechanism of the rotation has not been clarified. This review describes the inhibition mechanism of ATP hydrolysis in bacterial enzymes. In addition, properties of the F-type ATPase of Streptococcus mutans, which acts as a H+-pump in an acidic environment, are described. These findings may help in the development of novel antimicrobial agents.
Collapse
|
46
|
Wu R, Liu T, Wu S, Li H, Song R, Song B. Synthesis, Antibacterial Activity, and Action Mechanism of Novel Sulfonamides Containing Oxyacetal and Pyrimidine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9305-9318. [PMID: 35858046 DOI: 10.1021/acs.jafc.2c02099] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bacterial leaf blight (BLB) and bacterial leaf streak (BLS) are two serious bacterial diseases caused by Xanthomonas oryzae pv. oryzae (Xoo) and Xanthomonas oryzae pv. oryzicola (Xoc), respectively. However, the control of these diseases by conventional pesticides remains challenging due to development of resistances. We aimed to address this pending problem and developed a series of novel pyrimidine sulfonamide derivatives. Structurally, title compounds bear a unique oxyacetal group, which has a proven immune-activating effect. Compound E35 designed based on the 3D-QSAR model was demonstrated as the optimal in vitro activity against Xoo and Xoc, with EC50 values of 26.7 and 30.8 mg/L, respectively, which were higher than the positive controls bismerthiazol (29.9 and 32.7 mg/L) and thiodiazole copper (30.5 and 36.4 mg/L). On the prevention level, the biological activity test showed compound E35 had superior protective activity (43.7%) on BLS to thiodiazole copper (32.1%). The defense enzymes and proteomics results suggested that compound E35 could be a versatile candidate as it improved plant's resistance to disease.
Collapse
Affiliation(s)
- Rong Wu
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Ting Liu
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Sikai Wu
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Hongde Li
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Runjiang Song
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Baoan Song
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| |
Collapse
|
47
|
Zharova TV, Kozlovsky VS, Grivennikova VG. Interaction of Venturicidin and F o·F 1-ATPase/ATP Synthase of Tightly Coupled Subbacterial Particles of Paracoccus denitrificans in Energized Membranes. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:742-751. [PMID: 36171655 DOI: 10.1134/s0006297922080065] [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: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 06/16/2023]
Abstract
Proton-translocating Fo×F1-ATPase/synthase that catalyzes synthesis and hydrolysis of ATP is commonly considered to be a reversibly functioning complex. We have previously shown that venturicidin, a specific Fo-directed inhibitor, blocks the synthesis and hydrolysis of ATP with a significant difference in the affinity [Zharova, T. V. and Vinogradov, A. D. (2017) Biochim. Biophys. Acta, 1858, 939-944]. In this paper, we have studied in detail inhibition of Fo×F1-ATPase/synthase by venturicidin in tightly coupled membranes of Paracoccus denitrificans under conditions of membrane potential generation. ATP hydrolysis was followed by the ATP-dependent succinate-supported NAD+ reduction (potential-dependent reverse electron transfer) catalyzed by the respiratory chain complex I. It has been demonstrated that membrane energization did not affect the affinity of Fo×F1-ATPase/synthase for venturicidin. The dependence of the residual ATP synthase activity on the concentration of venturicidin approximated a linear function, whereas the dependence of ATP hydrolysis was sigmoidal: at low inhibitor concentrations venturicidin strongly inhibited ATP synthesis without decrease in the rate of ATP hydrolysis. A model is proposed suggesting that ATP synthesis and ATP hydrolysis are catalyzed by two different forms of Fo×F1.
Collapse
Affiliation(s)
- Tatyana V Zharova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Vladimir S Kozlovsky
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Vera G Grivennikova
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
48
|
Miranda-Astudillo H, Ostolga-Chavarría M, Cardol P, González-Halphen D. Beyond being an energy supplier, ATP synthase is a sculptor of mitochondrial cristae. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148569. [PMID: 35577152 DOI: 10.1016/j.bbabio.2022.148569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Mitochondrial F1FO-ATP synthase plays a key role in cellular bioenergetics; this enzyme is present in all eukaryotic linages except in amitochondriate organisms. Despite its ancestral origin, traceable to the alpha proteobacterial endosymbiotic event, the actual structural diversity of these complexes, due to large differences in their polypeptide composition, reflects an important evolutionary divergence between eukaryotic lineages. We discuss the effect of these structural differences on the oligomerization of the complex and the shape of mitochondrial cristae.
Collapse
Affiliation(s)
- Héctor Miranda-Astudillo
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcos Ostolga-Chavarría
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Pierre Cardol
- InBios/Phytosystems, Institut de Botanique, Université de Liège, Liège, Belgium
| | - Diego González-Halphen
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
49
|
Marshansky V. Discovery and Study of Transmembrane Rotary Ion-Translocating Nano-Motors: F-ATPase/Synthase of Mitochondria/Bacteria and V-ATPase of Eukaryotic Cells. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:702-719. [PMID: 36171652 DOI: 10.1134/s000629792208003x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 06/16/2023]
Abstract
This review discusses the history of discovery and study of the operation of the two rotary ion-translocating ATPase nano-motors: (i) F-ATPase/synthase (holocomplex F1FO) of mitochondria/bacteria and (ii) eukaryotic V-ATPase (holocomplex V1VO). Vacuolar adenosine triphosphatase (V-ATPase) is a transmembrane multisubunit complex found in all eukaryotes from yeast to humans. It is structurally and functionally similar to the F-ATPase/synthase of mitochondria/bacteria and the A-ATPase/synthase of archaebacteria, which indicates a common evolutionary origin of the rotary ion-translocating nano-motors built into cell membranes and invented by Nature billions of years ago. Previously we have published several reviews on this topic with appropriate citations of our original research. This review is focused on the historical analysis of the discovery and study of transmembrane rotary ion-translocating ATPase nano-motors functioning in bacteria, eukaryotic cells and mitochondria of animals.
Collapse
|
50
|
Todokoro Y, Kang SJ, Suzuki T, Ikegami T, Kainosho M, Yoshida M, Fujiwara T, Akutsu H. Chemical Conformation of the Essential Glutamate Site of the c-Ring within Thermophilic Bacillus F oF 1-ATP Synthase Determined by Solid-State NMR Based on its Isolated c-Ring Structure. J Am Chem Soc 2022; 144:14132-14139. [PMID: 35905443 DOI: 10.1021/jacs.2c03580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Proton translocation through the membrane-embedded Fo component of F-type ATP synthase (FoF1) is facilitated by the rotation of the Fo c-subunit ring (c-ring), carrying protons at essential acidic amino acid residues. Cryo-electron microscopy (Cryo-EM) structures of FoF1 suggest a unique proton translocation mechanism. To elucidate it based on the chemical conformation of the essential acidic residues of the c-ring in FoF1, we determined the structure of the isolated thermophilic Bacillus Fo (tFo) c-ring, consisting of 10 subunits, in membranes by solid-state NMR. This structure contains a distinct proton-locking conformation, wherein Asn23 (cN23) CγO and Glu56 (cE56) CδOH form a hydrogen bond in a closed form. We introduced stereo-array-isotope-labeled (SAIL) Glu and Asn into the tFoc-ring to clarify the chemical conformation of these residues in tFoF1-ATP synthase (tFoF1). Two well-separated 13C signals could be detected for cN23 and cE56 in a 505 kDa membrane protein complex, respectively, thereby suggesting the presence of two distinct chemical conformations. Based on the signal intensity and structure of the tFoc-ring and tFoF1, six pairs of cN23 and cE56 surrounded by membrane lipids take the closed form, whereas the other four in the a-c interface employ the deprotonated open form at a proportion of 87%. This indicates that the a-c interface is highly hydrophilic. The pKa values of the four cE56 residues in the a-c interface were estimated from the cN23 signal intensity in the open and closed forms and distribution of polar residues around each cE56. The results favor a rotation of the c-ring for ATP synthesis.
Collapse
Affiliation(s)
- Yasuto Todokoro
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Japan.,Technical Support Division, School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka 560-0043, Japan
| | - Su-Jin Kang
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Japan.,Department of Biophysics and Chemical Biology, Seoul National University, Gwanak-Gu, Seoul 151-742, Republic of Korea.,College of Pharmacy, Dongduk Women's University, Seoul 02748, Republic of Korea
| | - Toshiharu Suzuki
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Midori-ku, Yokohama 226-0026, Japan
| | - Takahisa Ikegami
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Masatsune Kainosho
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo 192-0397, Japan
| | - Masasuke Yoshida
- Department of Molecular Bioscience, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto 603-8555, Japan
| | - Toshimichi Fujiwara
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Japan
| | - Hideo Akutsu
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita 565-0871, Japan.,Department of Biophysics and Chemical Biology, Seoul National University, Gwanak-Gu, Seoul 151-742, Republic of Korea.,Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|