1
|
Palmai Z. Sucrose and Gibberellic Acid Binding Stabilize the Inward-Open Conformation of AtSWEET13: A Molecular Dynamics Study. Proteins 2025; 93:1141-1156. [PMID: 39815685 DOI: 10.1002/prot.26799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
In plants, sugar will eventually be exported transporters (SWEETs) facilitate the translocation of mono- and disaccharides across membranes and play a critical role in modulating responses to gibberellin (GA3), a key growth hormone. However, the dynamic mechanisms underlying sucrose and GA3 binding and transport remain elusive. Here, we employed microsecond-scale molecular dynamics (MD) simulations to investigate the influence of sucrose and GA3 binding on SWEET13 transporter motions. While sucrose exhibits high flexibility within the binding pocket, GA3 remains firmly anchored in the central cavity. Binding of both ligands increases the average channel radius along the transporter's principal axis. In contrast to the apo form, which retains its initial conformation throughout the simulation, ligand-bound complexes undergo a significant conformational transition characterized by further opening of the intracellular gate relative to the inward-open crystal structure (5XPD). This opening is driven by ligand-induced bending of helix V, stabilizing the inward-open state. Sucrose binding notably enhances the flexibility of the intracellular gate and amplifies anticorrelated motions between the N- and C-terminal domains at the intracellular side, suggesting an opening-closing motion of these domains. Principal component analysis revealed that this gating motion is most pronounced in the sucrose complex and minimal in the apo form, highlighting sucrose's ability to induce high-amplitude gating. Our binding free energy calculations indicate that SWEET13 has lower binding affinity for sucrose compared to GA3, consistent with its role in sugar transport. These results provide insight into key residues involved in sucrose and GA3 binding and transport, advancing our understanding of SWEET13 dynamics.
Collapse
Affiliation(s)
- Zoltan Palmai
- Institute of Transformative bio-Molecules, Nagoya University, Nagoya, Japan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
2
|
Molina Panadero I, Falcón Torres J, Hmadcha A, Princiotto S, Cutarella L, Mori M, Dallavalle S, Christodoulou MS, Smani Y. Antibacterial activity of tamoxifen derivatives against methicillin-resistant Staphylococcus aureus. Front Pharmacol 2025; 16:1549288. [PMID: 40371342 PMCID: PMC12075203 DOI: 10.3389/fphar.2025.1549288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
The development of new antimicrobial therapeutic strategies requires urgent attention to prevent the tens of millions of deaths predicted to occur by 2050 as a result of multidrug-resistant (MDR) bacterial infections. This study aimed to discover new tamoxifen derivatives with antimicrobial potential, particularly targeting methicillin-resistant Staphylococcus aureus (MRSA). The minimum inhibitory concentration (MIC) of 22 tamoxifen derivatives was determined against S. aureus reference and MRSA strains using microdilution assays. The antibacterial effects of selected tamoxifen derivatives against MRSA (USA7 strain) were assessed through bacterial growth assays. Additionally, bacterial membrane permeability and molecular dynamics (MD) simulation assays were performed. The MIC of the tamoxifen derivatives against reference S. aureus and MRSA strains ranged from to 16 to >64 μg/mL. Bacterial growth assays demonstrated that tamoxifen derivatives 2, 5, and 6, the only compounds bearing the electron-donating hydroxyl group in the para position on both phenyl rings of the tamoxifen skeleton, dose-dependently reduced the growth of the USA7 strain. Moreover, treatment of MRSA with derivatives 2 and 5 resulted in a slight increase of membrane permeabilization. Extensive MD simulations on the interaction between 5 and 6 and the S. aureus membrane model suggest that the compounds do not act by destabilizing the membrane integrity. These findings suggest that tamoxifen derivatives exhibit antibacterial activity against MRSA, potentially broadening the spectrum of available drug treatments for combating antimicrobial-resistant S. aureus.
Collapse
Affiliation(s)
- Irene Molina Panadero
- Andalusian Center of Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), University of Pablo de Olavide - Seville, Seville, Spain
| | - Javier Falcón Torres
- Andalusian Center of Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), University of Pablo de Olavide - Seville, Seville, Spain
| | - Abdelkrim Hmadcha
- Department of Molecular Biology and Biochemical Engineering, University of Pablo de Olavide, Seville, Spain
- Biosanitary Research Institute (IIB-VIU), Valencian International University (VIU), Valencia, Spain
| | - Salvatore Princiotto
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Luigi Cutarella
- Department f Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Mattia Mori
- Department f Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Michael S. Christodoulou
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Younes Smani
- Andalusian Center of Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), University of Pablo de Olavide - Seville, Seville, Spain
- Department of Molecular Biology and Biochemical Engineering, University of Pablo de Olavide, Seville, Spain
| |
Collapse
|
3
|
Santarossa CC, Li Y, Yousef S, Hasdemir HS, Rodriguez CC, Haase MB, Baek M, Coudray N, Pavek JG, Focke KN, Silverberg AL, Bautista C, Yeh J, Marty MT, Baker D, Tajkhorshid E, Ekiert DC, Bhabha G. LetA defines a structurally distinct transporter family involved in lipid trafficking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644421. [PMID: 40166208 PMCID: PMC11957124 DOI: 10.1101/2025.03.21.644421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Membrane transport proteins translocate diverse cargos, ranging from small sugars to entire proteins, across cellular membranes. A few structurally distinct protein families have been described that account for most of the known membrane transport processes. However, many membrane proteins with predicted transporter functions remain uncharacterized. We determined the structure of E. coli LetAB, a phospholipid transporter involved in outer membrane integrity, and found that LetA adopts a distinct architecture that is structurally and evolutionarily unrelated to known transporter families. LetA functions as a pump at one end of a ~225 Å long tunnel formed by its binding partner, MCE protein LetB, creating a pathway for lipid transport between the inner and outer membranes. Unexpectedly, the LetA transmembrane domains adopt a fold that is evolutionarily related to the eukaryotic tetraspanin family of membrane proteins, including TARPs and claudins. LetA has no detectable homology to known transport proteins, and defines a new class of membrane transporters. Through a combination of deep mutational scanning, molecular dynamics simulations, AlphaFold-predicted alternative states, and functional studies, we present a model for how the LetA-like family of membrane transporters may use energy from the proton-motive force to drive the transport of lipids across the bacterial cell envelope.
Collapse
|
4
|
Niu GH, Hsiao WC, Lee PH, Zheng LG, Yang YS, Huang WC, Hsieh CC, Chiu TY, Wang JY, Chen CP, Huang CL, You MS, Kuo YP, Wang CM, Wen ZH, Yu GY, Chen CT, Chi YH, Tung CW, Hsu SC, Yeh TK, Sung PJ, Zhang MM, Tsou LK. Orally Bioavailable and Site-Selective Covalent STING Inhibitor Derived from a Macrocyclic Marine Diterpenoid. J Med Chem 2025; 68:5471-5487. [PMID: 40014799 PMCID: PMC11912488 DOI: 10.1021/acs.jmedchem.4c02665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Pharmacological inhibition of the cGAS-STING-controlled innate immune pathway is an emerging therapeutic strategy for a myriad of inflammatory diseases. Here, we report GHN105 as an orally bioavailable covalent STING inhibitor. Late-stage diversification of the briarane-type diterpenoid excavatolide B allowed the installation of solubility-enhancing functional groups while enhancing its activity as a covalent STING inhibitor against multiple human STING variants, including the S154 variant responsible for a genetic autoimmune disease. Selectively engaging the membrane-proximal Cys91 residue of STING, GHN105 dose-dependently inhibited cGAS-STING signaling and type I interferon responses in cells and in vivo. Moreover, orally administered GHN105 exhibited on-target engagement in vivo and markedly reversed key pathological features in a delayed treatment of the acute colitis mouse model. Our study provided proof of concept that the synthetic briarane analog GHN105 serves as a safe, site-selective, and orally active covalent STING inhibitor and devises a regimen that allows long-term systemic administration.
Collapse
Affiliation(s)
- Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wan-Chi Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Po-Hsun Lee
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Li-Guo Zheng
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Yu-Shao Yang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chih-Chien Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - May-Su You
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Yi-Ping Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chien-Ming Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung 804201, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Shu-Ching Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 944401, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| |
Collapse
|
5
|
Yegnaswamy S, C SK, Aldaais E. Conformational dynamics of the membrane protein of MERS-CoV in comparison with SARS-CoV-2 in ERGIC complex. J Biomol Struct Dyn 2025:1-15. [PMID: 39755960 DOI: 10.1080/07391102.2024.2437529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/21/2024] [Indexed: 01/07/2025]
Abstract
The present study explores the conformational dynamics of the membrane protein of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) within the Endoplasmic Reticulum-Golgi Intermediate Compartment (ERGIC) complex using an all-atomistic molecular dynamics simulation approach. Significant structural changes were observed in the N-terminal, C-terminal, transmembrane, and beta-sheet sandwich domains of the MERS-CoV membrane protein. This study also highlights the structural similarities between the MERS-CoV and the SARS-CoV-2 membrane proteins, particularly in how both exhibit a distinct kink in the transmembrane helix caused by aromatic residue-lipid interactions. A structural expansion below the transmembrane and above the beta-sheet sandwich domain within the dimer was observed in all the M-proteins. This site on the beta-sheet sandwich domains near the C-terminal end could serve as a potential drug-binding site. Notably, a stable helical structure was identified in the C-terminal domain of the MERS-CoV membrane protein, whereas a proper secondary structural conformation was not observed in the SARS-CoV-2 membrane protein. Further, the SARS-CoV-2 membrane protein exhibited stronger binding to the lipid bilayer than the MERS-CoV, indicating its greater structural stability within the ERGIC complex. The structural similarity between the membrane protein of MERS-CoV and SARS-CoV-2 suggests the feasibility of employing a common inhibitor against these beta-coronaviruses. Furthermore, this analysis enhances our understanding of the membrane protein's interactions with proteins and lipids, paving the way for therapeutic developments against these viruses.
Collapse
Affiliation(s)
- Subha Yegnaswamy
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Selvaa Kumar C
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Ebtisam Aldaais
- College of Applied Medical Sciences, lmam Abdulrahman Bin Faisal University (lAU), Dammam, Saudi Arabia
| |
Collapse
|
6
|
Giangregorio N, Tonazzi A, Pierri CL, Indiveri C. Insights into Transient Dimerisation of Carnitine/Acylcarnitine Carrier (SLC25A20) from Sarkosyl/PAGE, Cross-Linking Reagents, and Comparative Modelling Analysis. Biomolecules 2024; 14:1158. [PMID: 39334924 PMCID: PMC11430254 DOI: 10.3390/biom14091158] [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: 07/16/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
The carnitine/acylcarnitine carrier (CAC) is a crucial protein for cellular energy metabolism, facilitating the exchange of acylcarnitines and free carnitine across the mitochondrial membrane, thereby enabling fatty acid β-oxidation and oxidative phosphorylation (OXPHOS). Although CAC has not been crystallised, structural insights are derived from the mitochondrial ADP/ATP carrier (AAC) structures in both cytosolic and matrix conformations. These structures underpin a single binding centre-gated pore mechanism, a common feature among mitochondrial carrier (MC) family members. The functional implications of this mechanism are well-supported, yet the structural organization of the CAC, particularly the formation of dimeric or oligomeric assemblies, remains contentious. Recent investigations employing biochemical techniques on purified and reconstituted CAC, alongside molecular modelling based on crystallographic AAC dimeric structures, suggest that CAC can indeed form dimers. Importantly, this dimerization does not alter the transport mechanism, a phenomenon observed in various other membrane transporters across different protein families. This observation aligns with the ping-pong kinetic model, where the dimeric form potentially facilitates efficient substrate translocation without necessitating mechanistic alterations. The presented findings thus contribute to a deeper understanding of CAC's functional dynamics and its structural parallels with other MC family members.
Collapse
Affiliation(s)
- Nicola Giangregorio
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126 Bari, Italy
| | - Annamaria Tonazzi
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126 Bari, Italy
| | - Ciro Leonardo Pierri
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari, Via E. Orabona, 4, 70125 Bari, Italy
| | - Cesare Indiveri
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Via Amendola 122/O, 70126 Bari, Italy
- Department DiBEST (Biologia, Ecologia, Scienze della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, Via Bucci 4C, 87036 Arcavacata di Rende, Italy
| |
Collapse
|
7
|
Ceccarelli M, Milenkovic S, Bodrenko IV. The Effect of Lipopolysaccharides on the Electrostatic Properties of Gram-Negative General Porins from Enterobacteriaceae. Chemphyschem 2024; 25:e202400147. [PMID: 38625051 DOI: 10.1002/cphc.202400147] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/17/2024]
Abstract
We investigated, by using all-atom molecular dynamics simulations, the effect of the outer membrane of Gram-negative bacteria, composed in the outer leaflet by polar/charged lipopolysaccharides (LPS), on the electrostatic properties of general porins from the Enterobacteriaceae family. General porins constitute the main path for the facilitated diffusion of polar antibiotics through the outer membrane. As model system we selected OmpK36 from Klebsiella pneumoniae, the ortholog of OmpC from Escherichia coli. This species presents high variability of amino acid composition of porins, with the effect to increase its resistance to the penetration of antibiotics. The various properties we analyzed seem to indicate that LPS acts as an independent layer without affecting the internal electrostatic properties of OmpK36. The only apparent effect on the microsecond time scale we sampled is the appearance of calcium ions, when present at moderate concentration in solution, inside the pore. However, we noticed increased fluctuations of the polarization density and only minor changes on its average value.
Collapse
Affiliation(s)
- Matteo Ceccarelli
- Department of Physics, University of Cagliari, Cittadella Universitaria, 09042, Monserrato, IT
| | - Stefan Milenkovic
- Department of Physics, University of Cagliari, Cittadella Universitaria, 09042, Monserrato, IT
| | - Igor V Bodrenko
- Istituto Nanoscienze, CNR, piazza San Silvestro 12, 56127, Pisa, Italy
- Lab NEST, Scuola Normale Superiore, piazza San Silvestro 12, 56127, Pisa, Italy
| |
Collapse
|
8
|
Chen J, Ding Y, Jiang C, Qu R, Wren JD, Georgescu C, Wang X, Reuter DN, Liu B, Giles CB, Mayr CH, Schiller HB, Dai J, Stipp CS, Subramaniyan B, Wang J, Zuo H, Huang C, Fung KM, Rice HC, Sonnenberg A, Wu D, Walters MS, Zhao YY, Kanie T, Hays FA, Papin JF, Wang DW, Zhang XA. CD151 Maintains Endolysosomal Protein Quality to Inhibit Vascular Inflammation. Circ Res 2024; 134:1330-1347. [PMID: 38557119 PMCID: PMC11081830 DOI: 10.1161/circresaha.123.323190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Tetraspanin CD151 is highly expressed in endothelia and reinforces cell adhesion, but its role in vascular inflammation remains largely unknown. METHODS In vitro molecular and cellular biological analyses on genetically modified endothelial cells, in vivo vascular biological analyses on genetically engineered mouse models, and in silico systems biology and bioinformatics analyses on CD151-related events. RESULTS Endothelial ablation of Cd151 leads to pulmonary and cardiac inflammation, severe sepsis, and perilous COVID-19, and endothelial CD151 becomes downregulated in inflammation. Mechanistically, CD151 restrains endothelial release of proinflammatory molecules for less leukocyte infiltration. At the subcellular level, CD151 determines the integrity of multivesicular bodies/lysosomes and confines the production of exosomes that carry cytokines such as ANGPT2 (angiopoietin-2) and proteases such as cathepsin-D. At the molecular level, CD151 docks VCP (valosin-containing protein)/p97, which controls protein quality via mediating deubiquitination for proteolytic degradation, onto endolysosomes to facilitate VCP/p97 function. At the endolysosome membrane, CD151 links VCP/p97 to (1) IFITM3 (interferon-induced transmembrane protein 3), which regulates multivesicular body functions, to restrain IFITM3-mediated exosomal sorting, and (2) V-ATPase, which dictates endolysosome pH, to support functional assembly of V-ATPase. CONCLUSIONS Distinct from its canonical function in strengthening cell adhesion at cell surface, CD151 maintains endolysosome function by sustaining VCP/p97-mediated protein unfolding and turnover. By supporting protein quality control and protein degradation, CD151 prevents proteins from (1) buildup in endolysosomes and (2) discharge through exosomes, to limit vascular inflammation. Also, our study conceptualizes that balance between degradation and discharge of proteins in endothelial cells determines vascular information. Thus, the IFITM3/V-ATPase-tetraspanin-VCP/p97 complexes on endolysosome, as a protein quality control and inflammation-inhibitory machinery, could be beneficial for therapeutic intervention against vascular inflammation.
Collapse
Affiliation(s)
- Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Chao Jiang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Rongmei Qu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | | | - Xuejun Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Beibei Liu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Cory B. Giles
- Oklahoma Medical Research Foundation, Oklahoma City, USA
| | | | | | - Jingxing Dai
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | | | - Jie Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Houjuan Zuo
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Chao Huang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Kar-Ming Fung
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Heather C. Rice
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - David Wu
- University of Chicago, Chicago, IL, USA
| | | | - You-Yang Zhao
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tomoharu Kanie
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Franklin A. Hays
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - James F. Papin
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Dao Wen Wang
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xin A. Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
- Lead contact
| |
Collapse
|
9
|
Ardino C, Sannio F, Poli G, Galati S, Dreassi E, Botta L, Docquier JD, D'Agostino I. An update on antibacterial AlkylGuanidino Ureas: Design of new derivatives, synergism with colistin and data analysis of the whole library. Eur J Med Chem 2024; 270:116362. [PMID: 38574637 DOI: 10.1016/j.ejmech.2024.116362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
Antimicrobial resistance (AMR) represents one of the most challenging global Public Health issues, with an alarmingly increasing rate of attributable mortality. This scenario highlights the urgent need for innovative medicinal strategies showing activity on resistant isolates (especially, carbapenem-resistant Gram-negative bacteria, methicillin-resistant S. aureus, and vancomycin-resistant enterococci) yielding new approaches for the treatment of bacterial infections. We previously reported AlkylGuanidino Ureas (AGUs) with broad-spectrum antibacterial activity and a putative membrane-based mechanism of action. Herein, new tetra- and mono-guanidino derivatives were designed and synthesized to expand the structure-activity relationships (SARs) and, thereby, tested on the same panel of Gram-positive and Gram-negative bacteria. The membrane-active mechanism of selected compounds was then investigated through molecular dynamics (MD) on simulated bacterial membranes. In the end, the newly synthesized series, along with the whole library of compounds (more than 70) developed in the last decade, was tested in combination with subinhibitory concentrations of the last resort antibiotic colistin to assess putative synergistic or additive effects. Moreover, all the AGUs were subjected to cheminformatic and machine learning analyses to gain a deeper knowledge of the key features required for bioactivity.
Collapse
Affiliation(s)
- Claudia Ardino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Filomena Sannio
- Department of Medical Biotechnologies, University of Siena, Viale Mario Bracci 16, I-53100, Siena, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, I-56126, Pisa, Italy
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, I-56126, Pisa, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy
| | - Lorenzo Botta
- Lead Discovery Siena s.r.l., Via Vittorio Alfieri 31, I-53019, Castelnuovo Berardenga, Italy; Department of Ecological and Biological Sciences, University of Tuscia, Largo dell'Università snc, I-01100, Viterbo, Italy
| | - Jean-Denis Docquier
- Department of Medical Biotechnologies, University of Siena, Viale Mario Bracci 16, I-53100, Siena, Italy; Lead Discovery Siena s.r.l., Via Vittorio Alfieri 31, I-53019, Castelnuovo Berardenga, Italy
| | - Ilaria D'Agostino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via Aldo Moro 2, I-53100, Siena, Italy; Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, I-56126, Pisa, Italy.
| |
Collapse
|
10
|
Princiotto S, Casciaro B, G Temprano A, Musso L, Sacchi F, Loffredo MR, Cappiello F, Sacco F, Raponi G, Fernandez VP, Iucci T, Mangoni ML, Mori M, Dallavalle S, Pisano C. The antimicrobial potential of adarotene derivatives against Staphylococcus aureus strains. Bioorg Chem 2024; 145:107227. [PMID: 38387400 DOI: 10.1016/j.bioorg.2024.107227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Multidrug-resistant (MDR) pathogens are severely impacting our ability to successfully treat common infections. Here we report the synthesis of a panel of adarotene-related retinoids showing potent antimicrobial activity on Staphylococcus aureus strains (including multidrug-resistant ones). Fluorescence and molecular dynamic studies confirmed that the adarotene analogues were able to induce conformational changes and disfunctions to the cell membrane, perturbing the permeability of the phospholipid bilayer. Since the major obstacle for developing retinoids is their potential cytotoxicity, a selected candidate was further investigated to evaluate its activity on a panel of human cell lines. The compound was found to be well tolerated, with IC50 5-15-fold higher than the MIC on S. aureus strains. Furthermore, the adarotene analogue had a good pharmacokinetic profile, reaching a plasma concentration of about 6 μM after 0.5 h after administration (150 mg/kg), at least twice the MIC observed against various bacterial strains. Moreover, it was demonstrated that the compound potentiated the growth-inhibitory effect of the poorly bioavailable rifaximin, when used in combination. Overall, the collected data pave the way for the development of synthetic retinoids as potential therapeutics for hard-to-treat infectious diseases caused by antibiotic-resistant Gram-positive pathogens.
Collapse
Affiliation(s)
- Salvatore Princiotto
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy
| | - Bruno Casciaro
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Alvaro G Temprano
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; Experimental Hepatology and Drug Targeting (HEVEPHARM) Group, Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain
| | - Loana Musso
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy
| | - Francesca Sacchi
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy
| | - Maria Rosa Loffredo
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Floriana Cappiello
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Sacco
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Giammarco Raponi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | | | | | - Maria Luisa Mangoni
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy.
| | | |
Collapse
|
11
|
Fu T, Zhang H, Zheng Q. Assessing the role of residue Phe108 of cytochrome P450 3A4 in allosteric effects of midazolam metabolism. Phys Chem Chem Phys 2024; 26:8807-8814. [PMID: 38421040 DOI: 10.1039/d3cp05270b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Cytochrome P450 3A4 (CYP3A4) is involved in the metabolism of more drugs in clinical use than any other xenobiotic-metabolizing enzyme. CYP3A4-mediated drug metabolism is usually allosterically modulated by substrate concentration (homotropic allostery) and other drugs (heterotropic allostery), exhibiting unusual kinetic profiles and regiospecific metabolism. Recent studies suggest that residue Phe108 (F108) of CYP3A4 may have an important role in drug metabolism. In this work, residue mutations were coupled with well-tempered metadynamics simulations to assess the importance of F108 in the allosteric effects of midazolam metabolism. Comparing the simulation results of the wild-type and mutation systems, we identify that the π-π interaction and steric effect between the F108 side chain and midazolam is favorable for the stable binding of substrate in the active site. F108 also plays an important role in the transition of substrate binding mode, which mainly induces the transition of substrate binding mode by forming π-π interactions with multiple aromatic rings of the substrate. Moreover, the side chain of F108 is closely related to the radius and depth of the 2a and 2f channels, and F108 may further regulate drug metabolism by affecting the pathway, orientation, or time of substrate entry into the CYP3A4 active site or product egress from the active site. Altogether, we suggest that F108 affects drug metabolism and regulatory mechanisms by affecting substrate binding stability, binding mode transition, and channel characteristics of CYP3A4. Our findings could promote the understanding of complicated allosteric mechanisms in CYP3A4-mediated drug metabolism, and the knowledge could be used for drug development and disease treatment.
Collapse
Affiliation(s)
- Tingting Fu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130023, China
| | - Hongxing Zhang
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130023, China
| | - Qingchuan Zheng
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun, 130023, China
| |
Collapse
|
12
|
Farci D, Milenkovic S, Iesu L, Tanas M, Ceccarelli M, Piano D. Structural characterization and functional insights into the type II secretion system of the poly-extremophile Deinococcus radiodurans. J Biol Chem 2024; 300:105537. [PMID: 38072042 PMCID: PMC10828601 DOI: 10.1016/j.jbc.2023.105537] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 01/21/2024] Open
Abstract
The extremophile bacterium D. radiodurans boasts a distinctive cell envelope characterized by the regular arrangement of three protein complexes. Among these, the Type II Secretion System (T2SS) stands out as a pivotal structural component. We used cryo-electron microscopy to reveal unique features, such as an unconventional protein belt (DR_1364) around the main secretin (GspD), and a cap (DR_0940) found to be a separated subunit rather than integrated with GspD. Furthermore, a novel region at the N-terminus of the GspD constitutes an additional second gate, supplementing the one typically found in the outer membrane region. This T2SS was found to contribute to envelope integrity, while also playing a role in nucleic acid and nutrient trafficking. Studies on intact cell envelopes show a consistent T2SS structure repetition, highlighting its significance within the cellular framework.
Collapse
Affiliation(s)
- Domenica Farci
- Department of Plant Physiology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland; Department of Life and Environmental Sciences, Università degli Studi di Cagliari, Cagliari, Italy; R&D Department, ReGenFix Laboratories, Sardara, Italy.
| | - Stefan Milenkovic
- Department of Physics and IOM/CNR, Università degli Studi di Cagliari, Monserrato, Italy
| | - Luca Iesu
- Department of Life and Environmental Sciences, Università degli Studi di Cagliari, Cagliari, Italy
| | - Marta Tanas
- Department of Life and Environmental Sciences, Università degli Studi di Cagliari, Cagliari, Italy
| | - Matteo Ceccarelli
- Department of Physics and IOM/CNR, Università degli Studi di Cagliari, Monserrato, Italy
| | - Dario Piano
- Department of Plant Physiology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland; Department of Life and Environmental Sciences, Università degli Studi di Cagliari, Cagliari, Italy; R&D Department, ReGenFix Laboratories, Sardara, Italy.
| |
Collapse
|
13
|
Jardón-Valadez E, Ulloa-Aguirre A. Tracking conformational transitions of the gonadotropin hormone receptors in a bilayer of (SDPC) poly-unsaturated lipids from all-atom molecular dynamics simulations. PLoS Comput Biol 2024; 20:e1011415. [PMID: 38206994 PMCID: PMC10807830 DOI: 10.1371/journal.pcbi.1011415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/24/2024] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
Glycoprotein hormone receptors [thyrotropin (TSHR), luteinizing hormone/chorionic gonadotropin (LHCGR), and follicle stimulating hormone (FSHR) receptors] are rhodopsin-like G protein-coupled receptors. These receptors display common structural features including a prominent extracellular domain with leucine-rich repeats (LRR) stabilized by β-sheets and a long and flexible loop known as the hinge region (HR), and a transmembrane (TM) domain with seven α-helices interconnected by intra- and extracellular loops. Binding of the ligand to the LRR resembles a hand coupling transversally to the α- and β-subunits of the hormone, with the thumb being the HR. The structure of the FSH-FSHR complex suggests an activation mechanism in which Y335 at the HR binds into a pocket between the α- and β-chains of the hormone, leading to an adjustment of the extracellular loops. In this study, we performed molecular dynamics (MD) simulations to identify the conformational changes of the FSHR and LHCGR. We set up a FSHR structure as predicted by AlphaFold (AF-P23945); for the LHCGR structure we took the cryo-electron microscopy structure for the active state (PDB:7FII) as initial coordinates. Specifically, the flexibility of the HR domain and the correlated motions of the LRR and TM domain were analyzed. From the conformational changes of the LRR, TM domain, and HR we explored the conformational landscape by means of MD trajectories in all-atom approximation, including a membrane of polyunsaturated phospholipids. The distances and procedures here defined may be useful to propose reaction coordinates to describe diverse processes, such as the active-to-inactive transition, and to identify intermediaries suited for allosteric regulation and biased binding to cellular transducers in a selective activation strategy.
Collapse
Affiliation(s)
- Eduardo Jardón-Valadez
- Departamento de Recursos de la Tierra, Unidad Lerma, Universidad Autónoma Metropolitana, Lerma de Villada, Estado de México, Mexico
| | - Alfredo Ulloa-Aguirre
- Instituto Nacional de Ciencias Medicas y Nutrición “Salvador Zubiran”. Mexico City, Mexico
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México. Mexico City, Mexico
| |
Collapse
|
14
|
Hsiao WC, Niu GH, Lo CF, Wang JY, Chi YH, Huang WC, Tung CW, Sung PJ, Tsou LK, Zhang MM. Marine diterpenoid targets STING palmitoylation in mammalian cells. Commun Chem 2023; 6:153. [PMID: 37463995 DOI: 10.1038/s42004-023-00956-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
Natural products are important sources of therapeutic agents and useful drug discovery tools. The fused macrocycles and multiple stereocenters of briarane-type diterpenoids pose a major challenge to total synthesis and efforts to characterize their biological activities. Harnessing a scalable source of excavatolide B (excB) from cultured soft coral Briareum stechei, we generated analogs by late-stage diversification and performed structure-activity analysis, which was critical for the development of functional excB probes. We further used these probes in a chemoproteomic strategy to identify Stimulator of Interferon Genes (STING) as a direct target of excB in mammalian cells. We showed that the epoxylactone warhead of excB is required to covalently engage STING at its membrane-proximal Cys91, inhibiting STING palmitoylation and signaling. This study reveals a possible mechanism-of-action of excB, and expands the repertoire of covalent STING inhibitors.
Collapse
Affiliation(s)
- Wan-Chi Hsiao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan
| | - Ping-Jyun Sung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 804201, Taiwan.
- National Museum of Marine Biology and Aquarium, Pingtung, 944401, Taiwan.
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, 404394, Taiwan.
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan.
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, 35053, Taiwan.
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan.
| |
Collapse
|
15
|
Li Y, Acharya A, Yang L, Liu J, Tajkhorshid E, Zgurskaya HI, Jackson M, Gumbart JC. Insights into substrate transport and water permeation in the mycobacterial transporter MmpL3. Biophys J 2023; 122:2342-2352. [PMID: 36926696 PMCID: PMC10257117 DOI: 10.1016/j.bpj.2023.03.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/04/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Mycobacteria, such as Mycobacterium tuberculosis, are characterized by a uniquely thick and waxy cell envelope that consists of two membranes, with a variety of mycolates comprising their outer membrane (OM). The protein Mycobacterial membrane protein Large 3 (MmpL3) is responsible for the transport of a primary OM component, trehalose monomycolate (TMM), from the inner (cytoplasmic) membrane (IM) to the periplasmic space, a process driven by the proton gradient. Although multiple structures of MmpL3 with bound substrates have been solved, the exact pathway(s) for TMM or proton transport remains elusive. Here, employing molecular dynamics simulations we investigate putative pathways for either transport species. We hypothesized that MmpL3 will cycle through similar conformational states as the related transporter AcrB, which we used as targets for modeling the conformation of MmpL3. A continuous water pathway through the transmembrane region was found in one of these states, illustrating a putative pathway for protons. Additional equilibrium simulations revealed that TMM can diffuse from the membrane into a binding pocket in MmpL3 spontaneously. We also found that acetylation of TMM, which is required for transport, makes it more stable within MmpL3's periplasmic cavity compared with the unacetylated form.
Collapse
Affiliation(s)
- Yupeng Li
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Atanu Acharya
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia
| | - Lixinhao Yang
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia
| | - Jinchan Liu
- Department of Molecular Biophysics and Biochemistry (MB&B), Yale University, New Haven, Connecticut
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois; Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Helen I Zgurskaya
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - James C Gumbart
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia; School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia.
| |
Collapse
|
16
|
Montero-Dominguez PA, Corzo G. Characterization of the coupling mechanism of scorpion β-neurotoxins on the voltage-gated sodium channel hNav1.6. J Biomol Struct Dyn 2023; 41:14419-14427. [PMID: 36812246 DOI: 10.1080/07391102.2023.2181629] [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: 11/17/2022] [Accepted: 02/12/2023] [Indexed: 02/24/2023]
Abstract
Scorpion β-neurotoxins represent a pharmacological group that affects voltage-gated sodium channels (Nav). Despite knowing the electrophysiological effect of these toxins on Nav channels, the molecular mechanism by which the union is carried out is still undetermined. In this study, computational techniques such as modeling, docking and molecular dynamics were used to elucidate the mechanism of interaction between scorpion β-neurotoxins using the neurotoxin nCssII and its recombinant variant CssII-RCR, which bind to the site-4, an extracellular receptor, of the human sodium channel hNav1.6. Different modes of interaction were observed for both toxins, where the main distinguishing feature was the interaction generated by the residue E15 on such site-4; that is, E15 in nCssII exhibits an interaction with the voltage-sensing domain II, and the same residue E15 of CssII-RCR exhibits an interaction with domain III. Despite this difference in interaction by E15, it is seen that both neurotoxins interact with similar regions of the voltage sensing domain such as the S3-S4 connecting loop (L834-E838) of the hNav1.6. Our simulations present a first approach to the mode of interaction of scorpion beta-neurotoxins in toxin-receptor complexes, being able to explain at the molecular level the phenomenon of voltage sensor entrapment generated by these toxins.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pavel Andrei Montero-Dominguez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Mor, México
| | - Gerardo Corzo
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca Mor, México
| |
Collapse
|
17
|
Antibacterial alkylguanidino ureas: Molecular simplification approach, searching for membrane-based MoA. Eur J Med Chem 2022; 231:114158. [DOI: 10.1016/j.ejmech.2022.114158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
|