1
|
Masia KJ, Mhlongo NN, Pooe OJ, Ibrahim MA, Kappo AP, Simelane MBC. Antiplasmodial potential of compounds isolated from Ziziphus mucronata and their binding to Plasmodium falciparum HGXPRT using biophysical and molecular docking studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5453-5463. [PMID: 39560751 PMCID: PMC11985687 DOI: 10.1007/s00210-024-03611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
The increasing resistance of Plasmodium parasites to currently available antiplasmodial therapies poses a significant challenge in treating malaria. Since ancient times, plants have served as a primary source of novel pharmacologically active compounds for drug development. Therefore, this study aimed to explore the antiplasmodial properties of pentacyclic triterpenes isolated from Ziziphus mucronata bark, with an emphasis on their mechanism of action. Dichloromethane and ethyl acetate extracts of the stem bark were subjected to silica gel column chromatography, which led to the isolation of three known triterpenoids: betulinic acid, methyl betulinate, and lupeol. The compounds were then evaluated for antiplasmodial activity against Plasmodium falciparum NF54 strains using the Plasmodium lactate dehydrogenase (pLDH) assay. In silico evaluation of the isolated compounds was conducted through molecular docking and further validated with in vitro experiments against a purified protein target, Plasmodium falciparum hypoxanthine-guanine-xanthine phosphoribosyltransferase (PfHGXPRT). Betulinic acid, methyl betulinate, and lupeol exhibited potent antiplasmodial activities with IC50 values of 20, 10.11, and 7.56 µg/mL, respectively. Lupeol exhibited the highest binding energy of - 7.6 kcal/mol. Differential scanning fluorimetry revealed that lupeol decreases the Tm of PfHGXPRT, thus decreasing the protein's thermal stability. At high concentrations, lupeol also increased protein absorbance, indicating the detection of hydrophobic amino acids and protein unfolding. This study proves that Z. mucronata could serve as a reservoir of effective agents for treating malaria, while also scientifically validating its use in traditional medicine. However, further experimental studies are required to substantiate its relevant therapeutic effects.
Collapse
Affiliation(s)
- Kgaugelo J Masia
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, 2006, South Africa
| | - Ndumiso N Mhlongo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Ofentse J Pooe
- School of Life Sciences, University of KwaZulu-Natal, Durban, 4000, Westville, South Africa
| | - Mohammed A Ibrahim
- Department of Biochemistry, Ahmadu Bello University, Zaria, 810107, Nigeria
| | - Abidemi P Kappo
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, 2006, South Africa
| | - Mthokozisi B C Simelane
- Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park Kingsway Campus, Johannesburg, 2006, South Africa.
| |
Collapse
|
2
|
Papay RS, Perez DM. Further In Vitro and Ex Vivo Pharmacological and Kinetic Characterizations of CCF219B: A Positive Allosteric Modulator of the α 1A-Adrenergic Receptor. Pharmaceuticals (Basel) 2025; 18:476. [PMID: 40283913 PMCID: PMC12030198 DOI: 10.3390/ph18040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Alterations in the adrenergic system have been associated with the pathophysiology of Alzheimer's disease (AD). A novel α1A-adrenergic receptor (AR)-positive allosteric modulator (PAM), CCF219B, has been shown to outperform donepezil with rescue of AD cognition/memory deficits with a reduction in amyloid biomarkers and without cardiovascular side effects. Initial pharmacological analysis in transfected cell lines revealed a signal bias with increased efficacy (but not potency) of cAMP signaling and ligand selectivity for norepinephrine (NE). As most GPCR allosteric modulators change the potency of agonists, we hypothesized and now report that CCF219B induced additional aspects of its allosteric interactions with NE that may provide mechanistic insight. Methods: Using Rat-1 fibroblasts stably transfected with α1A-AR, we determined the activation profile of pERK and p38 messengers by CCF219B in the presence of NE. Using membranes prepared from the stably transfected fibroblasts or from the brain of WT mice or the AD mouse model, hAPP(lon), equilibrium or kinetic radioligand-binding analyses were performed. Results: We identified p-ERK1/2 but not p38 as an additional signal pathway that is potentiated by CCF219B in the presence of NE. An analysis of binding studies of CCF219B in membranes derived from the brains of WT or hAPP(lon) mice revealed profiles that were time-dependent and resulted in an increase in α1A-AR expression that was unaltered in the presence of cycloheximide or when performed at 37 °C. hAPP(lon) mice displayed a reduction in α1A-AR-binding sites that were rescued upon prolonged incubation with CCF219B but also displayed a compensatory increase in α1B/D-AR subtype expression. Binding kinetics reveal that CCF219B can decrease the association rate of 3H-NE but only in the presence of GTP. The association rate increased for the radiolabeled antagonist, 125I-HEAT. There were no changes in the dissociation rate of either radiolabel. Conclusions: CCF219B affects the association but not the dissociation rate of NE and explains its ability to increase the active state of the receptor by promoting a pre-coupled conformation, consistent with increasing efficacy but not potency. Potentiation of pERK may contribute to CCF219B's ability to confer neuroprotection and be pro-cognitive in AD. CCF219B's ability to increase the expression of α1A-AR provides a positive feedback loop and strengthens the hypothesis that α1-AR subtypes may be involved in AD etiology and/or progression.
Collapse
Affiliation(s)
| | - Dianne M. Perez
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
3
|
Sozańska N, Klepka BP, Niedzwiecka A, Zhukova L, Dadlez M, Greb-Markiewicz B, Ożyhar A, Tarczewska A. The molecular properties of the bHLH TCF4 protein as an intrinsically disordered hub transcription factor. Cell Commun Signal 2025; 23:154. [PMID: 40149012 PMCID: PMC11948756 DOI: 10.1186/s12964-025-02154-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Transcription factor 4 (TCF4) is a member of the basic helix-loop-helix (bHLH) family of transcription factors that guides proper embryogenesis, particularly neurogenesis, myogenesis, heart development and hematopoiesis. The interaction of TCF4 with DNA is dependent on the presence of a conserved bHLH domain, particularly the presence of a basic (b) motif. Most mutations in the Tcf4 gene are either associated with the development of serious nervous system disorders, such as Pitt-Hopkins syndrome or schizophrenia, or are lethal. Although TCF4 is essential for the proper development and function of the human body, there is a lack of fundamental knowledge about the structure of TCF4 since structural studies were previously limited exclusively to its bHLH. METHODS Recombinant full-length TCF4 was expressed in bacterial cells and purified using chromatographic techniques. To compare the properties of TCF4 in its apo and holo form, we determined the dissociation constant (KD) of the TCF4:DNA complex using independent methods, including fluorescence polarization (FP), electrophoretic mobility shift assay (EMSA), and fluorescence correlation spectroscopy (FCS). Then we compared the properties of TCF4 in its apo and holo form in relation to the changes of the conformation of the polypeptide chain (hydrogen/deuterium exchange mass spectrometry; HDX-MS), hydrodynamic properties (e.g., sedimentation-velocity analytical ultracentrifugation; SV-AUC), and stability (thermal shift, circular dichroism; CD). RESULTS We demonstrate the molecular characteristics of TCF4, the dimer of which is one of the largest intrinsically disordered proteins (IDPs) described to date. According to our findings, the structure of TCF4 is extensively disordered. Only the bHLH domain exhibits a stable fold. Strikingly, Ephrussi-box (E-box) binding via the bHLH domain has no significant effect on the disordered nature of TCF4, but it does influence the dynamic of bHLH and stability of the protein. CONCLUSIONS We suggest that bHLH plays the role of an anchor localizing TCF4 to specific gene sequences. The dual nature of the TCF4 structure and the fact that the intrinsically disordered regions (IDRs) represent most of the protein sequence, suggest that TCF4 may act as a hub transcription factor regulating the expression of specific genes through the interaction of IDRs with gene-specific partners.
Collapse
Affiliation(s)
- Nikola Sozańska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland
| | - Barbara P Klepka
- Laboratory of Biological Physics, Institute of Physics, Polish Academy of Sciences, Aleja Lotnikow 32/46, Warsaw, PL-02668, Poland
| | - Anna Niedzwiecka
- Laboratory of Biological Physics, Institute of Physics, Polish Academy of Sciences, Aleja Lotnikow 32/46, Warsaw, PL-02668, Poland
| | - Lilia Zhukova
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, Warsaw, 02-106, Poland
| | - Michał Dadlez
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, Warsaw, 02-106, Poland
| | - Beata Greb-Markiewicz
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland
| | - Andrzej Ożyhar
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland
| | - Aneta Tarczewska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland.
| |
Collapse
|
4
|
Obisesan OA, Ofori S, Orobator ON, Sharma H, Groetecke E, Awuah SG. Discovery of a Pyrazolopyridinone-Based MYC Inhibitor That Selectively Engages Intracellular c-MYC and Disrupts MYC-MAX Heterodimerization. J Med Chem 2025; 68:6233-6251. [PMID: 40077826 DOI: 10.1021/acs.jmedchem.4c02556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
c-MYC is an oncogenic transcription factor that plays a crucial role in the regulation of downstream targets involved in proliferation, apoptosis, differentiation, metabolism, signaling, and immune response processes whose deregulation leads to the progression of different pathologies. The development of selective and potent small-molecule inhibitors of c-MYC remains a grand challenge in chemical biology and medicine due to its undruggability, derived from extensive intrinsic disorder. In this study, we identified a novel dihydro pyrazolo pyridinone scaffold, MY05, that selectively targets c-MYC in cells and disrupts MYC-MAX interaction. MY05 engages intracellular c-MYC, modulates c-MYC thermal stability, reduces c-MYC transcriptional targets, and inhibits proliferation in cancer cells and tumor growth in mice. In summary, we identified a novel compound that directly interacts with c-MYC to disrupt the transcriptional program.
Collapse
Affiliation(s)
- Oluwatosin A Obisesan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Himanshi Sharma
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Emma Groetecke
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- Markey NCI Comprehensive Cancer Center, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Bioelectronics and Nanomedicine Research Center, Lexington, Kentucky 40506, United States
| |
Collapse
|
5
|
Huang EYW, Kuang F, Wu H, Yu CX, Chen X, Vasku G, Nguyen LTA, Jeppe KJ, Coussens AK, Kwai BX, Leung IK. An integrated structural and biophysical approach to study carbon metabolism in Mycobacterium tuberculosis. QRB DISCOVERY 2025; 6:e15. [PMID: 40395558 PMCID: PMC12088919 DOI: 10.1017/qrd.2025.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 05/22/2025] Open
Abstract
Metabolic enzymes are the catalysts that drive the biochemical reactions essential for sustaining life. Many of these enzymes are tightly regulated by feedback mechanisms. To fully understand their roles and modulation, it is crucial to investigate the relationship between their structure, catalytic mechanism, and function. In this perspective, by using three examples from our studies on Mycobacterium tuberculosis (Mtb) isocitrate lyase and related proteins, we highlight how an integrated approach combining structural, activity, and biophysical data provides insights into their biological functions. These examples underscore the importance of employing fast-fail experiments at the early stages of a research project, emphasise the value of complementary techniques in validating findings, and demonstrate how in vitro data combined with chemical, biochemical, and physiological knowledge can lead to a broader understanding of metabolic adaptations in pathogenic bacteria. Finally, we address the unexplored questions in Mtb metabolism and discuss how we expand our approach to include microbiological and bioanalytical techniques to further our understanding. Such an integrated and interdisciplinary strategy has the potential to uncover novel regulatory mechanisms and identify new therapeutic opportunities for the eradication of tuberculosis. The approach can also be broadly applied to investigate other biochemical networks and complex biological systems.
Collapse
Affiliation(s)
- Evelyn Y.-W. Huang
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Francis Kuang
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Haozhe Wu
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Chai Xin Yu
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Xiaoxu Chen
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Glenda Vasku
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Le Thao Anh Nguyen
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Katherine J. Jeppe
- Monash Proteomics and Metabolomics Platform, Monash University, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Anna K. Coussens
- Infectious Diseases and Immune Defence Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Brooke X.C. Kwai
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Ivanhoe K.H. Leung
- School of Chemistry and Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
Zhang B, Matus MF, Yao Q, Song X, Wu Z, Hu W, Häkkinen H, Xie J. Unraveling the Stoichiometric Interactions and Synergism between Ligand-Protected Gold Nanoparticles and Proteins. J Am Chem Soc 2025; 147:6404-6414. [PMID: 39823220 DOI: 10.1021/jacs.4c09879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Nanomaterials that engage in well-defined and tunable interactions with proteins are pivotal for the development of advanced applications. Achieving a precise molecular-level understanding of nano-bio interactions is essential for establishing these interactions. However, such an understanding remains challenging and elusive. Here, we identified stoichiometric interactions of water-soluble gold nanoparticles (Au NPs) with bovine serum albumin (BSA), unraveling their synergism in manipulating emission of nano-bio conjugates in the second near-infrared (NIR-II) regime. Using Au25(p-MBS)18 (p-MBS = para-mercaptobenzenesulfonic acid) as paradigm particles, we achieved precise binding of Au NPs to BSA with definitive molar ratios of 1:1 and 2:1, which is unambiguously evidenced by high-resolution mass spectrometry and transmission electron microscopy. Molecular dynamics simulations identified well-defined binding sites, mediated by electrostatic interactions and hydrogen bonds between the p-MBS moieties on the Au25(p-MBS)18 surface and BSA. Particularly, positively charged residues on BSA were found to be pivotal. By careful control of the molar ratio of Au25(p-MBS)18 to BSA, atomically precise [Au25(p-MBS)18]x-BSA conjugates (x = 1 or 2) could be formed. Through a comprehensive spectroscopy study, an electron transfer process and synergistic effect were manifested in the Au25(p-MBS)18-BSA conjugates, leading to drastically enhanced emission in the NIR-II window. This work offers insights into the precise engineering of nanomaterial-protein interactions and opens new avenues for the development of next-generation nano-bio conjugates for nanotheranostics.
Collapse
Affiliation(s)
- Bihan Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| | - María Francisca Matus
- Departments of Physics and Chemistry, Nanoscience Center, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Qiaofeng Yao
- Key Laboratory of Organic Integrated Circuits, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology & State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Zhennan Wu
- State Key Laboratory of Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China
| | - Wenping Hu
- Key Laboratory of Organic Integrated Circuits, Ministry of Education & Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin 300072, China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Hannu Häkkinen
- Departments of Physics and Chemistry, Nanoscience Center, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou 350207, China
| |
Collapse
|
7
|
Mulaudzi VE, Adeosun IJ, Adewumi AT, Soliman MES, Cosa S. Helichrysum populifolium Compounds Inhibit MtrCDE Efflux Pump Transport Protein for the Potential Management of Gonorrhoea Infection. Int J Mol Sci 2024; 25:13310. [PMID: 39769078 PMCID: PMC11677219 DOI: 10.3390/ijms252413310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The progressive development of resistance in Neisseria gonorrhoeae to almost all available antibiotics has made it crucial to develop novel approaches to tackling multi-drug resistance (MDR). One of the primary causes of antibiotic resistance is the over-expression of the MtrCDE efflux pump protein, making this protein a vital target for fighting against antimicrobial resistance (AMR) in N. gonorrhoeae. This study was aimed at evaluating the potential MtrCDE efflux pump inhibitors (EPIs) and their stability in treating gonorrhoea infection. This is significant because finding novel EPIs would allow for the longer maintenance of antibiotics at therapeutic levels, thereby prolonging the susceptibility of currently available antibiotics. A virtual screening of the selected Helichrysum populifolium compounds (4,5-dicaffeoylquinic acid, apigeninin-7-glucoside, and carvacrol) was conducted to evaluate their potential EPI activity. An integrated computational framework consisting of molecular docking (MD), molecular mechanics generalized born, and surface area solvation (MMGBSA) analysis, molecular dynamics simulations (MDS), and absorption, distribution, metabolism, and excretion (ADME) properties calculations were conducted. Of the tested compounds, 4,5-dicaffeoylquinic acid revealed the highest molecular docking binding energies (-8.8 kcal/mol), equivalent MMGBSA binding free energy (-54.82 kcal/mol), indicative of consistent binding affinity with the MtrD protein, reduced deviations and flexibility (root mean square deviation (RMSD) of 5.65 Å) and, given by root mean square fluctuation (RMSF) of 1.877 Å. Carvacrol revealed a docking score of -6.0 kcal/mol and a MMGBSA computed BFE of -16.69 kcal/mol, demonstrating the lowest binding affinity to the MtrD efflux pump compared to the remaining test compounds. However, the average RMSD (4.45 Å) and RMSF (1.638 Å) of carvacrol-bound MtrD protein showed no significant difference from the unbound MtrD protein, except for the reference compounds, implying consistent MtrD conformation throughout simulations and indicates a desirable feature during drug design. Additionally, carvacrol obeyed the Lipinski rule of five which confirmed the compound's drug-likeness properties making it the most promising EPI candidate based on its combined attributes of a reasonable binding affinity, sustained stability during MDS, its obedience to the Lipinski rule of five and compliance with drug-likeness criteria. An in vitro validation of the potential EPI activities of H. populifolium compounds confirmed that 4,5-dicaffeoylquinic acid reduced the expulsion of the bis-benzimide dye by MtrCDE pump, while carvacrol showed low accumulation compared to other compounds. While 4,5-dicaffeoylquinic acid demonstrated the highest binding affinity in computational analysis and an EPI activity in vitro, it showed lower stability compared to the other compounds, as indicated in MDS. This leaves carvacrol, as a better EPI candidate for the management of gonorrhoea infection.
Collapse
Affiliation(s)
- Vhangani E. Mulaudzi
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| | - Idowu J. Adeosun
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| | - Adeniyi T. Adewumi
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (A.T.A.); (M.E.S.S.)
| | - Mahmoud E. S. Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (A.T.A.); (M.E.S.S.)
| | - Sekelwa Cosa
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| |
Collapse
|
8
|
Škulj S, Kožić M, Barišić A, Vega A, Biarnés X, Piantanida I, Barisic I, Bertoša B. Comparison of two peroxidases with high potential for biotechnology applications - HRP vs. APEX2. Comput Struct Biotechnol J 2024; 23:742-751. [PMID: 38298178 PMCID: PMC10828542 DOI: 10.1016/j.csbj.2024.01.001] [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: 10/01/2023] [Revised: 01/01/2024] [Accepted: 01/01/2024] [Indexed: 02/02/2024] Open
Abstract
Peroxidases are essential elements in many biotechnological applications. An especially interesting concept involves split enzymes, where the enzyme is separated into two smaller and inactive proteins that can dimerize into a fully active enzyme. Such split forms were developed for the horseradish peroxidase (HRP) and ascorbate peroxidase (APX) already. Both peroxidases have a high potential for biotechnology applications. In the present study, we performed biophysical comparisons of these two peroxidases and their split analogues. The active site availability is similar for all four structures. The split enzymes are comparable in stability with their native analogues, meaning that they can be used for further biotechnology applications. Also, the tertiary structures of the two peroxidases are similar. However, differences that might help in choosing one system over another for biotechnology applications were noticed. The main difference between the two systems is glycosylation which is not present in the case of APX/sAPEX2, while it has a high impact on the HRP/sHRP stability. Further differences are calcium ions and cysteine bridges that are present only in the case of HRP/sHRP. Finally, computational results identified sAPEX2 as the systems with the smallest structural variations during molecular dynamics simulations showing its dominant stability comparing to other simulated proteins. Taken all together, the sAPEX2 system has a high potential for biotechnological applications due to the lack of glycans and cysteines, as well as due to high stability.
Collapse
Affiliation(s)
- Sanja Škulj
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb HR-10000, Croatia
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Matej Kožić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb HR-10000, Croatia
| | - Antun Barišić
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb HR-10000, Croatia
| | - Aitor Vega
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017 Barcelona, Spain
| | - Xevi Biarnés
- Laboratory of Biochemistry, Institut Químic de Sarrià, Universitat Ramon Llull, Via Augusta 390, 08017 Barcelona, Spain
| | - Ivo Piantanida
- Division of Organic Chemistry & Biochemistry, Ruđer Bošković Institute, Bijenička Cesta 54, 10 000 Zagreb, Croatia
| | - Ivan Barisic
- Molecular Diagnostics, Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, Vienna 1210, Austria
- Eko Refugium, Crno Vrelo 2, Slunj 47240, Croatia
| | - Branimir Bertoša
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb HR-10000, Croatia
| |
Collapse
|
9
|
Howes JM, Harper MT. Application of the Cellular Thermal Shift Assay (CETSA) to validate drug target engagement in platelets. Platelets 2024; 35:2354833. [PMID: 38767506 DOI: 10.1080/09537104.2024.2354833] [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: 03/08/2024] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024]
Abstract
Small molecule drugs play a major role in the study of human platelets. Effective action of a drug requires it to bind to one or more targets within the platelet (target engagement). However, although in vitro assays with isolated proteins can be used to determine drug affinity to these targets, additional factors affect target engagement and its consequences in an intact platelet, including plasma membrane permeability, intracellular metabolism or compartmentalization, and level of target expression. Mechanistic interpretation of the effect of drugs on platelet activity requires comprehensive investigation of drug binding in the proper cellular context, i.e. in intact platelets. The Cellular Thermal Shift Assay (CETSA) is a valuable method to investigate target engagement within complex cellular environments. The assay is based on the principle that drug binding to a target protein increases that protein's thermal stability. In this technical report, we describe the application of CETSA to platelets. We highlight CETSA as a quick and informative technique for confirming the direct binding of drugs to platelet protein targets, providing a platform for understanding the mechanism of action of drugs in platelets, and which will be a valuable tool for investigating platelet signaling and function.
Collapse
Affiliation(s)
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Shechter S, Pal RK, Trovato F, Rozen O, Gage MJ, Avni D. p70S6K as a Potential Anti-COVID-19 Target: Insights from Wet Bench and In Silico Studies. Cells 2024; 13:1760. [PMID: 39513867 PMCID: PMC11545240 DOI: 10.3390/cells13211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/27/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
The onset of SARS-CoV-2 infection in 2019 sparked a global COVID-19 pandemic. This infection is marked by a significant rise in both viral and host kinase activity. Our primary objective was to identify a pivotal host kinase essential for COVID-19 infection and the associated phenomenon of the cytokine storm, which may lead to long-term COVID-19 complications irrespective of viral genetic variations. To achieve this, our study tracked kinase phosphorylation dynamics in RAW264.7 macrophages following SPIKE transfection over time. Among the kinases surveyed, p70S6K (RPS6KB1) exhibited a 3.5-fold increase in phosphorylation at S418. This significant change prompted the selection of p70S6K for in silico investigation, utilizing its structure bound to M2698 (PDB: 7N93). M2698, an oral dual Akt/p70S6K inhibitor with an IC50 of 1.1 nM, exhibited psychosis side effects in phase I clinical trials, potentially linked to its interaction with Akt2. Our secondary objective was to discover a small-molecule analogue of M2698 that exhibits a distinct binding preference for p70S6K over Akt2 through computational modeling and analysis. The in silico part of our project began with validating the prediction accuracy of the docking algorithm, followed by an OCA analysis pinpointing specific atoms on M2698 that could be modified to enhance selectivity. Subsequently, our investigation led to the identification of an analog of M2698, designated as S34, that showed a superior docking score towards p70S6K compared to Akt2. To further assess the stability of S34 in its protein-ligand (PL) complexes with p70S6K and Akt2, MD simulations were conducted. These simulations suggest that S34, on average, forms two hydrogen bond interactions with p70S6K, whereas it only forms one hydrogen bond interaction with Akt2. This difference in hydrogen bond interactions likely contributed to the observed larger root mean square deviation (RMSD) of 0.3 nm in the S34-Akt2 complex, compared to 0.1 nm in the S34-p70S6K complex. Additionally, we calculated free binding energy to predict the strength of the binding interactions of S34 to p70S6K and Akt2, which showed ~2-fold favorable binding affinity of S34 in the p70S6K binding pocket compared to that in the Akt2 binding pocket. These observations may suggest that the S34-p70S6K complex is more stable than the S34-Akt2 complex. Our work focused on identifying a host kinase target and predicting the binding affinity of a novel small molecule to accelerate the development of effective treatments. The wet bench results specifically highlight p70S6K as a compelling anti-COVID-19 target. Meanwhile, our in silico investigations address the known off-target effects associated with M2698 by identifying a close analog called S34. In conclusion, this study presents novel and intriguing findings that could potentially lead to clinical applications with further investigations.
Collapse
Affiliation(s)
- Sharon Shechter
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854, USA; (S.S.); (M.J.G.)
| | | | - Fabio Trovato
- Psivant Therapeutics, 451 D Street, Boston, MA 02210, USA;
| | - Or Rozen
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel;
| | - Matthew J. Gage
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854, USA; (S.S.); (M.J.G.)
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel;
| |
Collapse
|
11
|
Yang P, Wang W, Hu Y, Wang Y, Xu Z, Liao X. Exploring high hydrostatic pressure effects on anthocyanin binding to serum albumin and food-derived transferrins. Food Chem 2024; 452:139544. [PMID: 38723571 DOI: 10.1016/j.foodchem.2024.139544] [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: 02/18/2024] [Revised: 04/20/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
This study investigated the effects of high hydrostatic pressure (HHP) on the binding interactions of cyanindin-3-O-glucoside (C3G) to bovine serum albumin, human serum albumin (HSA), bovine lactoferrin, and ovotransferrin. Fluorescence quenching revealed that HHP reduced C3G-binding affinity to HSA, while having a largely unaffected role for the other proteins. Notably, pretreating HSA at 500 MPa significantly increased its dissociation constant with C3G from 24.7 to 34.3 μM. Spectroscopic techniques suggested that HSA underwent relatively pronounced tertiary structural alterations after HHP treatments. The C3G-HSA binding mechanisms under pressure were further analyzed through molecular dynamics simulation. The localized structural changes in HSA under pressure might weaken its interaction with C3G, particularly polar interactions such as hydrogen bonds and electrostatic forces, consequently leading to a decreased binding affinity. Overall, the importance of pressure-induced structural alterations in proteins influencing their binding with anthocyanins was highlighted, contributing to optimizing HHP processing for anthocyanin-based products.
Collapse
Affiliation(s)
- Peiqing Yang
- Beijing Key Laboratory for Food Non-thermal processing, Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, National Engineering Research Center for Fruit & Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Wenxin Wang
- Beijing Key Laboratory for Food Non-thermal processing, Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, National Engineering Research Center for Fruit & Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan, China.
| | - Yongtao Wang
- Beijing Key Laboratory for Food Non-thermal processing, Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, National Engineering Research Center for Fruit & Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Zhenzhen Xu
- Beijing Key Laboratory for Food Non-thermal processing, Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, National Engineering Research Center for Fruit & Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; Institute of Quality Standard & Testing Technology for Agro-Products, Key Laboratory of Agro-food Safety and Quality, Ministry of Agriculture and Rural Affairs, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Xiaojun Liao
- Beijing Key Laboratory for Food Non-thermal processing, Key Laboratory of Fruit & Vegetable Processing, Ministry of Agriculture and Rural Affairs, National Engineering Research Center for Fruit & Vegetable Processing, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
12
|
Chauhan C, Singh P, Muthu SA, Parvez S, Selvapandiyan A, Ahmad B. Plumbagin accelerates serum albumin's amyloid aggregation kinetics and generates fibril polymorphism by inducing non-native β-sheet structures. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:141028. [PMID: 38849109 DOI: 10.1016/j.bbapap.2024.141028] [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: 03/08/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/09/2024]
Abstract
The ligand-induced conformational switch of proteins has great significance in understanding the biophysics and biochemistry of their self-assembly. In this work, we have investigated the ability of plumbagin (PL), a hydroxynaphthoquinone compound found in the root of the medicinal plant Plumbago zeylanica, to modulate aggregation precursor state, aggregation kinetics and generate distinct fibril of human serum albumin (HSA). PL was found to moderately bind (binding constant Ka ∼ 10-4 M-1)) to domain-II of HSA in the stoichiometric ratio of 1:1. We found that PL-HSA complex aggregation was accelerated as compared to that of HSA aggregation and it may be through an independent pathway. We also detected that fibril produced in the presence of PL is wider in diameter, contains a higher amount of β-sheet (∼18%) and disordered (∼46%) structures, and is less stable. We concluded that the acceleration of aggregation reaction and generation of fibril polymorphism was mainly because of the higher extent of unfolding and high content of non-native β-sheet structure in the aggregation precursor state of PL-HSA complex. This study offers opportunities to explore the ability of ligand binding to modulate aggregation reactions and generate polymorphic protein fibrils.
Collapse
Affiliation(s)
- Chanchal Chauhan
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, Jamia Hamdard, New Delhi 10062, India
| | - Poonam Singh
- UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari Campus, Mumbai 400098, India
| | - Shivani A Muthu
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, Jamia Hamdard, New Delhi 10062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India
| | | | - Basir Ahmad
- Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
13
|
Li X, Tao Q, Hu Q, Ma N, Ma G. In vitro gastrointestinal digestion and fecal fermentation of Pleurotus eryngii proteins extracted using different methods: insights for the utilization of edible mushroom-based proteins as novel nutritional and functional components. Food Funct 2024; 15:8865-8877. [PMID: 39120615 DOI: 10.1039/d4fo02604g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Pleurotus eryngii (P. eryngii) protein is considered a high-quality protein because it is rich in essential amino acids and displays multiple significant functional characterizations that vary with its fabrication processes. We aimed to investigate the differences in P. eryngii protein extracted via alkaline extraction and acid precipitation (AA), cellulase complex alkaline extraction and acid precipitation (CAA), ultrasound-assisted alkaline extraction and acid precipitation (UAA), and salt dissolution (S) in terms of gastrointestinal digestion and fecal fermentation consequences. Protein hydrolysis and structural analysis were performed after in vitro gastrointestinal digestion, and it was found that AA showed the highest hydrolysis degree, whereas CAA showed the lowest. The results of fluorescence chromatography and infrared chromatography indicated that the reasons for the digestion difference might be the unfolding degrees of the protein tertiary structure and polysaccharide content, which is the major component of crude proteins and can prevent protein hydrolysis. Metagenomic analysis suggested that compared with other groups, AA had excellent biological functions, including regulating obesity and insulin-related microbiota. This study could provide a new theoretical basis for the P. eryngii protein as a novel type of nutritional and functional component and contributes to the development of a diversified emerging food protein supply system.
Collapse
Affiliation(s)
- Xinyi Li
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China.
| | - Qi Tao
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China.
| | - Qiuhui Hu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China.
| | - Ning Ma
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China.
| | - Gaoxing Ma
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China.
| |
Collapse
|
14
|
Zhu L, Wu H, Xu Z, Guo L, Zhao J. Analysis of the effect of cations on protein conformational stability using solid-state nanopores. Analyst 2024; 149:3186-3194. [PMID: 38639484 DOI: 10.1039/d4an00248b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
The conformation of proteins is closely related to their biological functions, and it is affected by many factors, including the type of cations in solution. However, it is difficult to detect the conformational changes of a protein in situ. As a single-molecule sensing technology, nanopores can convert molecular structural information into analyzable current signals within a reasonable time range. Herein, we detect and analyze the effects of two different types of monovalent cations (Na+ and Li+) on a model protein bovine serum albumin (BSA) conformation using SiNx nanopores with different diameters. The quantitative analysis results show that the excluded volume of BSA in LiCl salt solutions is larger than the value in NaCl solution, indicating that Li+ is more prone to unfolding the proteins and making them unstable. This study demonstrated that nanopores enable the in situ detection of the structure of proteins at the single-molecule level and provide a new approach for the quantitative analysis of proteins.
Collapse
Affiliation(s)
- Libo Zhu
- School of Medical Imaging, Wannan Medical College, Wuhu, 241002, China.
| | - Hongwen Wu
- Jiangxi Institute of Respiratory Disease, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhengyuan Xu
- School of Medical Imaging, Wannan Medical College, Wuhu, 241002, China.
| | - Lanying Guo
- School of Medical Imaging, Wannan Medical College, Wuhu, 241002, China.
| | - Jinsong Zhao
- School of Medical Imaging, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
15
|
Salau VF, Erukainure OL, Aljoundi A, Akintemi EO, Elamin G, Odewole OA. Exploring the inhibitory action of betulinic acid on key digestive enzymes linked to diabetes via in vitro and computational models: approaches to anti-diabetic mechanisms. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:411-432. [PMID: 38764437 DOI: 10.1080/1062936x.2024.2352729] [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: 03/20/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024]
Abstract
Phytochemicals are now increasingly exploited as remedial agents for the management of diabetes due to side effects attributable to commercial antidiabetic agents. This study investigated the structural and molecular mechanisms by which betulinic acid exhibits its antidiabetic effect via in vitro and computational techniques. In vitro antidiabetic potential was analysed via on α-amylase, α-glucosidase, pancreatic lipase and α-chymotrypsin inhibitory assays. Its structural and molecular inhibitory mechanisms were investigated using Density Functional Theory (DFT) analysis, molecular docking and molecular dynamics (MD) simulation. Betulinic acid significantly (p < 0.05) inhibited α-amylase, α-glucosidase, pancreatic lipase and α-chymotrypsin enzymes with IC50 of 70.02 μg/mL, 0.27 μg/mL, 1.70 μg/mL and 8.44 μg/mL, respectively. According to DFT studies, betulinic acid possesses similar reaction in gaseous phase and water due to close values observed for highest occupied molecular orbital (HOMO) and lowest occupied molecular orbital (LUMO) and the chemical descriptors. The dipole moment indicates that betulinic acid has high polarity. Molecular electrostatic potential surface revealed the electrophilic and nucleophilic attack-prone atoms of the molecule. Molecular dynamic studies revealed a stable complex between betulinic acid and α-amylase, α-glucosidase, pancreatic lipase and α-chymotrypsin. The study elucidated the potent antidiabetic properties of betulinic acid by revealing its conformational inhibitory mode of action on enzymes involved in the onset of diabetes.
Collapse
Affiliation(s)
- V F Salau
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - O L Erukainure
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - A Aljoundi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Attahadi, Tripoli, Libya
| | - E O Akintemi
- School of Chemistry and Physics, University of KwaZulu-Natal, Durban, South Africa
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - G Elamin
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - O A Odewole
- Department of Pure and Industrial Chemistry, Faculty of Physical Sciences, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
16
|
Renodon-Corniere A, Mikawa T, Kuwabara N, Ito K, Levitsky D, Iwasaki H, Takahashi M. Human Rad51 Protein Requires Higher Concentrations of Calcium Ions for D-Loop Formation than for Oligonucleotide Strand Exchange. Int J Mol Sci 2024; 25:3633. [PMID: 38612444 PMCID: PMC11011376 DOI: 10.3390/ijms25073633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Human Rad51 protein (HsRad51)-promoted DNA strand exchange, a crucial step in homologous recombination, is regulated by proteins and calcium ions. Both the activator protein Swi5/Sfr1 and Ca2+ ions stimulate different reaction steps and induce perpendicular DNA base alignment in the presynaptic complex. To investigate the role of base orientation in the strand exchange reaction, we examined the Ca2+ concentration dependence of strand exchange activities and structural changes in the presynaptic complex. Our results show that optimal D-loop formation (strand exchange with closed circular DNA) required Ca2+ concentrations greater than 5 mM, whereas 1 mM Ca2+ was sufficient for strand exchange between two oligonucleotides. Structural changes indicated by increased fluorescence intensity of poly(dεA) (a poly(dA) analog) reached a plateau at 1 mM Ca2+. Ca2+ > 2 mM was required for saturation of linear dichroism signal intensity at 260 nm, associated with rigid perpendicular DNA base orientation, suggesting a correlation with the stimulation of D-loop formation. Therefore, Ca2+ exerts two different effects. Thermal stability measurements suggest that HsRad51 binds two Ca2+ ions with KD values of 0.2 and 2.5 mM, implying that one step is stimulated by one Ca2+ bond and the other by two Ca2+ bonds. Our results indicate parallels between the Mg2+ activation of RecA and the Ca2+ activation of HsRad51.
Collapse
Affiliation(s)
| | - Tsutomu Mikawa
- RIKEN Center for Biosystems Dynamics Research, Yokohama 230-0045, Japan;
| | - Naoyuki Kuwabara
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan;
| | - Kentaro Ito
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan;
| | - Dmitri Levitsky
- Nantes Université, CNRS, US2B, UMR 6286, F-44000 Nantes, France; (A.R.-C.); (D.L.)
| | - Hiroshi Iwasaki
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan;
- Innovative Science Institute, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Masayuki Takahashi
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan;
| |
Collapse
|
17
|
Swargiary A, Daimari M, Swargiary A, Biswas A, Brahma D, Singha H. Identification of phytocompounds as potent inhibitors of sodium/glucose cotransporter-2 leading to diabetes treatment. J Biomol Struct Dyn 2024:1-14. [PMID: 38379332 DOI: 10.1080/07391102.2024.2319674] [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: 09/30/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Type-II diabetes, a major metabolic disorder has threatened the very existence of a healthy life since long ago. Commercially available antidiabetic drugs are known for several adverse effects. The present study attempted to identify potential phytocompounds as inhibitors of sodium/glucose cotransporter-2 (SGLT2), a major protein that helps in glucose re-absorption from renal tubules. A total of 28 phytocompounds were collected based on the literature survey. 3D co-ordinates of phytocompounds were collected from PubChem database. Molecular docking was carried out with SGLT2 protein and the best 3 docking complexes were subjected to molecular dynamics simulation for 100 ns. Free energy changes were also analyzed using MM/PBSA analysis. Phytocompounds were also analyzed for their drug-likeness and ADMET properties. Docking study observed a strong binding affinity of phytocompounds (> -7.0 kcal/mol). More than 10 phytocompounds showed better binding affinity compared to reference drugs. Further analysis of three best docking complexes when analyzed by MD simulation showed better stability and compactness of the complexes compared to reference drug, empagliflozin. MM/PBSA analysis also revealed that van der Waals force and electrostatic energy are the major binding energy involved in the complex formation. Like docking energy, free energy analysis also observed stronger binding energies (ΔGGAS) in SGLT2-phytocompound complexes compared to empagliflozin complex. All the phytocompounds showed drug-likeness and considerable ADMET properties. The study, therefore, suggests that Trifolirhizin-6'-monoacetate, Aspalathin, and Quercetin-3-glucoside could be a possible inhibitor of SGLT2 protein. However, further studies need to be carried out to reveal the exact mode of activity.
Collapse
Affiliation(s)
- Ananta Swargiary
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| | - Manita Daimari
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| | - Arup Swargiary
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| | - Arup Biswas
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| | - Dulur Brahma
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| | - Hiloljyoti Singha
- Pharmacology and Bioinformatics Laboratory, Department of Zoology, Bodoland University, Kokrajhar, Assam, India
| |
Collapse
|
18
|
Pandaleke TA, Handono K, Widasmara D, Susianti H. The immunomodulatory activity of Orthosiphon aristatus against atopic dermatitis: Evidence-based on network pharmacology and molecular simulations. J Taibah Univ Med Sci 2024; 19:164-174. [PMID: 38047238 PMCID: PMC10692725 DOI: 10.1016/j.jtumed.2023.10.005] [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: 05/15/2023] [Revised: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 12/05/2023] Open
Abstract
Objectives To explore the potential activity of Orthosiphon aristatus (OA) against atopic dermatitis (AD). Methods Phytocompounds from OA were identified through chromatography analysis, then continued to target identification and functional annotation to explore the potential target of OA. Then, network pharmacology from annotated proteins determined protein targets for OA phytocompounds. Protein with highest rank according to the betweenness and closeness algorithm then continued to molecular docking and validated through molecular dynamics analysis. Results Chromatography data analysis revealed thirty-six compounds, predominantly classified as carboxylic acid, fatty acyls, and polyphenols. Upon identifying these compounds, network biology-based target identification revealed their potential bioactivity in modulating inflammation in AD. Tumour Necrosis Factor-alpha (TNF-α) and Prostaglandin G/H synthase 2 (PTGS2) emerged as the most probable targets based on hub centrality in the protein-protein interaction network. Later, molecular docking analyses highlighted sixteen compounds with good inhibitory activity against these two proteins. Notably, molecular dynamics simulation revealed that three compounds out of the previous sixteen potential compounds were more likely to act as the TNF-α and PTGS2 inhibitor as well as their native inhibitor. Those compounds are (1R,9R)-5-Cyclohexyl-11- (propylsulfonyl)-7,11- diazatricyclo[7.3.1.02,7]trideca- 2,4-dien-6-one, also known as ZINC8297940, as the best TNF-α inhibitor along with dl-Leucineamide and Benazol P as the potential inhibitor of PTGS2. Conclusions These findings suggest that OA may exert therapeutic effects against AD by controlling inflammation through TNF-α and PTGS2 signalling pathways.
Collapse
Affiliation(s)
- Thigita A. Pandaleke
- Doctoral Program of Medical Science, Universitas Brawijaya, Malang, East Java, Indonesia
- Department of Dermatology and Venereology, Faculty of Medicine, Sam Ratulangi University, RD Kandou Hospital, Jl. Raya Tanawangko No.56, Manado 95163, North Sulawesi, Indonesia
| | - Kusworini Handono
- Department of Clinical Pathology, Faculty of Medicine, Universitas Brawijaya – Saiful Anwar Hospital, Malang, East Java, Indonesia
| | - Dhelya Widasmara
- Department of Dermatology and Venereology, Faculty of Medicine, Universitas Brawijaya – Saiful Anwar Hospital, Malang, East Java, Indonesia
| | - Hani Susianti
- Department of Clinical Pathology, Faculty of Medicine, Universitas Brawijaya – Saiful Anwar Hospital, Malang, East Java, Indonesia
| |
Collapse
|
19
|
Elamin G, Aljoundi A, Alahmdi MI, Abo-Dya NE, Soliman MES. Revealing the Role of the Arg and Lys in Shifting Paradigm from BTK Selective Inhibition to the BTK/HCK Dual Inhibition - Delving into the Inhibitory Activity of KIN-8194 against BTK, and HCK in the Treatment of Mutated BTKCys481 Waldenström Macroglobulinemia: A Computational Approach. Anticancer Agents Med Chem 2024; 24:813-825. [PMID: 36752293 DOI: 10.2174/1871520623666230208102609] [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: 07/22/2022] [Revised: 10/30/2022] [Accepted: 12/08/2022] [Indexed: 02/09/2023]
Abstract
BACKGROUND Despite the early success of Bruton's tyrosine kinase (BTK) inhibitors in the treatment of Waldenström macroglobulinemia (WM), these single-target drug therapies have limitations in their clinical applications, such as drug resistance. Several alternative strategies have been developed, including the use of dual inhibitors, to maximize the therapeutic potential of these drugs. OBJECTIVE Recently, the pharmacological activity of KIN-8194 was repurposed to serve as a 'dual-target' inhibitor of BTK and Hematopoietic Cell Kinase (HCK). However, the structural dual inhibitory mechanism remains unexplored, hence the aim of this study. METHODS Conducting predictive pharmacokinetic profiling of KIN-8194, as well as demonstrating a comparative structural mechanism of inhibition against the above-mentioned enzymes. RESULTS Our results revealed favourable binding affinities of -20.17 kcal/mol, and -35.82 kcal/mol for KIN-8194 towards HCK and BTK, respectively. Catalytic residues Arg137/174 and Lys42/170 in BTK and Arg303 and Lys75/173/244/247 in HCK were identified as crucial mediators of the dual binding mechanism of KIN-8194, corroborated by high per-residue energy contributions and consistent high-affinity interactions of these residues. Prediction of the pharmacokinetics and physicochemical properties of KIN-8194 further established its inhibitory potential, evidenced by the favourable absorption, metabolism, excretion, and minimal toxicity properties. Structurally, KIN-8194 impacted the stability, flexibility, solvent-accessible surface area, and rigidity of BTK and HCK, characterized by various alterations observed in the bound and unbound structures, which proved enough to disrupt their biological function. CONCLUSION These structural insights provided a baseline for the understanding of the dual inhibitory activity of KIN- 8194. Establishing the cruciality of the interactions between the KIN-8194 and Arg and Lys residues could guide the structure-based design of novel dual BTK/HCK inhibitors with improved therapeutic activities.
Collapse
Affiliation(s)
- Ghazi Elamin
- Department of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Aimen Aljoundi
- Department of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mohamed I Alahmdi
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk, 7149, Saudi Arabia
| | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tabuk University, Tabuk, 71491, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mahmoud E S Soliman
- Department of Pharmaceutical Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| |
Collapse
|
20
|
Peters XQ, Elamin G, Aljoundi A, Alahmdi MI, Abo-Dya NE, Sidhom PA, Tawfeek AM, Ibrahim MAA, Soremekun O, Soliman MES. Therapeutic Path to Triple Knockout: Investigating the Pan-inhibitory Mechanisms of AKT, CDK9, and TNKS2 by a Novel 2-phenylquinazolinone Derivative in Cancer Therapy- An In-silico Investigation Therapy. Curr Pharm Biotechnol 2024; 25:1288-1303. [PMID: 37581526 DOI: 10.2174/1389201024666230815145001] [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: 02/27/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Blocking the oncogenic Wnt//β-catenin pathway has of late been investigated as a viable therapeutic approach in the treatment of cancer. This involves the multi-targeting of certain members of the tankyrase-kinase family; Tankyrase 2 (TNKS2), Protein Kinase B (AKT), and Cyclin- Dependent Kinase 9 (CDK9), which propagate the oncogenic Wnt/β-catenin signalling pathway. METHODS During a recent investigation, the pharmacological activity of 2-(4-aminophenyl)-7-chloro- 3H-quinazolin-4-one was repurposed to serve as a 'triple-target' inhibitor of TNKS2, AKT and CDK9. Yet, the molecular mechanism that surrounds its multi-targeting activity remains unanswered. As such, this study aims to explore the pan-inhibitory mechanism of 2-(4-aminophenyl)-7-chloro-3H-quinazolin- 4-one towards AKT, CDK9, and TNKS2, using in silico techniques. RESULTS Results revealed favourable binding affinities of -34.17 kcal/mol, -28.74 kcal/mol, and -27.30 kcal/mol for 2-(4-aminophenyl)-7-chloro-3H-quinazolin-4-one towards TNKS2, CDK9, and AKT, respectively. Pan-inhibitory binding of 2-(4-aminophenyl)-7-chloro-3H-quinazolin-4-one is illustrated by close interaction with specific residues on tankyrase-kinase. Structurally, 2-(4-aminophenyl)-7-chloro- 3H-quinazolin-4-one had an impact on the flexibility, solvent-accessible surface area, and stability of all three proteins, which was illustrated by numerous modifications observed in the unbound as well as the bound states of the structures, which evidenced the disruption of their biological function. Prediction of the pharmacokinetics and physicochemical properties of 2-(4-aminophenyl)-7-chloro-3H-quinazolin-4- one further established its inhibitory potential, evidenced by the favourable absorption, metabolism, excretion, and minimal toxicity properties. CONCLUSION The following structural insights provide a starting point for understanding the paninhibitory activity of 2-(4-aminophenyl)-7-chloro-3H-quinazolin-4-one. Determining the criticality of the interactions that exist between the pyrimidine ring and catalytic residues could offer insight into the structure-based design of innovative tankyrase-kinase inhibitors with enhanced therapeutic effects.
Collapse
Affiliation(s)
- Xylia Q Peters
- Department of Pharmaceutical Science, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Ghazi Elamin
- Department of Pharmaceutical Science, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Aimen Aljoundi
- Department of Pharmaceutical Science, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mohamed Issa Alahmdi
- Department of Pharmaceutical Science, University of Tabuk, Tabuk, 7149, Saudi Arabia
| | - Nader E Abo-Dya
- Department of Pharmaceutical Science, University of Tabuk, Tabuk, 7149, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Peter A Sidhom
- Department of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Ahmed M Tawfeek
- Chemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mahmoud A A Ibrahim
- Department of Pharmaceutical Science, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
- Chemistry Department, Computational Chemistry Laboratory, Faculty of Science, Minia University, Minia, 61519, Egypt
| | - Opeyemi Soremekun
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Mahmoud E S Soliman
- Department of Pharmaceutical Science, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| |
Collapse
|
21
|
Cazacu N, Stan DL, Târcă R, Chilom CG. Binding of flavonoids to yeast aldehyde dehydrogenase: a molecular mechanism and computational approach. J Biomol Struct Dyn 2023; 41:11247-11254. [PMID: 36571489 DOI: 10.1080/07391102.2022.2160820] [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: 10/21/2022] [Accepted: 12/15/2022] [Indexed: 12/27/2022]
Abstract
The interaction of three flavonoids, apigenin, fisetin and quercetin with yeast aldehyde dehydrogenase, ALDH was studied by spectroscopic and molecular docking methods. A combination of both static and dynamic processes interaction mechanism for the binding of flavonoids with ALDH was found. The interaction takes place with moderate binding and the interaction was driven by hydrophobic contacts. The microenvironments of the fluorescent amino acids changed upon flavonoids binding. The distances between ALDH and flavonoids determined by Förster Resonant Energy Transfer (FRET) confirmed the results obtained by fluorescence. The structure of ALDH against thermal denaturation was stabilized by apigenin and destabilized by fisetin and quercetin. Molecular docking simulation showed that all flavonoids bind to the same site of ALDH and confirmed the moderate binding straight found in fluorescence.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nicoleta Cazacu
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| | - Diana L Stan
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| | - Raluca Târcă
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| | - Claudia G Chilom
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| |
Collapse
|
22
|
Nhieu J, Miller MC, Lerdall TA, Mayo KH, Wei LN. Molecular basis for cellular retinoic acid-binding protein 1 in modulating CaMKII activation. Front Mol Biosci 2023; 10:1268843. [PMID: 37822422 PMCID: PMC10562560 DOI: 10.3389/fmolb.2023.1268843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction: Cellular retinoic acid (RA)-binding protein 1 (CRABP1) is a highly conserved protein comprised of an anti-parallel, beta-barrel, and a helix-turn-helix segment outside this barrel. Functionally, CRABP1 is thought to bind and sequester cytosolic RA. Recently, CRABP1 has been established as a major mediator of rapid, non-genomic activity of RA in the cytosol, referred to as "non-canonical" activity. Previously, we have reported that CRABP1 interacts with and dampens the activation of calcium-calmodulin (Ca2+-CaM)-dependent kinase 2 (CaMKII), a major effector of Ca2+ signaling. Through biophysical, molecular, and cellular assays, we, herein, elucidate the molecular and structural mechanisms underlying the action of CRABP1 in dampening CaMKII activation. Results: We identify an interaction surface on CRABP1 for CaMKII binding, located on the beta-sheet surface of the barrel, and an allosteric region within the helix segment outside the barrel, where both are important for interacting with CaMKII. Molecular studies reveal that CRABP1 preferentially associates with the inactive form of CaMKII, thereby dampening CaMKII activation. Alanine mutagenesis of residues implicated in the CaMKII interaction results in either a loss of this preference or a shift of CRABP1 from associating with the inactive CaMKII to associating with the active CaMKII, which corresponds to changes in CRABP1's effect in modulating CaMKII activation. Conclusions: This is the first study to elucidate the molecular and structural basis for CRABP1's function in modulating CaMKII activation. These results further shed insights into CRABP1's functional involvement in multiple signaling pathways, as well as its extremely high sequence conservation across species and over evolution.
Collapse
Affiliation(s)
- Jennifer Nhieu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle C. Miller
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Thomas A. Lerdall
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kevin H. Mayo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
23
|
Sebők-Nagy K, Kóta Z, Kincses A, Fazekas ÁF, Dér A, László Z, Páli T. Spin-Label Electron Paramagnetic Resonance Spectroscopy Reveals Effects of Wastewater Filter Membrane Coated with Titanium Dioxide Nanoparticles on Bovine Serum Albumin. Molecules 2023; 28:6750. [PMID: 37836593 PMCID: PMC10574081 DOI: 10.3390/molecules28196750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
The accumulation of proteins in filter membranes limits the efficiency of filtering technologies for cleaning wastewater. Efforts are ongoing to coat commercial filters with different materials (such as titanium dioxide, TiO2) to reduce the fouling of the membrane. Beyond monitoring the desired effect of the retention of biomolecules, it is necessary to understand what the biophysical changes are in water-soluble proteins caused by their interaction with the new coated filter membranes, an aspect that has received little attention so far. Using spin-label electron paramagnetic resonance (EPR), aided with native fluorescence spectroscopy and dynamic light scattering (DLS), here, we report the changes in the structure and dynamics of bovine serum albumin (BSA) exposed to TiO2 (P25) nanoparticles or passing through commercial polyvinylidene fluoride (PVDF) membranes coated with the same nanoparticles. We have found that the filtering process and prolonged exposure to TiO2 nanoparticles had significant effects on different regions of BSA, and denaturation of the protein was not observed, neither with the TiO2 nanoparticles nor when passing through the TiO2-coated filter membranes.
Collapse
Affiliation(s)
- Krisztina Sebők-Nagy
- Institute of Biophysics, Biological Research Centre Szeged, 6726 Szeged, Hungary; (K.S.-N.); (Z.K.); (A.K.); (A.D.)
| | - Zoltán Kóta
- Institute of Biophysics, Biological Research Centre Szeged, 6726 Szeged, Hungary; (K.S.-N.); (Z.K.); (A.K.); (A.D.)
| | - András Kincses
- Institute of Biophysics, Biological Research Centre Szeged, 6726 Szeged, Hungary; (K.S.-N.); (Z.K.); (A.K.); (A.D.)
| | - Ákos Ferenc Fazekas
- Department of Biosystems Engineering, Faculty of Engineering, University of Szeged, 6725 Szeged, Hungary; (Á.F.F.); (Z.L.)
| | - András Dér
- Institute of Biophysics, Biological Research Centre Szeged, 6726 Szeged, Hungary; (K.S.-N.); (Z.K.); (A.K.); (A.D.)
| | - Zsuzsanna László
- Department of Biosystems Engineering, Faculty of Engineering, University of Szeged, 6725 Szeged, Hungary; (Á.F.F.); (Z.L.)
| | - Tibor Páli
- Institute of Biophysics, Biological Research Centre Szeged, 6726 Szeged, Hungary; (K.S.-N.); (Z.K.); (A.K.); (A.D.)
| |
Collapse
|
24
|
Cazacu N, Chilom CG. Modulation of the structural and functional properties of α1-antitrypsin by interaction with flavonoid luteolin. J Biomol Struct Dyn 2023; 41:7884-7891. [PMID: 36184736 DOI: 10.1080/07391102.2022.2127909] [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: 07/28/2022] [Accepted: 09/15/2022] [Indexed: 10/07/2022]
Abstract
α1-antitrypsin (A1AT) is a circulating serine protease inhibitor and an acute phase reactant, the deficiency of which can lead to liver failure and chronic lung disease. Flavonoid treatment may induce changes in α1-antitrypsin production in some human cells. The purpose of this study is to investigate the properties of the A1AT protein that interacts with the flavonoid luteolin, which exhibits numerous properties, including antioxidant properties. For this purpose, multi-spectroscopic (UV-Vis spectroscopy, fluorescence and FRET) methods and molecular docking were used. The intrinsic fluorescence of A1AT was quenched by luteolin through a static mechanism. Luteolin binds to one site of the A1AT protein, with a moderate binding constant, and the binding process was driven by entropy and hydrophobic interactions. Hydrophobicity around Trp decreased as a result of luteolin binding to the A1AT site and FRET occurred at a distance of 3.11 nm. Under the action of temperature, the stability of A1AT structure was decreased by the presence of luteolin. Molecular docking confirmed that luteolin binds to one site, with a moderate affinity. The results would give a better understanding of the functional changes that occurred in the structure of A1AT induced by luteolin binding, which may have implications in the field of pharmaceutical research.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nicoleta Cazacu
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| | - Claudia G Chilom
- Faculty of Physics, University of Bucharest, Măgurele, Ilfov, Romania
| |
Collapse
|
25
|
Alonso SDV, González Flecha FL. Fifty years of biophysics in Argentina. Biophys Rev 2023; 15:431-438. [PMID: 37681102 PMCID: PMC10480372 DOI: 10.1007/s12551-023-01114-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 09/09/2023] Open
Abstract
In 1972, a group of young Argentinean scientists nucleated in the so-called Membrane Club constituted the Biophysical Society of Argentina (SAB). Over the years, this Society has grown and embraced new areas of research and emerging technologies. In this commentary, we provide an overview of the early stages of biophysics development in Argentina and highlight some of the notable achievements made during the past five decades. The SAB Annual Meetings have been a platform for intense scientific discussions, and the Society has fostered numerous international connections, becoming a hallmark of SAB activities over these 50 years. Initially centered on membrane biophysics, SAB focus has since expanded to encompass diverse fields such as molecular, cellular, and systems biophysics.
Collapse
Affiliation(s)
- Silvia del V. Alonso
- Laboratorio de Bio-Nanotecnología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
- Grupo de Biología Estructural y Biotecnología (GBEyB), IMBICE (CONICET CCT-La Plata), La Plata, Argentina
| | - F. Luis González Flecha
- Laboratorio de Biofísica Molecular, Instituto de Química y Fisicoquímica Biológicas, Universidad de Buenos Aires – CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
Hocq R, Bottone S, Gautier A, Pflügl S. A fluorescent reporter system for anaerobic thermophiles. Front Bioeng Biotechnol 2023; 11:1226889. [PMID: 37476481 PMCID: PMC10355840 DOI: 10.3389/fbioe.2023.1226889] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
Owing to their inherent capacity to make invisible biological processes visible and quantifiable, fluorescent reporter systems have numerous applications in biotechnology. For classical fluorescent protein systems (i.e., GFP and derivatives), chromophore maturation is O2-dependent, restricting their applications to aerobic organisms. In this work, we pioneered the use of the oxygen-independent system FAST (Fluorescence Activating and absorption Shifting tag) in the thermophilic anaerobe Thermoanaerobacter kivui. We developed a modular cloning system that was used to easily clone a library of FAST expression cassettes in an E. coli-Thermoanaerobacter shuttle plasmid. FAST-mediated fluorescence was then assessed in vivo in T. kivui, and we observed bright green and red fluorescence for cells grown at 55°C. Next, we took advantage of this functional reporter system to characterize a set of homologous and heterologous promoters by quantifying gene expression, expanding the T. kivui genetic toolbox. Low fluorescence at 66°C (Topt for T. kivui) was subsequently investigated at the single-cell level using flow cytometry and attributed to plasmid instability at higher temperatures. Adaptive laboratory evolution circumvented this issue and drastically enhanced fluorescence at 66°C. Whole plasmid sequencing revealed the evolved strain carried functional plasmids truncated at the Gram-positive origin of replication, that could however not be linked to the increased fluorescence displayed by the evolved strain. Collectively, our work demonstrates the applicability of the FAST fluorescent reporter systems to T. kivui, paving the way for further applications in thermophilic anaerobes.
Collapse
Affiliation(s)
- Rémi Hocq
- Institute of Chemical, Environmental and Bioscience Engineering, Technische Universität Wien, Vienna, Austria
- Christian Doppler Laboratory for Optimized Expression of Carbohydrate-Active Enzymes, Institute of Chemical, Environmental and Bioscience Engineering, Technische Universität Wien, Vienna, Austria
| | - Sara Bottone
- Laboratoire des Biomolécules (LBM), Centre National de la Recherche Scientifique (CNRS), Sorbonne Université, École Normale Supérieure, Université PSL, Paris, France
- Institut Universitaire de France, Paris, France
| | - Arnaud Gautier
- Laboratoire des Biomolécules (LBM), Centre National de la Recherche Scientifique (CNRS), Sorbonne Université, École Normale Supérieure, Université PSL, Paris, France
- Institut Universitaire de France, Paris, France
| | - Stefan Pflügl
- Institute of Chemical, Environmental and Bioscience Engineering, Technische Universität Wien, Vienna, Austria
- Christian Doppler Laboratory for Optimized Expression of Carbohydrate-Active Enzymes, Institute of Chemical, Environmental and Bioscience Engineering, Technische Universität Wien, Vienna, Austria
| |
Collapse
|
27
|
Kang J, Seshadri M, Cupp-Sutton KA, Wu S. Toward the analysis of functional proteoforms using mass spectrometry-based stability proteomics. FRONTIERS IN ANALYTICAL SCIENCE 2023; 3:1186623. [PMID: 39072225 PMCID: PMC11281393 DOI: 10.3389/frans.2023.1186623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Functional proteomics aims to elucidate biological functions, mechanisms, and pathways of proteins and proteoforms at the molecular level to examine complex cellular systems and disease states. A series of stability proteomics methods have been developed to examine protein functionality by measuring the resistance of a protein to chemical or thermal denaturation or proteolysis. These methods can be applied to measure the thermal stability of thousands of proteins in complex biological samples such as cell lysate, intact cells, tissues, and other biological fluids to measure proteome stability. Stability proteomics methods have been popularly applied to observe stability shifts upon ligand binding for drug target identification. More recently, these methods have been applied to characterize the effect of structural changes in proteins such as those caused by post-translational modifications (PTMs) and mutations, which can affect protein structures or interactions and diversify protein functions. Here, we discussed the current application of a suite of stability proteomics methods, including thermal proteome profiling (TPP), stability of proteomics from rates of oxidation (SPROX), and limited proteolysis (LiP) methods, to observe PTM-induced structural changes on protein stability. We also discuss future perspectives highlighting the integration of top-down mass spectrometry and stability proteomics methods to characterize intact proteoform stability and understand the function of variable protein modifications.
Collapse
Affiliation(s)
- Ji Kang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Meena Seshadri
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Kellye A. Cupp-Sutton
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Si Wu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| |
Collapse
|
28
|
Wang Y, Aoki S, Nara K, Kikuchi Y, Jiao Z, Hasebe Y. Shield, Anchor, and Adhesive Roles of Methylene Blue in Tyrosinase Adsorbed on Carbon Felt for a Flow Injection Amperometric Enzyme Biosensor for Phenolic Substrates and Inhibitors. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:4676-4691. [PMID: 36961887 DOI: 10.1021/acs.langmuir.2c03483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Methylene blue (MB) acted as a stabilizer for preventing surface-induced denaturation of tyrosinase (TYR) adsorbed on a carbon felt (CF) surface, which is based on shield and anchor roles preventing the unfavorable conformational change of TYR on the hydrophobic CF surface. Furthermore, MB acted as an effective adhesive for TYR immobilization on CF. The resulting TYR and MB coadsorbed CF (TYR/MB-CF) worked as an excellent working electrode unit in an electrochemical detector in a flow injection amperometric biosensor, which allowed highly sensitive consecutive determination of not only TYR substrates but also competitive inhibitors. Simultaneous adsorption of TYR and MB from their mixed solution was much useful as compared with step-wise separated adsorption of TYR on the MB-adsorbed CF, which suggests that the binding interaction of MB with TYR in the solution phase is important for this phenomenon. Fluorescence and UV-vis spectroscopy revealed that not only electrostatic forces between the cationic MB and anionic amino acid residues of TYR but also hydrophobic interactions via the phenothiazine ring of MB play a principal binding driving force of MB with TYR at the surface of the TYR molecules. Synchronous fluorescence, three-dimensional fluorescence, and circular dichroism (CD) spectroscopy clarified that the conformation and the secondary structure of TYR slightly changed upon the MB binding, implying that MB binding leads to the modification of the original intramolecular bonding in part.
Collapse
Affiliation(s)
- Yue Wang
- School of Chemical Engineering, University of Science and Technology LiaoNing, Anshan, LiaoNing 114501, China
| | - Shiori Aoki
- Department of Life Science and Green Chemistry, Graduate School of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
| | - Kazuyuki Nara
- Department of Life Science and Green Chemistry, Faculty of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
| | - Yugo Kikuchi
- Department of Life Science and Green Chemistry, Faculty of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
| | - Zeting Jiao
- Department of Life Science and Green Chemistry, Graduate School of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
| | - Yasushi Hasebe
- Department of Life Science and Green Chemistry, Graduate School of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
- Department of Life Science and Green Chemistry, Faculty of Engineering, Saitama Institute of Technology, 1690, Fusaiji, Fukaya, Saitama 369-0293, Japan
| |
Collapse
|
29
|
Huo X, Liu H, Wang S, Yin S, Yin Z. The inhibitory effect and mechanism of small molecules on acetic anhydride-induced BSA acetylation and aggregation. Colloids Surf B Biointerfaces 2023; 225:113265. [PMID: 36931043 DOI: 10.1016/j.colsurfb.2023.113265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Protein acetylation is a significant post-translational modification, and hyperacetylation results in amyloid aggregation, which is closely related to neurodegenerative diseases (Alzheimer's disease, Huntington's disease, and so on). Therefore, it is significant to inhibit the hyperacetylation of proteins and their induced aggregation. In the present study, we aimed to explore the anti-acetylation and anti-amyloid properties of five small molecules (gallic acid, menadione, resveratrol, apigenin, and quercetin) in the process of acetic anhydride-induced protein hyperacetylation and its aggregation. Optical detection methods, such as SDS-PAGE, inverted fluorescence microscopy, and endogenous fluorescence spectroscopy, were used to investigate the effects of small molecules on protein acetylation, aggregation, and structure. In addition, fluorescence quenching and molecular docking techniques were used to explore the relationship between small molecules and acetylation. The results showed that gallic acid (200 μM), menadione (100 μM), quercetin (40 μM), resveratrol (5 μM), and apigenin (20 μM) (unmodified rates were 61.12 %, 67.76 %, 65.11 %, 62.66 %, and 67.81 %, respectively) had strong inhibitory effects on acetylation, and there was no significant difference (P < 0.05). In addition, gallic acid (200 μM), menadione (100 μM), and resveratrol (5 μM) (inhibition rates of 29.89 %, 26.53 %, and 26.09 %, respectively) had more substantial inhibitory effects on protein aggregation, indicating that the five small molecules could inhibit acetic anhydride-induced hyperacetylation and protein aggregation. The underlying mechanism might be that it could inhibit hyperacetylation and resist amyloid aggregation by interacting with proteins to occupy acetylation sites. Collectively, our findings showed that gallic acid, menadione, and resveratrol could potentially prevent and treat neurodegenerative diseases, such as Alzheimer's disease, by inhibiting acetylation and acetylation-induced aggregation.
Collapse
Affiliation(s)
- Xingli Huo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Huijun Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shengjie Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shanmei Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zongning Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
30
|
Zahirul Kabir M, Tayyab H, Erkmen C, Kurbanoglu S, Mohamad SB, Uslu B. Characterization of Climbazole-Bovine serum albumin interaction by experimental and in silico approaches. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 288:122197. [PMID: 36470090 DOI: 10.1016/j.saa.2022.122197] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/08/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Interactive association of an antifungal drug, climbazole (CBZ) with the carrier protein in bovine circulation, bovine serum albumin (BSA) was explored by fluorescence and absorption spectroscopy along with in silico techniques. The fluorescence and absorption spectral alterations of the protein upon addition of CBZ affirmed the complex foration between CBZ and BSA. The inverse temperature dependence behaviour of the KSV values as well as the hyperchromic result of the protein's absorption signals characterized CBZ-triggered quenching of BSA fluorescence as the static quenching. A weak binding affinity (Ka = 3.12-1.90-× 103 M-1) was reported towards the CBZ-BSA association process. Interpretation of thermodynamic data (entropy change = +14.68 J mol-1 K-1 and enthalpy change = -15.07 kJ mol-1) and in silico analyses anticipated that hydrophobic forces, van der Waals forces and hydrogen bonds were the key intermolecular forces in the complex stabilization. Inclusion of CBZ to BSA produced microenvironmental perturbations around Tyr and Trp residues, and also significantly defended temperature-induced destabilization of BSA. The binding locus of CBZ was detected in the proximity of Sudlow's sites I (subdomain IIA) and II (subdomain IIIA) of BSA, exhibiting greater preference towards site II, as revealed by competitive site-marker displacement investigations and in silico analysis. The stability of the CBZ-BSA complex was further validated by the molecular dynamics simulation assessments.
Collapse
Affiliation(s)
- Md Zahirul Kabir
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey
| | - Hafsa Tayyab
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Cem Erkmen
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey; Ankara University, The Graduate School of Health Sciences, 06110, Ankara, Turkey
| | - Sevinc Kurbanoglu
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey
| | - Saharuddin B Mohamad
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia; Centre of Research for Computational Sciences and Informatics for Biology, Bioindustry, Environment, Agriculture and Healthcare, University of Malaya, Kuala Lumpur, Malaysia
| | - Bengi Uslu
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560, Ankara, Turkey.
| |
Collapse
|
31
|
Zhang Y, Wang Y, Dong Y, Zhang Z, Hasebe Y, Zhu J, Liu Z, Gao E. Effect of Acridine Orange on Improving the Electrochemical Performance of Tyrosinase Adsorbed Sulfide Minerals Based Catechol Biosensor. ChemistrySelect 2023. [DOI: 10.1002/slct.202202444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Yan Zhang
- School of Chemical Engineering University of Science and Technology Liaoning 185 Qianshan Middle Road, High-tech Zone Anshan Liaoning 114051 China
| | - Yue Wang
- School of Chemical Engineering University of Science and Technology Liaoning 185 Qianshan Middle Road, High-tech Zone Anshan Liaoning 114051 China
| | - Yan Dong
- School of Chemical Engineering University of Science and Technology Liaoning 185 Qianshan Middle Road, High-tech Zone Anshan Liaoning 114051 China
| | - Zhiqiang Zhang
- School of Chemical Engineering University of Science and Technology Liaoning 185 Qianshan Middle Road, High-tech Zone Anshan Liaoning 114051 China
| | - Yasushi Hasebe
- Department of Life Science and Green Chemistry Faculty of Engineering Saitama Institute of Technology 1690 Fusaiji Fukaya Saitama 369-0293 Japan
| | - Jianmin Zhu
- Oxiranchem Holding Group Inc. No. 29 Donghuan Road, Hongwei District Liaoyang Liaoning China
| | - Zhaobin Liu
- Oxiranchem Holding Group Inc. No. 29 Donghuan Road, Hongwei District Liaoyang Liaoning China
| | - Enjun Gao
- School of Chemical Engineering University of Science and Technology Liaoning 185 Qianshan Middle Road, High-tech Zone Anshan Liaoning 114051 China
| |
Collapse
|
32
|
Hu G, Xi G, Yan H, Gao Z, Wu Z, Lu Z, Tu J. A molecular dynamics investigation of Taq DNA polymerase and its complex with a DNA substrate using a solid-state nanopore biosensor. Phys Chem Chem Phys 2022; 24:29977-29987. [PMID: 36472131 DOI: 10.1039/d2cp03993a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Proteins have a small volume difference by the diversity of amino acids, which make protein detection and identification a great challenge. Solid-state nanopore as label-free biosensors has attracted attention with high sensitivity. In this work, we investigated the Taq DNA polymerase before and after combining it with a DNA substrate on a solid-state nanopore through molecular dynamics. In simulation, we analyzed the contribution source of nanopore current blockage. In addition to considering the traditional physical exclusion volume model, the non-covalent interaction between the protein molecules and the pore wall also showed to affect the current blockage in the nanopore. When choosing pores of comparable size to protein molecules, the two states of Taq DNA polymerase produce differentiated non-covalent interactions with the pore wall, which enhanced the amplitude difference in current blockage. As a result, the two DNA polymerases can be distinguished through the distinct current blockage. However, when applying additional pulling force or increasing the pore size of the nanopore, the differences between the current blockages are not significant enough to distinguish. The introduction of the non-covalent interaction makes it clear to understand the current blockage differences, which guide the mechanism between molecules with similar structures or volumes.
Collapse
Affiliation(s)
- Gang Hu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Guohao Xi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Han Yan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zhuwei Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Ziqing Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Zuhong Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Jing Tu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
33
|
Abubakar M, Kandandapani S, Mohamed SB, Azizah Abd Halim A, Tayyab S. Shedding light on the Molecular Interaction Between the Hepatitis B Virus Inhibitor, Clevudine, and Human Serum Albumin: Thermodynamic, Spectroscopic, Microscopic, and In Silico Analyses. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Elamin G, Aljoundi A, Alahmdi MI, Abo-Dya NE, Soliman MES. Battling BTK mutants with noncovalent inhibitors that overcome Cys481 and Thr474 mutations in Waldenström macroglobulinemia therapy: structural mechanistic insights on the role of fenebrutinib. J Mol Model 2022; 28:355. [PMID: 36222928 DOI: 10.1007/s00894-022-05345-y] [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: 06/21/2022] [Accepted: 10/01/2022] [Indexed: 10/17/2022]
Abstract
Recently, the non-covalent Bruton tyrosine kinase (BTK) inhibitor fenebrutinib was presented as a therapeutic option with strong inhibitory efficacy against a single (C481S) and double (T474S/C481S) BTK variant in the treatment of Waldenström macroglobulinemia (WM). However, the molecular events surrounding its inhibition mechanism towards this variant remain unresolved. Herein, we employed in silico methods such as molecular dynamic simulation coupled with binding free energy estimations to explore the mechanistic activity of the fenebrutinib on (C481S) and (T474S/C481S) BTK variant, at a molecular level. Our investigations reveal that amino acid arginine contributed immensely to the total binding energy, this establishing the cruciality of amino acid residues, Arg132 and Arg156 in (C481S) and Arg99, Arg137, and Arg132 in (T474S/C481S) in the binding of fenebrutinib towards both BTK variants. The structural orientations of fenebrutinib within the respective hydrophobic pockets allowed favorable interactions with binding site residues, accounting for its superior binding affinity by 24.5% and relative high hydrogen bond formation towards (T474S/C481S) when compared with (C481S) BTK variants. Structurally, fenebrutinib impacted the stability, flexibility, and solvent accessible surface area of both BTK variants, characterized by various alterations observed in the bound and unbound structures, which proved enough to disrupt their biological function. Findings from this study, therefore, provide insights into the inhibitory mechanism of fenebrutinib at the atomistic level and reveal its high selectivity towards BTK variants. These insights could be key in designing and developing BTK mutants' inhibitors to treat Waldenström macroglobulinemia (WM).
Collapse
Affiliation(s)
- Ghazi Elamin
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Aimen Aljoundi
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mohamed Issa Alahmdi
- Faculty of Science, Department of Chemistry, University of Tabuk, Tabuk, 7149, Saudi Arabia
| | - Nader E Abo-Dya
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Tabuk University, Tabuk, 71491, Saudi Arabia.,Faculty of Pharmacy, Department of Pharmaceutical Organic Chemistry, Zagazig University, Zagazig, Egypt
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa.
| |
Collapse
|
35
|
Yim WWY, Yamamoto H, Mizushima N. A pulse-chasable reporter processing assay for mammalian autophagic flux with HaloTag. eLife 2022; 11:78923. [PMID: 35938926 PMCID: PMC9385206 DOI: 10.7554/elife.78923] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/08/2022] [Indexed: 01/18/2023] Open
Abstract
Monitoring autophagic flux is necessary for most autophagy studies. The autophagic flux assays currently available for mammalian cells are generally complicated and do not yield highly quantitative results. Yeast autophagic flux is routinely monitored with the GFP-based processing assay, whereby the amount of GFP proteolytically released from GFP-containing reporters (e.g., GFP-Atg8), detected by immunoblotting, reflects autophagic flux. However, this simple and effective assay is typically inapplicable to mammalian cells because GFP is efficiently degraded in lysosomes while the more proteolytically resistant RFP accumulates in lysosomes under basal conditions. Here, we report a HaloTag (Halo)-based reporter processing assay to monitor mammalian autophagic flux. We found that Halo is sensitive to lysosomal proteolysis but becomes resistant upon ligand binding. When delivered into lysosomes by autophagy, pulse-labeled Halo-based reporters (e.g., Halo-LC3 and Halo-GFP) are proteolytically processed to generate Haloligand when delivered into lysosomes by autophagy. Hence, the amount of free Haloligand detected by immunoblotting or in-gel fluorescence imaging reflects autophagic flux. We demonstrate the applications of this assay by monitoring the autophagy pathways, macroautophagy, selective autophagy, and even bulk nonselective autophagy. With the Halo-based processing assay, mammalian autophagic flux and lysosome-mediated degradation can be monitored easily and precisely.
Collapse
Affiliation(s)
- Willa Wen-You Yim
- 1Department of Biochemistry and Molecular Biology, University of Tokyo, Tokyo, Japan
| | | | - Noboru Mizushima
- Department of Biochemistry and Molecular Biology, University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Lyndem S, Gazi R, Belwal VK, Bhatta A, Jana M, Roy AS. Binding of bioactive esculin and esculetin with hen egg white lysozyme: Spectroscopic and computational methods to comprehensively elucidate the binding affinities, interacting forces, and conformational alterations at molecular level. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
37
|
Peters XQ, Agoni C, Soliman MES. Unravelling the Structural Mechanism of Action of 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione in Dual-Targeting Tankyrase 1 and 2: A Novel Avenue in Cancer Therapy. Cell Biochem Biophys 2022; 80:505-518. [PMID: 35637423 DOI: 10.1007/s12013-022-01076-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/21/2022] [Indexed: 11/03/2022]
Abstract
Tankyrase (TNKS) belonging to the poly(ADPribose) polymerase family, are known for their multi-functioning capabilities, and play an essential role in the Wnt β-catenin pathway and various other cellular processes. Although showing inhibitory potential at a nanomolar level, the structural dual-inhibitory mechanism of the novel TNKS inhibitor, 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione, remains unexplored. By employing advanced molecular modeling, this study provides these insights. Results of sequence alignments of binding site residues identified conserved residues; GLY1185 and ILE1224 in TNKS-1 and PHE1035 and PRO1034 in TNKS-2 as crucial mediators of the dual binding mechanism of 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione, corroborated by high per-residue energy contributions and consistent high-affinity interactions of these residues. Estimation of the binding free energy of 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione showed estimated total energy of -43.88 kcal/mol and -30.79 kcal/mol towards TNKS-1 and 2, respectively, indicating favorable analogous dual binding as previously reported. Assessment of the conformational dynamics of TNKS-1 and 2 upon the binding of 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione revealed similar structural changes characterized by increased flexibility and solvent assessible surface area of the residues inferring an analogous structural binding mechanism. Insights from this study show that peculiar, conserved residues are the driving force behind the dual inhibitory mechanism of 5-methyl-5-[4-(4-oxo-3H-quinazolin-2-yl)phenyl]imidazolidine-2,4-dione and could aid in the design of novel dual inhibitors of TNKS-1 and 2 with improved therapeutic properties.
Collapse
Affiliation(s)
- Xylia Q Peters
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Clement Agoni
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa.,West African Centre for Computational Analysis, Accra, Ghana
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa.
| |
Collapse
|
38
|
Sanchez Ramirez DO, Tonetti C, Cruz-Maya I, Guarino V, Peila R, Carletto RA, Varesano A, Vineis C. Design of cysteine-S-sulfonated keratin via pH driven processes: Micro-Structural Properties, biocidal activity and in vitro validation. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
39
|
Conde-Giménez M, Galano-Frutos JJ, Galiana-Cameo M, Mahía A, Victor BL, Salillas S, Velázquez-Campoy A, Brito RMM, Gálvez JA, Díaz-de-Villegas MD, Sancho J. Alchemical Design of Pharmacological Chaperones with Higher Affinity for Phenylalanine Hydroxylase. Int J Mol Sci 2022; 23:ijms23094502. [PMID: 35562892 PMCID: PMC9100405 DOI: 10.3390/ijms23094502] [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: 03/24/2022] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 11/17/2022] Open
Abstract
Phenylketonuria (PKU) is a rare metabolic disease caused by variations in a human gene, PAH, encoding phenylalanine hydroxylase (PAH), and the enzyme converting the essential amino acid phenylalanine into tyrosine. Many PKU-causing variations compromise the conformational stability of the encoded enzyme, decreasing or abolishing its catalytic activity, and leading to an elevated concentration of phenylalanine in the blood, which is neurotoxic. Several therapeutic approaches have been developed to treat the more severe manifestations of the disorder, but they are either not entirely effective or difficult to adhere to throughout life. In a search for novel pharmacological chaperones to treat PKU, a lead compound was discovered (compound IV) that exhibited promising in vitro and in vivo chaperoning activity on PAH. The structure of the PAH-IV complex has been reported. Here, using alchemical free energy calculations (AFEC) on the structure of the PAH-IV complex, we design a new generation of compound IV-analogues with a higher affinity for the enzyme. Seventeen novel analogues were synthesized, and thermal shift and isothermal titration calorimetry (ITC) assays were performed to experimentally evaluate their stabilizing effect and their affinity for the enzyme. Most of the new derivatives bind to PAH tighter than lead compound IV and induce a greater thermostabilization of the enzyme upon binding. Importantly, the correspondence between the calculated alchemical binding free energies and the experimentally determined ΔΔGb values is excellent, which supports the use of AFEC to design pharmacological chaperones to treat PKU using the X-ray structure of their complexes with the target PAH enzyme.
Collapse
Affiliation(s)
- María Conde-Giménez
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - Juan José Galano-Frutos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - María Galiana-Cameo
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - Alejandro Mahía
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - Bruno L. Victor
- Coimbra Chemistry Center-Institute of Molecular Sciences (CQC-IMS), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (B.L.V.); (R.M.M.B.)
| | - Sandra Salillas
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
| | - Adrián Velázquez-Campoy
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- CIBER de Enfermedades Hepáticas y Digestivas CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rui M. M. Brito
- Coimbra Chemistry Center-Institute of Molecular Sciences (CQC-IMS), Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal; (B.L.V.); (R.M.M.B.)
| | - José Antonio Gálvez
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain;
| | - María D. Díaz-de-Villegas
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain;
- Correspondence: (M.D.D.-d.-V.); (J.S.)
| | - Javier Sancho
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009 Zaragoza, Spain; (M.C.-G.); (J.J.G.-F.); (M.G.-C.); (A.M.); (S.S.); (A.V.-C.)
- Biocomputation and Complex Systems Physics Institute (BIFI)-GBsC-CSIC Joint Unit, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Correspondence: (M.D.D.-d.-V.); (J.S.)
| |
Collapse
|
40
|
Gupta DN, Rani R, Kokane AD, Ghosh DK, Tomar S, Sharma AK. Characterization of a cytoplasmic 2-Cys peroxiredoxin from Citrus sinensis and its potential role in protection from oxidative damage and wound healing. Int J Biol Macromol 2022; 209:1088-1099. [PMID: 35452700 DOI: 10.1016/j.ijbiomac.2022.04.086] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/02/2022] [Accepted: 04/11/2022] [Indexed: 12/20/2022]
Abstract
In present work, the recombinant cytoplasmic 2-Cys peroxiredoxin from Citrus sinensis (CsPrx) was purified and characterized. The peroxidase activity was examined with different substrates using DTT, a non-physiological electron donor. The conformational studies, in oxidized and reduced states, were performed using circular dichroism (CD) and fluorescence measurement. The CD analysis showed higher α-helical content for reduced state of the protein. The thermal stability studies of CsPrx by Differential Scanning Calorimetry (DSC) showed that oxidized state is more stable as compared to the reduced state of CsPrx. In vitro studies showed that the CsPrx provides a protective shield against ROS and free radicals that participate in the degradation of plasmid DNA. The pre-treatment of 10 μM CsPrx provide almost 100% protection against peroxide-mediated cell killing in the Vero cells. CsPrx showed significant cell proliferation and wound healing properties. The superior morphology of viable cells and wound closure was found at 20 μM CsPrx treated for 12 h. The results demonstrated that CsPrx is a multifaceted protein with a significant role in cell proliferation, wound healing and protection against hydrogen peroxide-induced cellular damage. This could be the first report of a plant peroxiredoxin being characterized for biomedical applications.
Collapse
Affiliation(s)
- Deena Nath Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, India
| | - Ruchi Rani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, India
| | - Amol D Kokane
- Plant Virology Laboratory, ICAR-Central Citrus Research Institute, Nagpur, India
| | - Dilip Kumar Ghosh
- Plant Virology Laboratory, ICAR-Central Citrus Research Institute, Nagpur, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, India
| | - Ashwani Kumar Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247 667, India.
| |
Collapse
|
41
|
Zargar S, Wani TA, Alsaif NA, Khayyat AIA. A Comprehensive Investigation of Interactions between Antipsychotic Drug Quetiapine and Human Serum Albumin Using Multi-Spectroscopic, Biochemical, and Molecular Modeling Approaches. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27082589. [PMID: 35458787 PMCID: PMC9029314 DOI: 10.3390/molecules27082589] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 12/28/2022]
Abstract
Quetiapine (QTP) is a short-acting atypical antipsychotic drug that treats schizophrenia or manic episodes of bipolar disorder. Human serum albumin (HSA) is an essential transport protein that transports hormones and various other ligands to their intended site of action. The interactions of QTP with HSA and their binding mechanism in the HSA-QTP system was studied using spectroscopic and molecular docking techniques. The UV-Vis absorption study shows hyperchromicity in the spectra of HSA on the addition of QTP, suggesting the complex formation and interactions between QTP and HSA. The results of intrinsic fluorescence indicate that QTP quenched the fluorescence of HSA and confirmed the complex formation between HSA and QTP, and this quenching mechanism was a static one. Thermodynamic analysis of the HSA-QTP system confirms the involvement of hydrophobic forces, and this complex formation is spontaneous. The competitive displacement and molecular docking experiments demonstrated that QTP is preferentially bound to HSA subdomain IB. Furthermore, the CD experiment results showed conformational changes in the HSA-QTP system. Besides this, the addition of QTP does not affect the esterase-like activity of HSA. This study will help further understand the credible mechanism of transport and delivery of QTP via HSA and design new QTP-based derivatives with greater efficacy.
Collapse
Affiliation(s)
- Seema Zargar
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh 11451, Saudi Arabia; (S.Z.); (A.I.A.K.)
| | - Tanveer A. Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
- Correspondence:
| | - Nawaf A. Alsaif
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Arwa Ishaq A. Khayyat
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 22452, Riyadh 11451, Saudi Arabia; (S.Z.); (A.I.A.K.)
| |
Collapse
|
42
|
The inhibitory effects of natural antioxidants on protein glycation as well as aggregation induced by methylglyoxal and underlying mechanisms. Colloids Surf B Biointerfaces 2022; 212:112360. [PMID: 35131714 DOI: 10.1016/j.colsurfb.2022.112360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/14/2021] [Accepted: 01/21/2022] [Indexed: 01/12/2023]
Abstract
The non-enzymatic glycation of protein can result in the formation of advanced glycation end-products (AGEs), leading to the deposition of amyloid proteins, and it's essential for the pathogenesis of diabetes complications and amyloid diseases. Reactive dicarbonyl compounds, such as methylglyoxal (MGO), are one of the most reactive glycating agents. Therefore, it's crucially necessary to inhibit protein glycation and aggregation induced by MGO. In the present study, we aimed to systemically investigate the anti-glycation and anti-fibrillization activities of eight natural antioxidants, including apigenin, quercetin (Que), catechin, resveratrol (Res), and gallic acid (GA), L-ascorbic acid (L-AA), limonene, and β-carotene, during MGO-induced protein glycation and aggregation. Furthermore, the underlying mechanisms were clarified. The formation of AGEs and the degree of protein aggregation were characterized by optical detection, flow cytometry, and so on. The results demonstrated that eight selected natural antioxidants could inhibit glycation and protein aggregation induced by MGO via the synergy of scavenging free radicals, capturing MGO, and interacting with proteins, among which GA (300 μM) and Res (15 μM) had higher inhibition rates on both argpyrimidine (specific fluorescent AGEs, 17% and 22%, respectively) and protein amyloid aggregation (42% and 29%, respectively). These findings suggested that antioxidants could act as potential inhibitors of AGEs and glycation-induced protein aggregation, which were expected to become a new strategy for the prevention and treatment of diabetes and amyloid diseases. Besides, these inhibition mechanisms provided valuable insights into the design and development of candidate drugs for the prevention and treatment of AGEs and protein aggregation-related diseases.
Collapse
|
43
|
Naik R, Seetharamappa J. Elucidating the binding mechanism of an antimigraine agent with a model protein: insights from molecular spectroscopic, calorimetric and computational approaches. J Biomol Struct Dyn 2022; 41:3686-3701. [PMID: 35322751 DOI: 10.1080/07391102.2022.2053747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Sumatriptan (SUM), a serotonin activator used to treat migraines and cluster headaches. Molecular spectroscopic methods including fluorescence quenching, time dependent fluorescence, FRET, absorption, circular dichroism, differential scanning calorimetric and computational approaches were employed to unravel the interaction between sumatriptan and bovine serum albumin (BSA). The fluorescence quenching studies suggested the interaction between SUM and BSA with a moderate binding with the binding constant (Kb) in the order of 104. The findings of temperature and time dependent fluorescence quenching studies confirmed the role of static quenching mechanism. Thermodynamic parameters suggested the key role of electrostatic force in the interaction of SUM with BSA. Absorption and CD spectral studies revealed the bioenvironmental changes around the Trp in BSA upon binding of SUM. Calorimetric based thermal denaturation results confirmed that the thermal stability of BSA was improved in the presence of SUM. resulted in the this decreased flexibility of protein chain. Site competitive studies indicated SUM was located in the hydrophobic cavity of site I which was further confirmed by the docking and dynamic simulation studies. Additionally, molecular dynamics simulations inferred the microenvironmental condition around the SUM and the amino acids and forces involved in the binding of SUM with BSA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Roopa Naik
- Department of Chemistry, Karnatak University, Dharwad, Karnataka, India
| | - J Seetharamappa
- Department of Chemistry, Karnatak University, Dharwad, Karnataka, India
| |
Collapse
|
44
|
Binding of α-lipoic acid to human serum albumin: spectroscopic and molecular modeling studies. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02858-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Meena VK, Kumar V, Karalia S, Garima, Sundd M. Ellagic Acid Modulates Uninduced as well as Mutation and Metal-Induced Aggregation of α-Synuclein: Implications for Parkinson's Disease. ACS Chem Neurosci 2021; 12:3598-3614. [PMID: 34506119 DOI: 10.1021/acschemneuro.1c00317] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
α-Synuclein (αS) is an intrinsically disordered protein whose aggregation and deposition in Lewy bodies is involved in the progression of Parkinson's disease (PD) and other related disorders. The aggregation process of αS is also triggered by mutations like A53T and E46K in the SNCA gene and disruption in metal-ion homeostasis. Currently, there is no obviating therapy available in the market that could effectively prevent the progression of the disease. In this backdrop, there exists an emerging need to consider naturally occurring polyphenols and flavonoids as potential therapeutic agents against PD. In this study, we demonstrate the modulatory effect of ellagic acid (EA) against wild-type as well as mutation and metal-induced aggregation of αS. Thioflavin T (ThT) fluorescence assay suggests that EA acts on the nucleation phase of αS fibrillization, thereby increasing the lag phase from 21.33 ± 3.01 to 48.20 ± 5.05 h and reducing the fibrils growth rate from 4.60 ± 2.06 to 0.890 ± 0.36 h-1. 8-Anilino-1-naphthalene sulfonic acid (ANS), Congo red (CR), and intrinsic fluorescence studies indicate that the interaction of EA with αS facilitates the structural changes in the protein that lead to inhibition of fibril formation. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) images illustrate that the size of fibrils diminishes up to 100 nm in the presence of EA. Dot blot and seeding experiments put forward that EA directs the αS aggregation toward off-pathway fibrillization. Our 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay deciphers the role of EA in minimizing the αS fibril-induced toxicity, thereupon leading to an increase in cell viability. Also, EA attenuates both mutations as well as metal-induced αS fibrillization and disaggregates the preexisting fibrils. Additionally, computational studies elucidate that EA preferentially interacts with the N-terminal and NAC domain of αS. Hence, this work reveals the aggregation inhibition mechanism of EA and provides considerable therapeutic interventions against PD and related disorders.
Collapse
Affiliation(s)
- Vinod Kumar Meena
- NMR-II Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Vijay Kumar
- NMR-II Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Shivani Karalia
- NMR-II Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Garima
- NMR-II Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Monica Sundd
- NMR-II Lab, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
46
|
Ehsan M, Wang H, Cecchetti C, Mortensen JS, Du Y, Hariharan P, Nygaard A, Lee HJ, Ghani L, Guan L, Loland CJ, Byrne B, Kobilka BK, Chae PS. Maltose-bis(hydroxymethyl)phenol (MBPs) and Maltose-tris(hydroxymethyl)phenol (MTPs) Amphiphiles for Membrane Protein Stability. ACS Chem Biol 2021; 16:1779-1790. [PMID: 34445864 DOI: 10.1021/acschembio.1c00578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Membrane protein structures provide a fundamental understanding of their molecular actions and are of importance for drug development. Detergents are widely used to solubilize, stabilize, and crystallize membrane proteins, but membrane proteins solubilized in conventional detergents are prone to denaturation and aggregation. Thus, developing novel detergents with enhanced efficacy for protein stabilization remains important. We report herein the design and synthesis of a class of phenol-derived maltoside detergents. Using two different linkers, we prepared two sets of new detergents, designated maltose-bis(hydroxymethyl)phenol (MBPs) and maltose-tris(hydroxymethyl)phenol (MTPs). The evaluation of these detergents with three transporters and two G-protein coupled receptors allowed us to identify a couple of new detergents (MBP-C9 and MTP-C12) that consistently conferred enhanced stability to all tested proteins compared to a gold standard detergent (DDM). Furthermore, the data analysis based on the detergent structures provides key detergent features responsible for membrane protein stabilization that together will facilitate the future design of novel detergents.
Collapse
Affiliation(s)
- Muhammad Ehsan
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, South Korea
| | - Haoqing Wang
- Department of Molecular and Cellular Physiology, Stanford University, California 94305, United States
| | - Cristina Cecchetti
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Jonas S. Mortensen
- Department of Neuroscience, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Yang Du
- Department of Molecular and Cellular Physiology, Stanford University, California 94305, United States
| | - Parameswaran Hariharan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Andreas Nygaard
- Department of Neuroscience, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Ho Jin Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, South Korea
| | - Lubna Ghani
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, South Korea
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Claus J. Loland
- Department of Neuroscience, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Brian K. Kobilka
- Department of Molecular and Cellular Physiology, Stanford University, California 94305, United States
| | - Pil Seok Chae
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, South Korea
| |
Collapse
|
47
|
Yang H, Khan S, Sun A, Bai Q, Cheng H, Akhtari K. Enhancement of interferon gamma stability as an anticancer therapeutic protein against hepatocellular carcinoma upon interaction with calycosin. Int J Biol Macromol 2021; 185:813-820. [PMID: 34186122 DOI: 10.1016/j.ijbiomac.2021.06.159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/13/2021] [Accepted: 06/24/2021] [Indexed: 11/30/2022]
Abstract
The stability of IFN-γ as a therapeutic protein can play a key role on its anticancer effects. Herein, we explored the thermodynamic parameters and conformational stability of IFN-γ in the presence of calycosin, the main active compound of Radix astragali, by different biophysical and theoretical analysis. Afterwards, the improved anticancer effects of IFN-γ-calycosin interaction relative to IFN-γ alone were assessed on hepatocellular carcinoma (HepG2) cell line by MTT and caspase assays. ITC data indicated that upon interaction of calycosin with IFN-γ the binding and thermodynamic parameters were as follows: Kd = 1.9 μM, ΔG° = -32.45 kJ/mol, ΔH° = -11.91 kJ/mol, and TΔS° = 20.54 kJ/mol. ANS/synchronous fluorescence, CD and UV-Vis spectroscopy studies indicated that the interaction between calycosin and IFN-γ caused the folding of the IFN-γ backbone in to a more packed structure with enhanced α-helix content and higher melting temperature (Tm) value. The spectroscopic outcomes were then verified by molecular docking and molecular dynamic analysis. It was also shown that after incubation of the IFN-γ samples at 50 °C for 60 min in the presence of calycosin (5 μM), the IFN-γ-calycosin system showed a significant antiproliferative effects against hepatocellular carcinoma (HepG2) cells through caspase-9/3 activation and this anticancer effect was more pronounced than free IFN-γ. This data may provide useful information about the development of IFN-γ-based therapeutic platforms.
Collapse
Affiliation(s)
- Huiyu Yang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Aimin Sun
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Qian Bai
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.
| | - Haowei Cheng
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Keivan Akhtari
- Department of Physics, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
48
|
Poonan P, Agoni C, Soliman MES. Dual-Knockout of Mutant Isocitrate Dehydrogenase 1 and 2 Subtypes Towards Glioma Therapy: Structural Mechanistic Insights on the Role of Vorasidenib. Chem Biodivers 2021; 18. [PMID: 33982420 DOI: 10.1002/cbdv.202100110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023]
Abstract
Recently, Vorasidenib (AG-881) has been reported as a therapeutic alternative that exerts potent dual inhibitory activity against mIDH1/2 towards the treatment of low-grade glioma. However, structural and dynamic events associated with its dual inhibition mechanism remain unclear. As such, we employ integrative computer-assisted atomistic techniques to provide thorough structural and dynamic insights. Our analysis proved that the dual-targeting ability of AG-881 is mediated by Val255/Val294 within the binding pockets of both mIDH1 and mIDH2 which are shown to elicit a strong intermolecular interaction, thus favoring binding affinity. The structural orientations of AG-881 within the respective hydrophobic pockets allowed favorable interactions with binding site residues which accounted for its high binding free energy of -28.69 kcal/mol and -19.89 kcal/mol towards mIDH1 and mIDH2, respectively. Interestingly, upon binding, AG-881 was found to trigger systemic alterations of mIDH1 and mIDH2 characterized by restricted residue flexibility and a reduction in exposure of residues to the solvent surface area. As a result of these structural alterations, crucial interactions of the mutant enzymes were inhibited, a phenomenon that results in a suppression of the production of oncogenic stimulator 2-HG. Findings therefore provide thorough structural and dynamic insights associated with the dual inhibitory activity of AG-881 towards glioma therapy.
Collapse
Affiliation(s)
- Preantha Poonan
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Clement Agoni
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| |
Collapse
|
49
|
Errasti-Murugarren E, Bartoccioni P, Palacín M. Membrane Protein Stabilization Strategies for Structural and Functional Studies. MEMBRANES 2021; 11:membranes11020155. [PMID: 33671740 PMCID: PMC7926488 DOI: 10.3390/membranes11020155] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
Accounting for nearly two-thirds of known druggable targets, membrane proteins are highly relevant for cell physiology and pharmacology. In this regard, the structural determination of pharmacologically relevant targets would facilitate the intelligent design of new drugs. The structural biology of membrane proteins is a field experiencing significant growth as a result of the development of new strategies for structure determination. However, membrane protein preparation for structural studies continues to be a limiting step in many cases due to the inherent instability of these molecules in non-native membrane environments. This review describes the approaches that have been developed to improve membrane protein stability. Membrane protein mutagenesis, detergent selection, lipid membrane mimics, antibodies, and ligands are described in this review as approaches to facilitate the production of purified and stable membrane proteins of interest for structural and functional studies.
Collapse
Affiliation(s)
- Ekaitz Errasti-Murugarren
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
- Correspondence: (E.E.-M.); (M.P.)
| | - Paola Bartoccioni
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
| | - Manuel Palacín
- Laboratory of Amino acid Transporters and Disease, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Spain;
- CIBERER (Centro Español en Red de Biomedicina de Enfermedades Raras), 28029 Barcelona, Spain
- Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, 08028 Barcelona, Spain
- Correspondence: (E.E.-M.); (M.P.)
| |
Collapse
|
50
|
Minić S, Annighöfer B, Hélary A, Hamdane D, Hui Bon Hoa G, Loupiac C, Brûlet A, Combet S. Effect of Ligands on HP-Induced Unfolding and Oligomerization of β-Lactoglobulin. Biophys J 2020; 119:2262-2274. [PMID: 33129832 DOI: 10.1016/j.bpj.2020.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022] Open
Abstract
To probe intermediate states during unfolding and oligomerization of proteins remains a major challenge. High pressure (HP) is a powerful tool for studying these problems, revealing subtle structural changes in proteins not accessible by other means of denaturation. Bovine β-lactoglobulin (BLG), the main whey protein, has a strong propensity to bind various bioactive molecules such as retinol and resveratrol, two ligands with different affinity and binding sites. By combining in situ HP-small-angle neutron scattering (SANS) and HP-ultraviolet/visible absorption spectroscopy, we report the specific effects of these ligands on three-dimensional conformational and local changes in BLG induced by HP. Depending on BLG concentration, two different unfolding mechanisms are observed in situ under pressures up to ∼300 MPa: either a complete protein unfolding, from native dimers to Gaussian chains, or a partial unfolding with oligomerization in tetramers mediated by disulfide bridges. Retinol, which has a high affinity for the BLG hydrophobic cavity, significantly stabilizes BLG both in three-dimensional and local environments by shifting the onset of protein unfolding by ∼100 MPa. Increasing temperature from 30 to 37°C enhances the hydrophobic stabilization effects of retinol. In contrast, resveratrol, which has a low binding affinity for site(s) on the surface of the BLG, does not induce any significant effect on the structural changes of BLG due to pressure. HP treatment back and forth up to ∼300 MPa causes irreversible covalent oligomerization of BLG. Ab initio modeling of SANS shows that the oligomers formed from the BLG-retinol complex are smaller and more elongated compared to BLG without ligand or in the presence of resveratrol. By combining HP-SANS and HP-ultraviolet/visible absorption spectroscopy, our strategy highlights the crucial role of BLG hydrophobic cavity and opens up new possibilities for the structural determination of HP-induced protein folding intermediates and irreversible oligomerization.
Collapse
Affiliation(s)
- Simeon Minić
- Université Paris-Saclay, Laboratoire Léon-Brillouin, UMR12 CEA-CNRS, CEA-Saclay, Gif-sur-Yvette CEDEX, France.
| | - Burkhard Annighöfer
- Université Paris-Saclay, Laboratoire Léon-Brillouin, UMR12 CEA-CNRS, CEA-Saclay, Gif-sur-Yvette CEDEX, France
| | - Arnaud Hélary
- Université Paris-Saclay, Laboratoire Léon-Brillouin, UMR12 CEA-CNRS, CEA-Saclay, Gif-sur-Yvette CEDEX, France
| | - Djemel Hamdane
- Laboratoire de Chimie des Processus Biologiques, CNRS-UMR 8229, Collège de France, Paris CEDEX 05, France
| | - Gaston Hui Bon Hoa
- National Institute of Health and Medical Research (INSERM), Paris, France
| | - Camille Loupiac
- Université de Bourgogne Franche-Comté, AgroSup Dijon, UMRA 02.102 Procédés Alimentaires et Microbiologiques, Equipe Physico-Chimie des Aliments et du Vin, Dijon, France
| | - Annie Brûlet
- Université Paris-Saclay, Laboratoire Léon-Brillouin, UMR12 CEA-CNRS, CEA-Saclay, Gif-sur-Yvette CEDEX, France
| | - Sophie Combet
- Université Paris-Saclay, Laboratoire Léon-Brillouin, UMR12 CEA-CNRS, CEA-Saclay, Gif-sur-Yvette CEDEX, France.
| |
Collapse
|