1
|
Dasari S, Kalyaanamoorthy S. Impact of Phosphorylation and O-GlcNAcylation on the Binding Affinity of R4 Tau Peptide to Microtubule and Its Conformational Preference upon Dissociation. J Chem Inf Model 2025; 65:1570-1584. [PMID: 39871444 DOI: 10.1021/acs.jcim.4c02109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/29/2025]
Abstract
Tau is a microtubule (MT)-associated protein that binds to and stabilizes the MTs of neurons. Due to its intrinsically disordered nature, it undergoes several post-translational modifications (PTMs) that are intricately linked to both the physiological and pathophysiological roles of Tau. Prior research has shown phosphorylation and O-GlcNAcylation to have contrasting effects on Tau aggregation; however, the precise molecular mechanisms and potential synergistic effects of these modifications remain elusive. In this article, we study the impact of phosphorylation at S352, and S356, as well as the phosphorylation of O-GlcNAcylation at S356, individually and in combination, on the binding of the R4 (336-367) peptide with MTs by performing classical molecular dynamics (MD) simulations. By analyzing the binding free energies of the Tau-MT complex, we found that both individual and combined phosphorylation at S352 and S356 sites decreased the affinity of the R4 peptide toward MT. Surprisingly, O-GlcNAcylation, a likely neuroprotective modification, at S356 also decreased the binding affinity of Tau to MT similar to the single phosphorylation systems (pS352 or pS356) but was observed to maintain major interactions with MT comparable to unmodified R4. Additionally, we investigated the impact of phosphorylation at both sites and the interplay between phosphorylation at S352 and O-GlcNAcylation at S356, which showed that the latter preserved the interactions and affinity of the Tau with MT better than dual phosphorylation, though still not as effectively as single phosphorylation. These findings suggest that O-GlcNAcylation at residue S356 has a moderate destabilizing effect. We also performed replica-exchange MD simulations of the R4 peptide to understand the changes in conformational preferences upon phosphorylation, O-GlcNAcylation, and a combination of both modifications. Both individual and combined phosphorylation of R4 peptide at S352, and S356, sites induced salt-bridge interactions with positively charged side chains of lysine and arginine amino acids. However, O-GlcNAcylation at S356 induced secondary structural changes on the R4 peptide, leading to the formation of a β-sheet structure, consistent with previous experimental observations. Interestingly, simultaneous phosphorylation at S352 and the phosphorylation of O-GlcNAcylation at S356 resulted in conformations promoting salt-bridges and β-sheets. Thus, our study provides atomistic insights into the impact of PTMs on the binding of Tau peptide to MT and its conformational preferences upon dissociation.
Collapse
Affiliation(s)
- Sathish Dasari
- Department of Chemistry, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Subha Kalyaanamoorthy
- Department of Chemistry, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Waterloo Artificial Intelligence Institute, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
2
|
Saxena V, Patil P, Khodke P, Kumbhar BV. Exploring purine analogues as inhibitors against Katanin, a microtubule severing enzyme using molecular modeling approach. Sci Rep 2024; 14:32095. [PMID: 39738711 PMCID: PMC11686324 DOI: 10.1038/s41598-024-83723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/15/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Katanin, a key protein in cellular architecture, plays a crucial role in severing microtubules, which are vital components of the cytoskeleton. Given its central involvement in cell division and proliferation, katanin represents a promising target for therapeutic intervention, particularly in cancer treatment. Inhibiting katanin's function could potentially hinder the uncontrolled growth of cancerous cells, making it an attractive target for novel anti-cancer therapies. Previous studies have shown that purine-based compounds exhibit a strong affinity for microtubule-severing enzymes. In this study, we aim to identify potential purine-type inhibitors of katanin using molecular modeling techniques. A total of 276,280 purine-type compounds from the PubChem database were subjected to structure-based high-throughput virtual screening, followed by ADME prediction, PASS analysis, and molecular docking studies. These efforts led to the identification of two potent compounds: PubChem CID 122589735 and 123629569, which demonstrated strong binding interactions with katanin. Molecular dynamics simulations further revealed that these compounds effectively altered katanin's conformation when compared to ATP. Additionally, binding energy calculations indicated that PubChem CID 122589735 exhibited the strongest binding affinity for katanin, with the binding free energy ranking as follows: 122589735 > 123629569 > ATP. Our findings suggest that the screened compounds, particularly PubChem CID 122589735, hold promise as potential katanin inhibitor. These compounds could play a significant role in the development of new anti-cancer therapies targeting a variety of carcinoma. Future research, including in vitro and in vivo studies, is essential to assess the efficacy and safety of these inhibitors, paving the way for innovative cancer treatments.
Collapse
Affiliation(s)
- Vibhuti Saxena
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Pruthanka Patil
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Purva Khodke
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Vile Parle (West), Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
3
|
Wagh P, Savaliya S, Joshi B, Vyas B, Kuperkar K, Lalan M, Shah P. Discerning computational, in vitro and in vivo investigations of self-assembling empagliflozin polymeric micelles in type-2 diabetes. Drug Deliv Transl Res 2024; 14:3568-3584. [PMID: 39103594 DOI: 10.1007/s13346-024-01658-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 06/24/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Empagliflozin (EMPA) is an SGLT2 inhibitor, a new class of anti-diabetic medication, indicated for treating type-2 diabetes. Its low permeability, poor solubility and bioavailability limits its use in management of diabetes. The study was aimed to formulate EMPA loaded polymeric micelles (PMs) to overcome these obstacles in oral absorption. METHODOLOGY In silico studies-molecular docking, molecular dynamic simulation (MDS), and quantum chemical calculation were employed to study the interaction of EMPA with different polymers. EMPA loaded TPGS polymeric micelles (EMPA-TPGS-PMs) were formulated by direct dissolution method and characterized in terms of surface morphology, entrapment, particle size, in vitro drug release, and in vitro cytotoxicity (HEK293 cells). In vivo pharmacokinetic and pharmacodynamic studies were also performed. RESULTS The results suggested a good interaction between TPGS and EMPA with lowest binding energy compared to other polymers. Further MDS results and DFT calculations validated the stable binding of the complex hence TPGS was selected for further wet lab experiments. The EMPA-TPGS complex displayed lower value of Total energy (T.E.) than its individual components, indicating the overall stability of the complex while, the energy band gap (∆E) value lied between the two individual molecules, signifying the better electron transfer between HOMO and LUMO of the complex. Based on the solubility, entrapment and cytotoxicity studies, 5% TPGS was selected for formulating drug loaded micelles. EMPA-TPGS5-PMs presented a size of 9.008 ± 1.25 nm, Polydispersity index (PDI) of 0.254 ± 0.100, a controlled release behaviour upto 24 h. SEM and AFM images of the nanoformulation suggested spherical particles whereas, DSC, and PXRD studies confirmed the loss of crystallinity of EMPA. A 3.12-folds higher AUC and a greater reduction in blood glucose levels was exhibited by EMPA-TPGS5-PMs in comparison to EMPA-SUSP in mice model. CONCLUSION EMPA-TPGS-PMs has exhibited better bio absorption and therapeutic effectiveness in diabetes treatment. This improved performance would open the possibility of dose reduction, reduced dosing frequency & dose-related side effects, improving pharmaco-economics and thereby improved overall compliance to the patient. However, this translation from bench to bedside would necessitate studies in higher animals and human volunteers.
Collapse
Affiliation(s)
- Priti Wagh
- Department of Pharmaceutics, Maliba Pharmacy College, Uka Tarsadia University, Bardoli-Mahuva Road,At & Po, Tarsadi, Bardoli, Gujarat, 394350, India
| | - Shivani Savaliya
- Department of Pharmaceutics, Maliba Pharmacy College, Uka Tarsadia University, Bardoli-Mahuva Road,At & Po, Tarsadi, Bardoli, Gujarat, 394350, India
| | - Bhrugesh Joshi
- C.G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Tarsadi, Bardoli, Gujarat, 394350, India
| | - Bhavin Vyas
- Department of Pharmacology, Maliba Pharmacy College, Uka Tarsadia University, Tarsadi, Bardoli, Gujarat, 394350, India
| | - Ketan Kuperkar
- Department of Chemistry, Sardar Vallabhbhai National Institute of Technology (SVNIT), Surat, Gujarat, 395007, India
| | - Manisha Lalan
- Parul Institute of Pharmacy and Research, Parul University, Waghodia, Vadodara, Gujarat, 391760, India
| | - Pranav Shah
- Department of Pharmaceutics, Maliba Pharmacy College, Uka Tarsadia University, Bardoli-Mahuva Road,At & Po, Tarsadi, Bardoli, Gujarat, 394350, India.
| |
Collapse
|
4
|
Peng L, Zhang Z, Li Q, Song Z, Yan C, Ling H. Unveiling the multifaceted pathogenesis and therapeutic drugs of Alzheimer's disease: A comprehensive review. Heliyon 2024; 10:e39217. [PMID: 39629139 PMCID: PMC11612466 DOI: 10.1016/j.heliyon.2024.e39217] [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] [Academic Contribution Register] [Received: 03/31/2024] [Revised: 08/02/2024] [Accepted: 10/09/2024] [Indexed: 12/07/2024] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder characterized by the accumulation of β-amyloid (Aβ) plaques and tau phosphorylation-induced neurofibrillary tangles. This review comprehensively summarizes AD pathogenesis and related factors, drawing on a wealth of authoritative reports and research findings. Specifically, we delve into the intricate mechanisms underlying AD pathology, including Aβ deposition, tau protein phosphorylation, cholinergic dysfunction, neuroinflammation, mitochondrial oxidative stress, ferroptosis, imbalance in the gut microbiota, and microRNA dysregulation. We also explored the effects of these factors on the brain, including synaptic damage and cognitive impairment. Moreover, our review highlights the associations between the pathogenesis of AD and inflammatory cytokines in the peripheral blood and cerebrospinal fluid, dysbiosis of the gut microbiota, and changes in microRNA expression. Overall, we provided a systematic and illustrative overview of the pathogenesis and therapeutic drugs for AD, offering help in the prevention and treatment of this condition.
Collapse
Affiliation(s)
- Liting Peng
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhiming Zhang
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, 423000, Hunan, China
| | - Qi Li
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Zhenjiang Song
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| | - Canqun Yan
- The Health Management Center, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongyan Ling
- Department of Physiology, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China
| |
Collapse
|
5
|
Khanal P, Dwivedi PSR, Patil VS, Shetty A, S A, Aga A, R A, Javaid A, Bhandare VV. Barosmin against postprandial hyperglycemia: outputs from computational prediction to functional responses in vitro. J Biomol Struct Dyn 2024; 42:4489-4505. [PMID: 37458811 DOI: 10.1080/07391102.2023.2233631] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2023] [Accepted: 05/28/2023] [Indexed: 05/16/2024]
Abstract
Previously, barosmin has been demonstrated to possess anti-diabetic action. However, its effect to inhibit α-amylase and α-glucosidase, including glucose utilization efficacy, has yet to be revealed. Hence, the current study attempted to assess the efficiency of barosmin in inhibiting the α-amylase, α -glucosidase, and dipeptidyl peptidase 4 enzymes, including glucose uptake efficacy. Molecular docking and simulation were performed using AutoDock Vina and Gromacs respectively followed by gene ontology analysis using the database for annotation, visualization, and integrated discovery. Further, in vitro enzyme inhibitory activities and glucose uptake assay were performed in L6 cell lines. Density functional theory analysis detailed mechanistic insights into the crucial interaction sites of barosmin of which the electron-dense region was prone to nucleophilic attack (O-atoms) whereas hydroxyl groups (-OH) showed affinity for electrophilic attacks. Barosmin showed good binding affinity with α-amylase (-9.2 kcal/mol), α-glucosidase (-10.7 kcal/mol), and dipeptidyl peptidase 4 (-10.0 kcal/mol). Barosmin formed stable nonbonded contacts with active site residues of aforementioned enzymes throughout 200 ns molecular dynamics simulation. Further, it regulated pathway concerned with glucose homeostasis i.e. tumor necrosis factor signaling pathway. In addition, barosmin showed α-amylase (IC50= 95.77 ± 23.33 µg/mL), α-glucosidase (IC50= 68.13 ± 2.95 µg/mL), and dipeptidyl peptidase 4 (IC50= 13.27 ± 1.99 µg/mL) inhibitory activities including glucose uptake efficacy in L6 cell lines (EC50= 12.46 ± 0.90 µg/mL) in the presence of insulin. This study presents the efficacy of the barosmin to inhibit α-amylase and α-glucosidase and glucose uptake efficacy in L6 cell lines via the use of multiple system biology tools and in vitro techniques.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pukar Khanal
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Prarambh S R Dwivedi
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Vishal S Patil
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, India
| | - Ankith Shetty
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Adithya S
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Afra Aga
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Akshith R
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | - Aarif Javaid
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences, Nitte Deemed to be University, Mangalore, India
| | | |
Collapse
|
6
|
Bhattacharya K, Khanal P, Patil VS, Dwivedi PSR, Chanu NR, Chaudhary RK, Deka S, Chakraborty A. Computational pharmacology profiling of borapetoside C against melanoma. J Biomol Struct Dyn 2024; 42:3233-3248. [PMID: 37203884 DOI: 10.1080/07391102.2023.2213333] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/25/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023]
Abstract
Melanoma,also known as a 'black tumor', begins in the melanocytes when cells (that produce pigment) grows out of control. Immunological dysregulation, which raises the risk for multiple illnesses, including melanoma, may be influenced by stress tiggered through viral infection, long term effects of ultraviolet radiation, environmental pollutants etc. Borapetoside C is one of the phytoconstituents from Tinospora crispa, and its biological source has been reported for its antistress property. Network pharmacology and KEGG pathway analysis of borapetoside C-regulated proteins were conducted to identify the hub genes involved in melanoma development. Further, a molecular docking was performed between borapetoside C and targets involved in melanoma. Further, the top 3 complexes were selected based on the binding energy to conduct molecular dynamics simulations to evaluate the stability of ligand-protein complex followed by principal component analysis and dynamic cross-correlation matrix. In addition, borapetoside C was also screened for its pharmacokinetics and toxicity profile. Network Pharmacology studies and KEGG pathway analysis revealed 8 targets involved in melanoma. Molecular docking between borapetoside C and targets involved in melanoma identified 3 complexes with minimum binding i.e. borapetoside C- MAP2K1, MMP9, and EGFR. Further, molecular dynamics simulations showed a stable complex of borapetoside C with MMP9 and EGFR. The present study suggested that borapetoside C may target MMP9 and EGFR to possess an anti-melanoma property. This finding can be useful in developing a novel therapeutic agent against melanoma from a natural source.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kunal Bhattacharya
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
- Royal School of Pharmacy, The Assam Royal Global University, Guwahati, Assam, India
| | - Pukar Khanal
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Vishal S Patil
- KLE College of Pharmacy, KLE Academy of Higher Education and Research, Belagavi, India
| | - Prarambh S R Dwivedi
- Department of Pharmacology, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Nongmaithem Randhoni Chanu
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
- Faculty of Pharmaceutical Science, Assam Downtown University, Guwahati, Assam, India
| | - Raushan Kumar Chaudhary
- Department of Pharmacy Practice, NGSM Institute of Pharmaceutical Sciences (NGSMIPS), Nitte (Deemed to be University), Mangalore, India
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
| | - Arup Chakraborty
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
| |
Collapse
|
7
|
Beerwala FA, Kolambkar SV, Patil VS, Darasaguppe HR, Khatib NA, Bhandare VV, Hegde HV, Roy S. Decoding the alpha-amylase inhibitory activity of Garcinia indica Choisy by computational and experimental studies. SOUTH AFRICAN JOURNAL OF BOTANY 2024; 165:14-29. [DOI: 10.1016/j.sajb.2023.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2025]
|
8
|
Nogales E, Kellogg E. Structure challenges in the multivalency of Tau-microtubule interactions. Cytoskeleton (Hoboken) 2024; 81:53-56. [PMID: 37702417 PMCID: PMC10873104 DOI: 10.1002/cm.21788] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/25/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
Structural studies aiming to visualize the interaction of Tau with microtubules (MTs) face several challenges, the main concerning the fact that Tau has multiple MT-interacting regions. In particular, the four (or three) pseudo-repeats of Tau bind to identical elements along the MT lattice but do it through non-identical residues. In addition, any given Tau molecule can use all its repeats or just one for its engagement with MTs. Finally, the binding of one Tau is not necessarily in register with respect to the next one. The mismatch in the MT and Tau repeats, therefore, challenges conventional modes of image analysis when visualizing these samples using cryo-electron microscopy. This commentary is dedicated to those challenges and ways to circumvent them while aiming for an atomic description of the Tau-tubulin interaction.
Collapse
Affiliation(s)
- Eva Nogales
- Molecular and Cell Biology Department, University of California, Berkeley, CA, USA
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Elizabeth Kellogg
- Structural Biology Department, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
9
|
Joshi BP, Bhandare VV, Vankawala M, Patel P, Patel R, Vyas B, Krishnamurty R. Friedelin, a novel inhibitor of CYP17A1 in prostate cancer from Cassia tora. J Biomol Struct Dyn 2023; 41:9695-9720. [PMID: 36373336 DOI: 10.1080/07391102.2022.2145497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022]
Abstract
In prostate cancer (PC), drugs targeting CYP17A1 have shown great success in regulating PC progression. However, successful drug molecules show adverse side effects and therapeutic resistance in PC. Therefore, we proposed to discover the potent phytochemical-based inhibitor against CYP17A1 using virtual screening. In this study, a phytochemicals library of ∼13800 molecules was selected to screen the best possible inhibitors against CYP17A1. A molecular modelling approach investigated detailed intermolecular interactions, their structural stability, and binding affinity. Further, in vitro and in vivo studies were performed to confirm the anticancer activity of identified potential inhibitor against CYP17A1. Friedelin from Cassia tora (CT) is identified as the best possible inhibitor from the screened library. MD simulation study reveals stable binding of Friedelin to conserved binding pocket of CYP17A1 with higher binding affinity than studied control, that is, Orteronel. Friedelin was tested on hormone-sensitive (22Rv1) and insensitive (DU145) cell lines and the IC50 value was found to be 72.025 and 81.766 µg/ml, respectively. CT extract showed a 25.28% IC50 value against 22Rv1, ∼92.6% increase in late Apoptosis/Necrosis, and three folds decrease in early apoptosis in treated cells compared to untreated cells. Further, animal studies show a marked decrease in prostate weight by 39.6% and prostate index by 36.5%, along with a reduction in serum PSA level by 71.7% and testosterone level by 92.4% compared to the testosterone group, which was further validated with histopathological studies. Thus, we propose Friedelin and CT extract as potential leads, which could be taken further for drug development in PC.[Figure: see text]Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Mahima Vankawala
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Prittesh Patel
- C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Tarsadi, Surat, Gujarat, India
| | - Rajesh Patel
- Bioinformatics and Supercomputer Lab., Department of Biosciences (UGC-SAP-DRS-II & DST-FIST-I), Veer Narmad South Gujarat University, Surat, Gujarat, India
| | - Bhavin Vyas
- Department of Pharmacology, Maliba Pharmacy College, Uka Tarsadia University, Tarsadi, Surat, Gujarat, India
| | - Ramar Krishnamurty
- C. G. Bhakta Institute of Biotechnology, Uka Tarsadia University, Tarsadi, Surat, Gujarat, India
| |
Collapse
|
10
|
Kumari S, Kumar P. Design and Computational Analysis of an MMP9 Inhibitor in Hypoxia-Induced Glioblastoma Multiforme. ACS OMEGA 2023; 8:10565-10590. [PMID: 36969457 PMCID: PMC10035023 DOI: 10.1021/acsomega.3c00441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 01/21/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The main therapeutic difficulties in treating hypoxia-induced glioblastoma multiforme (GBM) are toxicity of current treatments and the resistance brought on by the microenvironment. More effective therapeutic alternatives are urgently needed to reduce tumor lethality. Hence, we screened plant-based natural product panels intending to identify novel drugs without elevating drug resistance. We explored GEO for the hypoxia GBM model and compared hypoxic genes to non-neoplastic brain cells. A total of 2429 differentially expressed genes expressed exclusively in hypoxia were identified. The functional enrichment analysis demonstrated genes associated with GBM, further PPI network was constructed, and biological pathways associated with them were explored. Seven webtools, including GEPIA2.0, TIMER2.0, TCGA-GBM, and GlioVis, were used to validate 32 hub genes discovered using Cytoscape tool in GBM patient samples. Four GBM-specific hypoxic hub genes, LYN, MMP9, PSMB9, and TIMP1, were connected to the tumor microenvironment using TIMER analysis. 11 promising hits demonstrated positive drug-likeness with nontoxic characteristics and successfully crossed blood-brain barrier and ADMET analyses. Top-ranking hits have stable intermolecular interactions with the MMP9 protein according to molecular docking, MD simulation, MM-PBSA, PCA, and DCCM analyses. Herein, we have reported flavonoids, 7,4'-dihydroxyflavan, (3R)-3-(4-hydroxybenzyl)-6-hydroxy-8-methoxy-3,4-dihydro-2H-1-benzopyran, and 4'-hydroxy-7-methoxyflavan, to inhibit MMP9, a novel hypoxia gene signature that could serve as a promising predictor in various clinical applications, including GBM diagnosis, prognosis, and targeted therapy.
Collapse
|
11
|
Man VH, He X, Gao J, Wang J. Phosphorylation of Tau R2 Repeat Destabilizes Its Binding to Microtubules: A Molecular Dynamics Simulation Study. ACS Chem Neurosci 2023; 14:458-467. [PMID: 36669127 PMCID: PMC10032563 DOI: 10.1021/acschemneuro.2c00611] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2022] [Accepted: 01/05/2023] [Indexed: 01/22/2023] Open
Abstract
Phosphorylation, the most popular post-translational modification of tau protein, plays an important role in regulating tau physiological functions. However, aberrant phosphorylation attenuates the binding affinity of tau to a microtubule (MT), resulting in MT destabilization followed by accumulation of neurofibrillary tangles in the brain. There are in total 85 potential phosphorylation sites in a full-length tau protein, and about half of them are abnormal as they occur in tau of Alzheimer's disease (AD) brain only. In this work, we investigated the impact of abnormal Ser289, Ser293, and Ser289/Ser293 phosphorylation on tau R2-MT binding and the conformation of tau R2 using molecular dynamics simulation. We found that the phosphorylation significantly affected R2-MT interaction and reduced the binding affinity of tau R2 peptides to MTs. Free energy decomposition analysis suggested that the post-translational modified residues themselves made a significant contribution to destabilize tau repeat R2-MT binding. Therefore, the phosphorylation may attenuate the binding affinity of tau to MTs. Additionally, the phosphorylation also enhanced helix-coil transition of monomeric R2 peptides, which may result in the acceleration of tau aggregation. Since these phosphorylated sites have not been examined in previous experimental studies, our finding through all-atom molecular dynamics simulations and free energy analysis can inspire experimental scientists to investigate the impact of the phosphorylation on MT binding and aggregation of full-length tau and the pathological roles of the phosphorylation at those sites in AD development through in vitro/in vivo assays.
Collapse
Affiliation(s)
- Viet Hoang Man
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, School of Pharmacy, University of
Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Xibing He
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, School of Pharmacy, University of
Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Jie Gao
- Department
of Neuroscience, The Ohio State University
Wexner Medical Center, Columbus, Ohio43210, United States
| | - Junmei Wang
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, School of Pharmacy, University of
Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| |
Collapse
|
12
|
DasNandy A, Patil VS, Hegde HV, Harish DR, Roy S. Elucidating type 2 diabetes mellitus risk factor by promoting lipid metabolism with gymnemagenin: An in vitro and in silico approach. Front Pharmacol 2022; 13:1074342. [PMID: 36582536 PMCID: PMC9792475 DOI: 10.3389/fphar.2022.1074342] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction: Adipose tissue functions as a key endocrine organ which releases multiple bioactive substances and regulate obesity-linked complications. Dysregulation of adipocyte differentiation, triglyceride metabolism, adipokines production and lipid transport contributes to impaired lipid metabolism resulting in obesity, insulin resistance and type 2 diabetes. Gymnema sylvestre plant is frequently used in Ayurveda for treatment of diabetes and obesity. Gymnemagenin is a major bioactive compound of Gymnema sylvestre. The present study was undertaken to elucidate the role of gymnemagenin in lipid metabolism by in vitro and computational approaches. Methods: A panel of twelve genes viz., Fasn, Lipe, Lpl, Pparg, Plin2, Cidea, Scd1, Adipoq, Lep, Ccl2, Fabp4, and Slc2a4, essential in lipid metabolism were selected and gene expression pattern and triglyceride content were checked in adipocytes (3T3L1 cells) with/without treatment of gymnemagenin by Real time PCR and colorimetric estimation, respectively. Mode of action of gymnemagenin on Pparg and Fabp4 was accomplished by computational studies. Gene set enrichment and network pharmacology were performed by STRING and Cytoscape. Molecular docking was performed by AutoDock vina by POAP pipeline. Molecular dynamics, MM-PBSA were done by Gromacs tool. Results: In vitro study showed that gymnemagenin improved triglyceride metabolism by up regulating the expression of lipase genes viz., Lipe and Lpl which hydrolyse triglyceride. Gymnemagenin also up regulated the expression of anti-inflammatory gene Adipoq. Importantly, gymnemagenin treatment up regulated the expression of Pparg gene and the downstream target genes (Plin2, Cidea, and Scd1) which are associated with adipogenesis. However, gymnemagenin has no effect on expression of Fabp4, codes for a lipid transport protein. In silico study revealed that gymnemagenin targeted 12 genes were modulating 6 molecular pathways involved in diabetes and obesity. Molecular docking and dynamics revealed that gymnemagenin stably bind to active site residue of Pparg and failed to bind to Fabp4 active site compared to its standard molecules throughout 100 ns MD production run. Gymnemagenin scored binding free energy of -177.94 and -25.406 kJ/mol with Pparg and Fabp4, respectively. Conclusion: Gymnemagenin improved lipid metabolism by increasing triglyceride hydrolysis (lipolysis), up regulating the crucial gene of adipogenesis and increasing the expression of anti-inflammatory adipokine proving its therapeutic importance as anti-obesity and anti-diabetic phytocompound.
Collapse
|
13
|
Khanal P, Patil VS, Bhandare VV, Dwivedi PS, Shastry C, Patil B, Gurav SS, Harish DR, Roy S. Computational investigation of benzalacetophenone derivatives against SARS-CoV-2 as potential multi-target bioactive compounds. Comput Biol Med 2022; 146:105668. [PMID: 35667894 PMCID: PMC9135652 DOI: 10.1016/j.compbiomed.2022.105668] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 02/08/2023]
Abstract
Benzalacetophenones, precursors of flavonoids are aromatic ketones and enones and possess the immunostimulant as well as antiviral activities. Thus, benzalacetophenones were screened against the COVID-19 that could be lethal in patients with compromised immunity. We considered ChEBI recorded benzalacetophenone derivative(s) and evaluated their activity against 3C-like protease (3CLpro), papain-like protease (PLpro), and spike protein of SARS-Cov-2 to elucidate their possible role as antiviral agents. The probable targets for each compound were retrieved from DIGEP-Pred at 0.5 pharmacological activity and all the modulated proteins were enriched to identify the probably regulated pathways, biological processes, cellular components, and molecular functions. In addition, molecular docking was performed using AutoDock 4 and the best-identified hits were subjected to all-atom molecular dynamics simulation and binding energy calculations using molecular mechanics Poisson-Boltzmann surface area (MMPBSA). The compound 4-hydroxycordoin showed the highest druglikeness score and regulated nine proteins of which five were down-regulated and four were upregulated. Similarly, enrichment analysis identified the modulation of multiple pathways concerned with the immune system as well as pathways related to infectious and non-infectious diseases. Likewise, 3'-(3-methyl-2-butenyl)-4′-O-β-d-glucopyranosyl-4,2′-dihydroxychalcone with 3CLpro, 4-hydroxycordoin with PLpro and mallotophilippen D with spike protein receptor-binding domain showed highest binding affinity, revealed stable interactions during the simulation, and scored binding free energy of −26.09 kcal/mol, −16.28 kcal/mol, and −39.2 kcal/mol, respectively. Predicted anti-SARS-CoV-2 activities of the benzalacetophenones reflected the requirement of wet lab studies to develop novel antiviral candidates.
Collapse
|
14
|
Feizabadi MS, Castillon VJ. The Effect of Tau and Taxol on Polymerization of MCF7 Microtubules In Vitro. Int J Mol Sci 2022; 23:ijms23020677. [PMID: 35054875 PMCID: PMC8776089 DOI: 10.3390/ijms23020677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2021] [Revised: 12/27/2021] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Overexpression of Tau protein in breast cancer cells is identified as an indicator for potential resistance to taxane-based therapy. As reported findings have been obtained mostly from clinical studies, the undetermined underlying mechanism of such drug resistance needs to be thoroughly explored through comprehensive in vitro evaluations. Tau and Taxol bind to the beta tubulin site in microtubules’ structure. This is of particular interest in breast cancer, as microtubules of these cancer cells are structurally distinct from some other microtubules, such as neuronal microtubules, due to their unique beta tubulin isotype distribution. The observed changes in the in vitro polymerization of breast cancer microtubules, and the different function of some molecular motors along them, leave open the possibility that the drug resistance mechanism can potentially be associated with different responses of these microtubules to Tau and Taxol. We carried out a series of parallel experiments to allow comparison of the in vitro dual effect of Tau and Taxol on the polymerization of MCF7 microtubules. We observed a concentration-dependent demotion-like alteration in the self-polymerization kinetics of Tau-induced MCF7 microtubules. In contrast, microtubules polymerized under the simultaneous effects of Tau and Taxol showed promoted assembly as compared with those observed in Tau-induced microtubules. The analysis of our data obtained from the length of MCF7 microtubules polymerized under the interaction with Tau and Taxol in vitro suggests that the phenomenon known as drug resistance in microtubule-targeted drugs such as Taxol may not be directly linked to the different responses of microtubules to the drug. The effect of the drug may be mitigated due to the simultaneous interactions with other microtubule-associated proteins such as Tau protein. The observed regulatory effect of Tau and Taxol on the polymerization of breast cancer microtubules in vitro points to additional evidence for the possible role of tubulin isotypes in microtubules’ functions.
Collapse
|
15
|
Alıcı H, Demir K. Investigation of the stability and the helix-tail interaction of sCT and its various charged mutants based on comparative molecular dynamics simulations. Chem Phys 2021. [DOI: 10.1016/j.chemphys.2020.111057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/22/2022]
|
16
|
Alıcı H. In silico analysis: structural insights about inter-protofilaments interactions for α-synuclein (50–57) fibrils and its familial mutation. MOLECULAR SIMULATION 2020. [DOI: 10.1080/08927022.2020.1786084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 10/23/2022]
Affiliation(s)
- Hakan Alıcı
- Department of Physics, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
17
|
Kumbhar BV, Bhandare VV. Exploring the interaction of Peloruside-A with drug resistant αβII and αβIII tubulin isotypes in human ovarian carcinoma using a molecular modeling approach. J Biomol Struct Dyn 2020; 39:1990-2002. [DOI: 10.1080/07391102.2020.1745689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/17/2023]
Affiliation(s)
- Bajarang Vasant Kumbhar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | | |
Collapse
|