1
|
Förster CY, Shityakov S, Stavrakis S, Scheper V, Lenarz T. Interplay between noise-induced sensorineural hearing loss and hypertension: pathophysiological mechanisms and therapeutic prospects. Front Cell Neurosci 2025; 19:1523149. [PMID: 40260077 PMCID: PMC12009814 DOI: 10.3389/fncel.2025.1523149] [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: 11/05/2024] [Accepted: 03/21/2025] [Indexed: 04/23/2025] Open
Abstract
More than 5% of the global population suffers from disabling hearing loss, primarily sensorineural hearing loss (SNHL). SNHL is often caused by factors such as vascular disorders, viral infections, ototoxic drugs, systemic inflammation, age-related labyrinthine membrane degeneration, and noise-induced hearing loss (NIHL). NIHL, in particular, leads to changes in blood-labyrinth-barrier (BLB) physiology, increased permeability, and various health issues, including cardiovascular disease, hypertension, diabetes, neurological disorders, and adverse reproductive outcomes. Recent advances in neuromodulation and vector-based approaches offer hope for overcoming biological barriers such as the BLB in the development of innovative treatments. Computational methods, including molecular docking, molecular dynamics simulations, QSAR/QSPR analysis with machine/deep learning algorithms, and network pharmacology, hold potential for identifying drug candidates and optimizing their interactions with BLB transporters, such as the glutamate transporter. This paper provides an overview of NIHL, focusing on its pathophysiology; its impact on membrane transporters, ion channels, and BLB structures; and associated symptoms, comorbidities, and emerging therapeutic approaches. Recent advancements in neuromodulation and vector-based strategies show great promise in overcoming biological barriers such as BLB, facilitating the development of innovative treatment options. The primary aim of this review is to examine NIHL in detail and explore its underlying mechanisms, physiological effects, and cutting-edge therapeutic strategies for its effective management and prevention.
Collapse
Affiliation(s)
- Carola Y. Förster
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint Petersburg, Russia
| | - Stavros Stavrakis
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence “Hearing4all”, German Research Foundation, Hannover, Germany
| |
Collapse
|
2
|
Rezaeianzadeh O, Asghari S, Tajbakhsh M, Khalilpour A, Shityakov S. Synthesis and application of diazenyl sulfonamide-based schiff bases as potential BRCA2 active inhibitors against MCF-7 breast cancer cell line. Sci Rep 2025; 15:6661. [PMID: 39994448 PMCID: PMC11850876 DOI: 10.1038/s41598-025-91113-w] [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: 10/18/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
In this study, a library of novel sulfonamide-based Schiff bases 3a-j was synthesized in high yield (75 to 89%). The FTIR, 1H NMR, and 13C NMR spectroscopic techniques and mass analysis were used to characterize the synthesized compounds. Their anticancer activity was assessed in vitro on the breast cancer (MCF-7) and healthy human breast epithelial (MCF-10 A) cell lines over 48 and 72 h using the MTT assay. Most of the synthesized compounds demonstrated promising activity, with compound 3i showing particularly high efficacy at 48 and 72 h (IC50 = 4.85 ± 0.006 and 4.25 ± 0.009 µM) against the MCF-7 breast cancer cell line. Furthermore, molecular docking studies were performed for compounds 3a-j with the PDB: (3UV7) protein of the breast cancer susceptibility gene 2 (BRCA2). The obtained results revealed that compound 3i has the strongest binding affinity energy (-7.99 kcal/mol), consistent with the obtained experimental data. Additionally, molecular dynamics (MD) simulation assays confirm the formation of a stable 3i-BRCA2 complex with strong binding affinity through the formation of hydrogen bonds. Antioxidant activities were determined by in vitro assay DPPH cation radical activity method. Interestingly, the compound 3j (IC50 = 12.36 ± 0.55 µM) had comparable activity with ascorbic acid (IC50 = 13.58 ± 0.38 µM) in the antioxidant assay. The results of this research could potentially contribute to the development of new therapeutic agents useful in fighting caused by breast cancer.
Collapse
Affiliation(s)
- Olia Rezaeianzadeh
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran
| | - Sakineh Asghari
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran.
| | - Mahmood Tajbakhsh
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, 47416- 95447, Iran
| | - Asieh Khalilpour
- Department of Environmental Health Engineering, Babol University of Medicinal Sciences, Babol, Iran
| | - Sergey Shityakov
- Infochemistry Scientific Center, ITMO University, Lomonosova str. 9, 191002, Saint Petersburg, Russia
| |
Collapse
|
3
|
Isakova AM, Skorb EV, Shityakov S. NeuroClick: Advanced Software for Designing Blood-Brain Barrier-Permeable Drugs Using Click Reaction Simulations. Curr Protoc 2024; 4:e70050. [PMID: 39513559 DOI: 10.1002/cpz1.70050] [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] [Indexed: 11/15/2024]
Abstract
NeuroClick is a software tool designed for the in silico execution of azide-alkyne cycloaddition reactions, commonly known as click reactions. We developed this graphical user interface application to expedite the drug discovery process by generating libraries of 1,2,3-triazole compounds. NeuroClick enables users to input reagent SMILES strings, rapidly generating and screening extensive combinatorial libraries at a pace of 10,000 molecules per minute. The software applies stringent criteria to ensure the relevance and accuracy of the generated compounds, excluding molecules without azide groups or those with multiple reactive functional groups to maintain dataset integrity. NeuroClick incorporates advanced filtering options based on Lipinski's rule of five and blood-brain barrier (BBB) permeability predictors, allowing researchers to identify drug-like molecules with potential central nervous system activity. The software's high-throughput and user-friendly interface significantly enhance the efficiency of early-stage drug development by facilitating the exploration of vast chemical spaces and identifying promising lead compounds for further development. This article provides comprehensive guidance on the installation, usage, and features of NeuroClick, ensuring that users can leverage its full potential in their research endeavors. © 2024 Wiley Periodicals LLC. Basic Protocol: NeuroClick workflow for generating BBB-permeating drugs.
Collapse
Affiliation(s)
- Anastasiia M Isakova
- Laboratory of Cheminformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russian Federation
| | - Ekaterina V Skorb
- Laboratory of Cheminformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russian Federation
| | - Sergey Shityakov
- Laboratory of Cheminformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russian Federation
| |
Collapse
|
4
|
Shityakov S, Förster CY, Skorb E. Comparative in silico analysis of CNS-active molecules targeting the blood-brain barrier choline transporter for Alzheimer's disease therapy. In Silico Pharmacol 2024; 12:71. [PMID: 39099798 PMCID: PMC11291784 DOI: 10.1007/s40203-024-00245-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024] Open
Abstract
This study investigated the blood‒brain barrier (BBB) permeability of the central nervous system (CNS)-active compounds donepezil (DON), methionine (MET), and memantine (MEM) by employing a comprehensive in silico approach. These compounds are of particular interest for Alzheimer's disease (AD) therapy. Rigid-flexible molecular docking simulations indicated favorable binding affinities of all the compounds with BBB-ChT, with DON exhibiting the highest binding affinity (ΔGbind = -10.26 kcal/mol), predominantly mediated by significant hydrophobic interactions. In silico kinetic profiling suggested the stability of the DON/BBB-ChT complex, with ligand release prompted by conformational changes. 3D molecular alignment corroborated a minor conformational shift for DON in its minimal binding energy pose. Predictions indicated that active transport mechanisms notably enhance the brain distribution of donepezil compared to that of MET and MEM. Additionally, DON and MEM exhibited low mutagenic probabilities, while MET was identified as highly mutagenic. Overall, these findings highlight the potential of donepezil for superior BBB penetration, primarily through active transport mechanisms, underscoring the need for further validation through in vitro and in vivo studies for effective AD treatment. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00245-w.
Collapse
Affiliation(s)
- Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint- Petersburg, Russian Federation
| | - Carola Y Förster
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University of Würzburg, 97080 Würzburg, Germany
| | - Ekaterina Skorb
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint- Petersburg, Russian Federation
| |
Collapse
|
5
|
Kumar V, Banerjee A, Roy K. Breaking the Barriers: Machine-Learning-Based c-RASAR Approach for Accurate Blood-Brain Barrier Permeability Prediction. J Chem Inf Model 2024; 64:4298-4309. [PMID: 38700741 DOI: 10.1021/acs.jcim.4c00433] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2024]
Abstract
The intricate nature of the blood-brain barrier (BBB) poses a significant challenge in predicting drug permeability, which is crucial for assessing central nervous system (CNS) drug efficacy and safety. This research utilizes an innovative approach, the classification read-across structure-activity relationship (c-RASAR) framework, that leverages machine learning (ML) to enhance the accuracy of BBB permeability predictions. The c-RASAR framework seamlessly integrates principles from both read-across and QSAR methodologies, underscoring the need to consider similarity-related aspects during the development of the c-RASAR model. It is crucial to note that the primary goal of this research is not to introduce yet another model for predicting BBB permeability but rather to showcase the refinement in predicting the BBB permeability of organic compounds through the introduction of a c-RASAR approach. This groundbreaking methodology aims to elevate the accuracy of assessing neuropharmacological implications and streamline the process of drug development. In this study, an ML-based c-RASAR linear discriminant analysis (LDA) model was developed using a dataset of 7807 compounds, encompassing both BBB-permeable and -nonpermeable substances sourced from the B3DB database (freely accessible from https://github.com/theochem/B3DB), for predicting BBB permeability in lead discovery for CNS drugs. The model's predictive capability was then validated using three external sets: one containing 276,518 natural products (NPs) from the LOTUS database (accessible from https://lotus.naturalproducts.net/download) for data gap filling, another comprising 13,002 drug-like/drug compounds from the DrugBank database (available from https://go.drugbank.com/), and a third set of 56 FDA-approved drugs to assess the model's reliability. Further diversifying the predictive arsenal, various other ML-based c-RASAR models were also developed for comparison purposes. The proposed c-RASAR framework emerged as a powerful tool for predicting BBB permeability. This research not only advances the understanding of molecular determinants influencing CNS drug permeability but also provides a versatile computational platform for the rapid assessment of diverse compounds, facilitating informed decision-making in drug development and design.
Collapse
Affiliation(s)
- Vinay Kumar
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Arkaprava Banerjee
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Kunal Roy
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
6
|
Khalid H, Fareed MM, Dandekar T, Shityakov S. Calcineurin and mTOR inhibitors in kidney transplantation: integrative metamodeling on transplant survival and kidney function. Int Urol Nephrol 2024; 56:1403-1414. [PMID: 37751051 DOI: 10.1007/s11255-023-03754-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/18/2023] [Indexed: 09/27/2023]
Abstract
In our study, we examined the efficacy of mTOR (mammalian target of rapamycin) inhibitors, specifically rapamycin (Rap), compared to calcineurin inhibitors (CNIs) in kidney transplantation. By conducting a comprehensive search across reputable databases (EMBASE, Scopus, PubMed, Cochrane, and Crossref), we gathered data for a six-month post-transplantation period. Our analysis revealed that mTOR inhibitor administration resulted in improved glomerular filtration rate (GFR) and serum creatinine levels. However, it is important to note that the mTOR inhibitor group had a higher incidence of acute rejection after biopsy. Through molecular modeling, we observed that Rap exhibited a superior binding affinity for mTOR compared to CNIs' binding to calcineurin, probably contributing to the transplant rejection. Our meta-analysis supports the cautious use of an optimal mTOR inhibitor in conjunction with careful consideration of clinical features when minimizing CNIs early in the transplantation process. This is because mTOR inhibitors have complementary mechanisms of action, a low nephrotoxicity profile, and favorable outcomes in serum creatinine and GFR, which contribute to improved transplant survival.
Collapse
Affiliation(s)
- Hina Khalid
- Faculty of Life Sciences, Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan.
| | - Muhammad Mazhar Fareed
- School of Science and Engineering, Department of Computer Science, Università degli studi di Verona, Verona, Italy
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russian Federation
| |
Collapse
|
7
|
Isakova AM, Kovalenko AA, Skorb EV, Shityakov S. NeuroClick: software for mimicking click reaction to generate drug-like molecules permeating the blood-brain barrier. Future Med Chem 2024; 16:389-398. [PMID: 38372134 DOI: 10.4155/fmc-2023-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Background: Traditional methods for chemical library generation in virtual screening often impose limitations on the accessible chemical space or produce synthetically irrelevant structures. Incorporating common chemical reactions into generative algorithms could offer significant benefits. Materials & methods: In this study, we developed NeuroClick, a graphical user interface software designed to perform in silico azide-alkyne cycloaddition, a widely utilized synthetic approach in modern medicinal chemistry. Results & conclusion: NeuroClick facilitates the generation and filtering of large combinatorial libraries at a remarkable rate of 10,000 molecules per minute. Moreover, the generated products can be filtered to identify subsets of pharmaceutically relevant compounds based on Lipinski's rule of five and blood-brain barrier permeability prediction. We demonstrate the utility of NeuroClick by generating and filtering several thousand molecules for dopamine D3 receptor ligand screening.
Collapse
Affiliation(s)
- Anastasiia M Isakova
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint Petersburg, Russian Federation
| | - Alexander A Kovalenko
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint Petersburg, Russian Federation
| | - Ekaterina V Skorb
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint Petersburg, Russian Federation
| | - Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint Petersburg, Russian Federation
| |
Collapse
|
8
|
Aaddouz M, Azzaoui K, Sabbahi R, Youssoufi MH, Yahyaoui MI, Asehraou A, El Miz M, Hammouti B, Shityakov S, Siaj M, Mejdoubi E. Cheminformatics-Based Design and Synthesis of Hydroxyapatite/Collagen Nanocomposites for Biomedical Applications. Polymers (Basel) 2023; 16:85. [PMID: 38201750 PMCID: PMC10780405 DOI: 10.3390/polym16010085] [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: 12/04/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
This paper presents a novel cheminformatics approach for the design and synthesis of hydroxyapatite/collagen nanocomposites, which have potential biomedical applications in tissue engineering, drug delivery, and orthopedic and dental implants. The nanocomposites are synthesized by the co-precipitation method with different ratios of hydroxyapatite and collagen. Their mechanical, biological, and degradation properties are analyzed using various experimental and computational techniques. Attenuated total reflection-Fourier-transform infrared spectroscopy, thermogravimetric analysis, and X-ray diffraction unveil the low crystallinity and nanoscale particle size of hydroxyapatite (22.62 nm) and hydroxyapatite/collagen composites (14.81 nm). These findings are substantiated by scanning electron microscopy with energy-dispersive X-ray spectroscopy, confirming the Ca/P ratio between 1.65 and 1.53 and attesting to the formation of non-stoichiometric apatites in all samples, further validated by molecular simulation. The antimicrobial activity of the nanocomposites is evaluated in vitro against several bacterial and fungal strains, demonstrating their medical potential. Additionally, in silico analyses are performed to predict the absorption, distribution, metabolism, and excretion properties and the bioavailability of the collagen samples. This study paves the way for the development of novel biomaterials using chemoinformatics tools and methods, facilitating the optimization of design and synthesis parameters, as well as the prediction of biological outcomes. Future research directions should encompass the investigation of in vivo biocompatibility and bioactivity of the nanocomposites, while exploring further applications and functionalities of these innovative materials.
Collapse
Affiliation(s)
- Mohamed Aaddouz
- Laboratory of Applied Chemistry and Environment, Team: Mineral Chemistry of Solids, Department of Chemistry, Faculty of Sciences, Mohammed 1st University, P.O. Box 717, Oujda 60000, Morocco; (M.H.Y.); (E.M.)
| | - Khalil Azzaoui
- Laboratory of Engineering, Electrochemistry, Modeling and Environment, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco;
- Euromed Research Center, Euromed Polytechnic School, Euro-Mediterranean University of Fes, P.O. Box 15, Fes 30070, Morocco;
| | - Rachid Sabbahi
- Euromed Research Center, Euromed Polytechnic School, Euro-Mediterranean University of Fes, P.O. Box 15, Fes 30070, Morocco;
- Higher School of Technology, Ibn Zohr University, Quartier 25 Mars, P.O. Box 3007, Laayoune 70000, Morocco
| | - Moulay Hfid Youssoufi
- Laboratory of Applied Chemistry and Environment, Team: Mineral Chemistry of Solids, Department of Chemistry, Faculty of Sciences, Mohammed 1st University, P.O. Box 717, Oujda 60000, Morocco; (M.H.Y.); (E.M.)
| | - Meryem Idrissi Yahyaoui
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed Premier University, Oujda 60000, Morocco; (M.I.Y.); (A.A.)
| | - Abdeslam Asehraou
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed Premier University, Oujda 60000, Morocco; (M.I.Y.); (A.A.)
| | - Mohamed El Miz
- Laboratory of Molecular Chemistry, Materials and Environment (LCM2E), Multidisciplinary Faculty of Nador, University Mohamed I, Nador 60700, Morocco;
| | - Belkheir Hammouti
- Euromed Research Center, Euromed Polytechnic School, Euro-Mediterranean University of Fes, P.O. Box 15, Fes 30070, Morocco;
| | - Sergey Shityakov
- Department of Bioinformatics, Würzburg University, 97074 Würzburg, Germany;
| | - Mohamed Siaj
- Department of Chemistry, Université du Québec à Montréal, NanoQAM/QCAM, Montréal, QC H3C 3P8, Canada
| | - Elmiloud Mejdoubi
- Laboratory of Applied Chemistry and Environment, Team: Mineral Chemistry of Solids, Department of Chemistry, Faculty of Sciences, Mohammed 1st University, P.O. Box 717, Oujda 60000, Morocco; (M.H.Y.); (E.M.)
| |
Collapse
|
9
|
Sivamaruthi BS, Kapoor DU, Kukkar RR, Gaur M, Elossaily GM, Prajapati BG, Chaiyasut C. Mesoporous Silica Nanoparticles: Types, Synthesis, Role in the Treatment of Alzheimer's Disease, and Other Applications. Pharmaceutics 2023; 15:2666. [PMID: 38140007 PMCID: PMC10747102 DOI: 10.3390/pharmaceutics15122666] [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: 10/03/2023] [Revised: 10/25/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Globally, many individuals struggle with Alzheimer's disease (AD), an unrelenting and incapacitating neurodegenerative condition. Despite notable research endeavors, effective remedies for AD remain constrained, prompting the exploration of innovative therapeutic avenues. Within this context, silica-based nanoplatforms have emerged with pronounced potential due to their unique attributes like expansive surface area, customizable pore dimensions, and compatibility with living systems. These nanoplatforms hold promise as prospective interventions for AD. This assessment provides a comprehensive overview encompassing various forms of mesoporous silica nanoparticles (MSNs), techniques for formulation, and their applications in biomedicine. A significant feature lies in their ability to precisely guide and control the transport of therapeutic agents to the brain, facilitated by the adaptability of these nanoplatforms as drug carriers. Their utility as tools for early detection and monitoring of AD is investigated. Challenges and prospects associated with harnessing MSNs are studied, underscoring the imperative of stringent safety evaluations and optimization of how they interact with the body. Additionally, the incorporation of multifunctional attributes like imaging and targeting components is emphasized to enhance their efficacy within the intricate milieu of AD. As the battle against the profound repercussions of AD persists, MSNs emerge as a promising avenue with the potential to propel the development of viable therapeutic interventions.
Collapse
Affiliation(s)
- Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand;
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Devesh U. Kapoor
- Department of Pharmacy, Dr. Dayaram Patel Pharmacy College, Bardoli 394601, Gujarat, India;
| | - Rajiv R. Kukkar
- School of Pharmacy, Raffles University, Neemrana 301705, Rajasthan, India
| | - Mansi Gaur
- Rajasthan Pharmacy College, Rajasthan University of Health Sciences, Jaipur 302033, Rajasthan, India
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana 384012, Gujarat, India
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
10
|
T P, Katta B, Lulu S S, Sundararajan V. Gene expression analysis reveals GRIN1, SYT1, and SYN2 as significant therapeutic targets and drug repurposing reveals lorazepam and lorediplon as potent inhibitors to manage Alzheimer's disease. J Biomol Struct Dyn 2023; 42:10352-10373. [PMID: 37691428 DOI: 10.1080/07391102.2023.2256878] [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: 04/24/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
Alzheimer's disease (AD) is a slowly progressive neurodegenerative disease and a leading cause of dementia. We aim to identify key genes for the development of therapeutic targets and biomarkers for potential treatments for AD. Meta-analysis was performed on six microarray datasets and identified the differentially expressed genes between healthy and Alzheimer's disease samples. Thereafter, we filtered out the common genes which were present in at least four microarray datasets for downstream analysis. We have constructed a gene-gene network for the common genes and identified six hub genes. Furthermore, we investigated the regulatory mechanisms of these hub genes by analysing their interaction with miRNAs and transcription factors. The gene ontology analysis results highlighted the enriched terms significantly associated with hub genes. Through an extensive literature survey, we found that three of the hub genes including GRIN1, SYN2, and SYT1 were critically involved in disease development. To leverage existing drugs for potential repurposing, we predicted drug-gene interaction using the drug-gene interaction database, and performed molecular docking studies. The docking results revealed that the drug compounds had strong interactions and favorable binding with selected hub genes. Lorazepam exhibits a binding energy of -7.3 kcal/mol with GRIN1, Lorediplon exhibits binding energies of -7.7 kcal/mol and -6.3 kcal/mol with the SYT1, and SYN2 respectively. In addition, 100 ns molecular dynamics simulations were carried out for the top complexes and apo protein as well. Furthermore, the MM-PBSA free energy calculations also revealed that these complexes are stable and had favorable energies. According to our study, the identified hub gene could serve as a biomarker as well as a therapeutic target for AD, and the proposed repurposed drug molecules appear to have promising efficacy in treating the disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Premkumar T
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Bhavana Katta
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Sajitha Lulu S
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Vino Sundararajan
- Integrative Multiomics Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
11
|
Fareed MM, Dutta K, Dandekar T, Tarabonda H, Skorb EV, Shityakov S. In silico investigation of nonsynonymous single nucleotide polymorphisms in BCL2 apoptosis regulator gene to design novel protein-based drugs against cancer. J Cell Biochem 2022; 123:2044-2056. [PMID: 36146908 DOI: 10.1002/jcb.30330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/04/2022] [Accepted: 09/10/2022] [Indexed: 12/24/2022]
Abstract
BCL2 apoptosis regulator gene encodes Bcl-2 pro-survival protein, which plays an important role to evade apoptosis in various cancers. Moreover, single nucleotide polymorphisms (SNPs) in the BCL2 gene can be nonsynonymous (nsSNPs), which might affect the protein stability and probably its function. Therefore, we implement cutting-edge computational techniques based on the Spherical Polar Fourier and Monte-Carlo algorithms to investigate the impact of these SNPs on the B cell lymphoma-2 (Bcl-2) stability and therapeutic potential of protein-based molecules to inhibit this protein. As a result, we identified two nsSNPs (Q118R and R129C) to be deleterious and highly conserved, having a negative effect on protein stability. Additionally, molecular docking and molecular dynamics simulations confirmed the decreased binding affinity of mutated Bcl-2 variants to bind three-helix bundle protein inhibitor as these mutations occurred in the protein-protein binding site. Overall, this computational approach investigating nsSNPs provides a useful basis for designing novel molecules to inhibit Bcl-2 pro-survival pathway in malignant cells.
Collapse
Affiliation(s)
- Muhammad Mazhar Fareed
- Department of Computer Science, School of Science and Engineering, Università degli studi di Verona, Verona, Italy
| | - Kunal Dutta
- Department of Human Physiology, Vidyasagar University, Midnapore, West Bengal, India
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Herman Tarabonda
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russia
| | - Ekaterina V Skorb
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russia
| | - Sergey Shityakov
- Laboratory of Chemoinformatics, Infochemistry Scientific Center, ITMO University, Saint-Petersburg, Russia
| |
Collapse
|
12
|
Linking Cerebrovascular Dysfunction to Age-Related Hearing Loss and Alzheimer’s Disease—Are Systemic Approaches for Diagnosis and Therapy Required? Biomolecules 2022; 12:biom12111717. [DOI: 10.3390/biom12111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Alzheimer’s disease (AD), the most common cause of dementia in the elderly, is a neurodegenerative disorder associated with neurovascular dysfunction, cognitive decline, and the accumulation of amyloid β peptide (Aβ) in the brain and tau-related lesions in neurons termed neurofibrillary tangles (NFTs). Aβ deposits and NFT formation are the central pathological hallmarks in AD brains, and the majority of AD cases have been shown to exhibit a complex combination of systemic comorbidities. While AD is the foremost common cause of dementia in the elderly, age-related hearing loss (ARHL) is the most predominant sensory deficit in the elderly. During aging, chronic inflammation and resulting endothelial dysfunction have been described and might be key contributors to AD; we discuss an intriguing possible link between inner ear strial microvascular pathology and blood–brain barrier pathology and present ARHL as a potentially modifiable and treatable risk factor for AD development. We present compelling evidence that ARHL might well be seen as an important risk factor in AD development: progressive hearing impairment, leading to social isolation, and its comorbidities, such as frailty, falls, and late-onset depression, link ARHL with cognitive decline and increased risk of dementia, rendering it tempting to speculate that ARHL might be a potential common molecular and pathological trigger for AD. Additionally, one could speculate that amyloid-beta might damage the blood–labyrinth barrier as it does to the blood–brain barrier, leading to ARHL pathology. Finally, there are options for the treatment of ARHL by targeted neurotrophic factor supplementation to the cochlea to improve cognitive outcomes; they can also prevent AD development and AD-related comorbidity in the future.
Collapse
|
13
|
The Role of Clusterin Transporter in the Pathogenesis of Alzheimer’s Disease at the Blood–Brain Barrier Interface: A Systematic Review. Biomolecules 2022; 12:biom12101452. [PMID: 36291661 PMCID: PMC9599067 DOI: 10.3390/biom12101452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is considered a chronic and debilitating neurological illness that is increasingly impacting older-age populations. Some proteins, including clusterin (CLU or apolipoprotein J) transporter, can be linked to AD, causing oxidative stress. Therefore, its activity can affect various functions involving complement system inactivation, lipid transport, chaperone activity, neuronal transmission, and cellular survival pathways. This transporter is known to bind to the amyloid beta (Aβ) peptide, which is the major pathogenic factor of AD. On the other hand, this transporter is also active at the blood–brain barrier (BBB), a barrier that prevents harmful substances from entering and exiting the brain. Therefore, in this review, we discuss and emphasize the role of the CLU transporter and CLU-linked molecular mechanisms at the BBB interface in the pathogenesis of AD.
Collapse
|
14
|
Atilano SR, Abedi S, Ianopol NV, Singh MK, Norman JL, Malik D, Falatoonzadeh P, Chwa M, Nesburn AB, Kuppermann BD, Kenney MC. Differential Epigenetic Status and Responses to Stressors between Retinal Cybrids Cells with African versus European Mitochondrial DNA: Insights into Disease Susceptibilities. Cells 2022; 11:2655. [PMID: 36078063 PMCID: PMC9454894 DOI: 10.3390/cells11172655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial (mt) DNA can be classified into haplogroups, which represent populations with different geographic origins. Individuals of maternal African backgrounds (L haplogroup) are more prone to develop specific diseases compared those with maternal European-H haplogroups. Using a cybrid model, effects of amyloid-β (Amyβ), sub-lethal ultraviolet (UV) radiation, and 5-Aza-2'-deoxycytidine (5-aza-dC), a methylation inhibitor, were investigated. Amyβ treatment decreased cell metabolism and increased levels of reactive oxygen species in European-H and African-L cybrids, but lower mitochondrial membrane potential (ΔΨM) was found only in African-L cybrids. Sub-lethal UV radiation induced higher expression levels of CFH, EFEMP1, BBC3, and BCL2L13 in European-H cybrids compared to African-L cybrids. With respect to epigenetic status, the African-L cybrids had (a) 4.7-fold higher total global methylation levels (p = 0.005); (b) lower expression patterns for DNMT3B; and (c) elevated levels for HIST1H3F. The European-H and African-L cybrids showed different transcription levels for CFH, EFEMP1, CXCL1, CXCL8, USP25, and VEGF after treatment with 5-aza-dC. In conclusion, compared to European-H haplogroup cybrids, the African-L cybrids have different (i) responses to exogenous stressors (Amyβ and UV radiation), (ii) epigenetic status, and (iii) modulation profiles of methylation-mediated downstream complement, inflammation, and angiogenesis genes, commonly associated with various human diseases.
Collapse
Affiliation(s)
- Shari R. Atilano
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Sina Abedi
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Narcisa V. Ianopol
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Mithalesh K. Singh
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - J Lucas Norman
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Deepika Malik
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Payam Falatoonzadeh
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - Anthony B. Nesburn
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Baruch D. Kuppermann
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
| | - M. Cristina Kenney
- Gavin Herbert Eye Institute, Ophthalmology Research Laboratory, University of California Irvine, Hewitt Hall, Room 2028, 843 Health Science Rd., Irvine, CA 92697, USA
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
15
|
Pourabdi L, Küçükkılınç TT, Khoshtale F, Ayazgök B, Nadri H, Farokhi Alashti F, Forootanfar H, Akbari T, Shafiei M, Foroumadi A, Sharifzadeh M, Shafiee Ardestani M, Abaee MS, Firoozpour L, Khoobi M, Mojtahedi MM. Synthesis of New 3-Arylcoumarins Bearing N-Benzyl Triazole Moiety: Dual Lipoxygenase and Butyrylcholinesterase Inhibitors With Anti-Amyloid Aggregation and Neuroprotective Properties Against Alzheimer’s Disease. Front Chem 2022; 9:810233. [PMID: 35127652 PMCID: PMC8812461 DOI: 10.3389/fchem.2021.810233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
A novel series of coumarin derivatives linked to the N-benzyl triazole group were synthesized and evaluated against 15-lipoxygenase (15-LOX), and acetyl- and butyrylcholinesterase (AChE and BuChE) to find the most potent derivative against Alzheimer’s disease (AD). Most of the compounds showed weak to moderate activity against ChEs. Among the most active BuChE and 15-LOX inhibitors, 8l and 8n exhibited an excellent neuroprotective effect, higher than the standard drug (quercetin) on the PC12 cell model injured by H2O2 and significantly reduced aggregation of amyloid Aβ1-42, with potencies of 1.44 and 1.79 times higher than donepezil, respectively. Compound 8l also showed more activity than butylated hydroxytoluene (BHT) as the reference antioxidant agent in reducing the levels of H2O2 activated by amyloid β in BV2 microglial cells. Kinetic and ligand–enzyme docking studies were also performed for better understanding of the mode of interaction between the best BuChE inhibitor and the enzyme. Considering the acceptable BuChE and 15-LOX inhibition activities as well as significant neuroprotection, and anti-amyloid aggregation activities, 8l and 8n could be considered as potential MTDLs for further modification and studies against AD.
Collapse
Affiliation(s)
- Ladan Pourabdi
- Department of Organic Chemistry and Natural Products, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | | | - Fatemeh Khoshtale
- Department of Organic Chemistry and Natural Products, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Beyza Ayazgök
- Faculty of Pharmacy, Department of Biochemistry, Hacettepe University, Ankara, Turkey
| | - Hamid Nadri
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farid Farokhi Alashti
- Department of Organic Chemistry and Natural Products, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Hamid Forootanfar
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Mohammad Shafiei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Sciences Research Center, The institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - M. Saeed Abaee
- Department of Organic Chemistry and Natural Products, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Khoobi
- Pharmaceutical Sciences Research Center, The institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Mohammad M. Mojtahedi, ; Mehdi Khoobi, ,
| | - Mohammad M. Mojtahedi
- Department of Organic Chemistry and Natural Products, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
- *Correspondence: Mohammad M. Mojtahedi, ; Mehdi Khoobi, ,
| |
Collapse
|