1
|
Abu-Hashem AA, Abdelgawad AA, Gouda MA. Chemistry and Biological Activity of Thieno[3,4- b]quinoline, Thieno[3,4- c]
quinolone, Thieno[3,2- g]quinoline and Thieno[2,3- g]quinoline Derivatives:
A Review (Part IX). MINI-REV ORG CHEM 2024; 21:764-778. [DOI: 10.2174/1570193x20666230601151439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 07/10/2024]
Abstract
Abstract:
Over the previous decades, thieno-quinoline derivatives have acquired great interest due to
their synthetic and biological applications. These reports have been disclosed on Thienoquinoline
synthesis such as thieno[3,4-b]quinoline; thieno[3,4-c]quinolone; thieno [3,2-g]quinoline; thieno[2,3-
g] quinoline; spiro-thieno[2,3-g]quinoline; benzo[b]thiophen-iso- quinoline derivatives, and therefore
in the existent review, we provided an inclusive update on the synthesis of thienoquinolines. Characterization
of the preparation methods and reactivity is categorized based on their types of reactions as
addition, alkylation, chlorination, acylation, oxidation, reduction, cyclization and cyclo-condensation.
Hence, this study will help the researchers to obtain knowledge from the last literature research to
conquer their resolve problems in designing new compounds and processes.
Collapse
Affiliation(s)
- Ameen A. Abu-Hashem
- Chemistry Department, Faculty of Science, Jazan University, Jazan, 45142, Saudi Arabia
| | - Ahmed A.M. Abdelgawad
- Chemistry Department, Faculty of Science, Jazan University, Jazan, 45142, Saudi Arabia
| | - Moustafa A. Gouda
- Department of Chemistry,
Faculty of Science and Arts, Taibah University, Ulla, Medina, Saudi Arabia
- Department of Chemistry, Faculty of Science,
Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
2
|
Garemilla S, Kumari R, Kumar R. CDK5 as a therapeutic tool for the treatment of Alzheimer's disease: A review. Eur J Pharmacol 2024; 978:176760. [PMID: 38901526 DOI: 10.1016/j.ejphar.2024.176760] [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: 05/06/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024]
Abstract
Alzheimer's disease (AD) remains one of the most challenging and prevalent neurodegenerative disorders worldwide. Despite extensive research efforts, effective treatments for AD are lacking, emphasising the need for a deeper understanding of its underlying molecular mechanisms. Cyclin-dependent kinase 5 (CDK5), a serine/threonine kinase primarily associated with cell cycle regulation and neuronal development, has emerged as a key player in AD pathology. This review article comprehensively explores the multifaceted roles of CDK5 in the pathogenesis of AD. We begin by elucidating the physiological functions of CDK5 in normal brain development and neuronal maintenance, highlighting its involvement in synaptic plasticity, neurotransmitter release, and cytoskeletal dynamics. Subsequently, we delve into the dysregulation of CDK5 activity observed in AD, encompassing aberrant hyperactivation, and dysregulated protein interactions. Moreover, we discuss the intricate interplay between CDK5 and AD-related proteins, including amyloid-beta precursor protein (APP) and tau protein, elucidating their collective impact on disease progression. Finally, we described various approaches available for the inhibition of CDK-5, which can be explored as future therapeutic intervention for AD. Through synthesizing evidence from in vitro studies, animal models, and clinical investigations, this review provides a comprehensive overview of the intricate relationship between CDK5 dysregulation and AD pathogenesis, offering insights that may inform future therapeutic interventions strategies.
Collapse
Affiliation(s)
- Sandilya Garemilla
- Department of Life Sciences, GITAM School of Sciences, GITAM (Deemed to Be) University, Visakhapatnam, India; The George Washington University, Washington, DC, USA
| | - Richa Kumari
- Department of Life Sciences, GITAM School of Sciences, GITAM (Deemed to Be) University, Visakhapatnam, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, Maharashtra, India
| | - Rahul Kumar
- Department of Life Sciences, GITAM School of Sciences, GITAM (Deemed to Be) University, Visakhapatnam, India.
| |
Collapse
|
3
|
Gopinathan A, Sankhe R, Rathi E, Kodi T, Upadhya R, Pai KSR, Kishore A. An in silico drug repurposing approach to identify HDAC1 inhibitors against glioblastoma. J Biomol Struct Dyn 2024:1-14. [PMID: 38686917 DOI: 10.1080/07391102.2024.2335293] [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: 12/02/2023] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
Despite considerable improvement in therapy and diagnosis, brain tumors remain a global public health concern. Among all brain tumors, 80% are due to Glioblastoma. The average survival rate of a patient once diagnosed with glioblastoma is 15 months. Lately, the role of peptidase enzymes, especially Neprilysin, a neutral endopeptidase, is gaining attention for its role in tumor growth regulation. Neprilysin expressions are positively correlated with several tumors including GBM and reduced expression of NEP protein is associated with the pathogenesis of multiple tumors. One of the main reasons for NEP protein downregulation is the action of Histone deacetylase (HDAC) enzymes, especially HDAC1. Additionally, studies have reported that increased levels of HDAC1 are responsible for downregulating NEP gene expression. Hence, HDAC1 inhibition can be a good target to elevate NEP levels, which can be a good therapeutic approach to GBM. This study utilizes the computational drug repurposing tool, Schrodinger Maestro to identify HDAC1 inhibitors from the ZINC15 database.1379 FDA-approved drugs from the ZINC15 database were screened through molecular docking. Based on docking score and ligand-protein interaction, the top ten molecules were selected which were then subjected to binding energy calculation and molecular dynamics (MD) simulations. The three most active drugs from the MD simulations- ZINC22010649 (Panobinostat), ZINC4392649 (Tasimelteon) and ZINC1673 (Melphalan), were tested on C6 and U87 MG glioblastoma cells for cytotoxicity and HDAC1 protein levels using western blot analysis. Among the three drugs, Panobinostat exhibited potent cytotoxic action and showed a significant reduction in the HDAC1 protein levels.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Adarsh Gopinathan
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Runali Sankhe
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ekta Rathi
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Triveni Kodi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Raghavendra Upadhya
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
4
|
Li Z, Yin B, Zhang S, Lan Z, Zhang L. Targeting protein kinases for the treatment of Alzheimer's disease: Recent progress and future perspectives. Eur J Med Chem 2023; 261:115817. [PMID: 37722288 DOI: 10.1016/j.ejmech.2023.115817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/02/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
Alzheimer's disease (AD) is a serious neurodegenerative disease characterized by memory impairment, mental retardation, impaired motor balance, loss of self-care and even death. Among the complex and diverse pathological changes in AD, protein kinases are deeply involved in abnormal phosphorylation of Tau proteins to form intracellular neuronal fiber tangles, neuronal loss, extracellular β-amyloid (Aβ) deposits to form amyloid plaques, and synaptic disturbances. As a disease of the elderly, the growing geriatric population is directly driving the market demand for AD therapeutics, and protein kinases are potential targets for the future fight against AD. This perspective provides an in-depth look at the role of the major protein kinases (GSK-3β, CDK5, p38 MAPK, ERK1/2, and JNK3) in the pathogenesis of AD. At the same time, the development of different protein kinase inhibitors and the current state of clinical advancement are also outlined.
Collapse
Affiliation(s)
- Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Bo Yin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Shuangqian Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhigang Lan
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
5
|
Bhurta D, Bharate SB. Discovery of Pongol, the Furanoflavonoid, as an Inhibitor of CDK7/Cyclin H/MAT1 and Its Preliminary Structure-Activity Relationship. ACS OMEGA 2023; 8:1291-1300. [PMID: 36643464 PMCID: PMC9835647 DOI: 10.1021/acsomega.2c06733] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/01/2022] [Indexed: 06/13/2023]
Abstract
Natural products have been a great source of leads for cancer drug discovery. The cyclin-dependent kinases (CDKs) play a vital role in the initiation and progression of cancer. The CDK-activating kinase, CDK7/cyclin H/MAT1, has recently gained tremendous attention in targeted cancer drug discovery. Herein, we screened a small library of pure natural products in an ADP-Glo CDK7/H kinase assay that yielded a series of furano- and naphthoflavonoids among actives. Pongol (SBN-88), the hydroxy-substituted furanoflavonoid, inhibits CDK7/H as well as CDK9/T1 with IC50 values of 0.93 and 0.83 μM, respectively, and >20-fold selectivity over CDK2/E1 (IC50 > 20 μM). The molecular docking and molecular dynamics simulation revealed that the presence of phenolic -OH in pongol is vital for kinase inhibition, as its absence resulted in a significant loss in activity (e.g., lanceolatin B). The prime MM-GBSA calculations revealed the presence of strong lipophilic and H-bonding interactions of pongol with CDKs.
Collapse
Affiliation(s)
- Deendyal Bhurta
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sandip B. Bharate
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
- Academy
of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
6
|
Cdk5-p25 as a key element linking amyloid and tau pathologies in Alzheimer's disease: Mechanisms and possible therapeutic interventions. Life Sci 2022; 308:120986. [PMID: 36152679 DOI: 10.1016/j.lfs.2022.120986] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022]
Abstract
Despite the fact that the small atypical serine/threonine cyclin-dependent kinase 5 (Cdk5) is expressed in a number of tissues, its activity is restricted to the central nervous system due to the neuron-only localization of its activators p35 and p39. Although its importance for the proper development and function of the brain and its role as a switch between neuronal survival and death are unmistakable and unquestionable, Cdk5 is nevertheless increasingly emerging, as supported by a large number of publications on the subject, as a therapeutic target of choice in the fight against Alzheimer's disease. Thus, its aberrant over activation via the calpain-dependent conversion of p35 into p25 is observed during the pathogenesis of the disease where it leads to the hyperphosphorylation of the β-amyloid precursor protein and tau. The present review highlights the pivotal roles of the hyperactive Cdk5-p25 complex activity in contributing to the development of Alzheimer's disease pathogenesis, with a particular emphasis on the linking function between Aβ and tau that this kinase fulfils and on the fact that Cdk5-p25 is part of a deleterious feed forward loop giving rise to a molecular machinery runaway leading to AD pathogenesis. Additionally, we discuss the advances and challenges related to the possible strategies aimed at specifically inhibiting Cdk5-p25 activity and which could lead to promising anti-AD therapeutics.
Collapse
|
7
|
Multitargeting the Action of 5-HT 6 Serotonin Receptor Ligands by Additional Modulation of Kinases in the Search for a New Therapy for Alzheimer's Disease: Can It Work from a Molecular Point of View? Int J Mol Sci 2022; 23:ijms23158768. [PMID: 35955902 PMCID: PMC9368844 DOI: 10.3390/ijms23158768] [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: 07/20/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022] Open
Abstract
In view of the unsatisfactory treatment of cognitive disorders, in particular Alzheimer’s disease (AD), the aim of this review was to perform a computer-aided analysis of the state of the art that will help in the search for innovative polypharmacology-based therapeutic approaches to fight against AD. Apart from 20-year unrenewed cholinesterase- or NMDA-based AD therapy, the hope of effectively treating Alzheimer’s disease has been placed on serotonin 5-HT6 receptor (5-HT6R), due to its proven, both for agonists and antagonists, beneficial procognitive effects in animal models; however, research into this treatment has so far not been successfully translated to human patients. Recent lines of evidence strongly emphasize the role of kinases, in particular microtubule affinity-regulating kinase 4 (MARK4), Rho-associated coiled-coil-containing protein kinase I/II (ROCKI/II) and cyclin-dependent kinase 5 (CDK5) in the etiology of AD, pointing to the therapeutic potential of their inhibitors not only against the symptoms, but also the causes of this disease. Thus, finding a drug that acts simultaneously on both 5-HT6R and one of those kinases will provide a potential breakthrough in AD treatment. The pharmacophore- and docking-based comprehensive literature analysis performed herein serves to answer the question of whether the design of these kind of dual agents is possible, and the conclusions turned out to be highly promising.
Collapse
|
8
|
Abu-Hashem AA, El-Gazzar ABA, Abdelgawad AAM, Gouda MA. Synthesis and chemical reactions of thieno[3,2- c]quinolines from arylamine derivatives, part (V): a review. PHOSPHORUS SULFUR 2022. [DOI: 10.1080/10426507.2021.2012176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Ameen A. Abu-Hashem
- Photochemistry Department (Heterocyclic Unit), National Research Centre, Dokki, Giza, Egypt
- Chemistry Department, Faculty of Science, Jazan University, Jazan, Saudi Arabia
| | - A. B. A. El-Gazzar
- Photochemistry Department (Heterocyclic Unit), National Research Centre, Dokki, Giza, Egypt
| | - Ahmed A. M. Abdelgawad
- Chemistry Department, Faculty of Science, Jazan University, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants Department, Desert Research Center, Cairo, Egypt
| | - Moustafa A. Gouda
- Department of Chemistry, Faculty of Science and Arts, Taibah University, Ulla, Medina, Saudi Arabia
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
9
|
Xie Z, Hou S, Yang X, Duan Y, Han J, Wang Q, Liao C. Lessons Learned from Past Cyclin-Dependent Kinase Drug Discovery Efforts. J Med Chem 2022; 65:6356-6389. [PMID: 35235745 DOI: 10.1021/acs.jmedchem.1c02190] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibition of cyclin-dependent kinases (CDKs) has become an effective therapeutic strategy for treating various diseases, especially cancer. Over almost three decades, although great efforts have been made to discover CDK inhibitors, many of which have entered clinical trials, only four CDK inhibitors have been approved. In the process of CDK inhibitor development, many difficulties and misunderstandings have hampered their discovery and clinical applications, which mainly include inadequate understanding of the biological functions of CDKs, less attention paid to pan- and multi-CDK inhibitors, nonideal isoform selectivity of developed selective CDK inhibitors, overlooking the metabolic stability of early discovered CDK inhibitors, no effective resistance solutions, and a lack of available combination therapy and effective biomarkers for CDK therapies. After reviewing the mechanisms of CDKs and the research progress of CDK inhibitors, this perspective summarizes and discusses these difficulties or lessons, hoping to facilitate the successful discovery of more useful CDK inhibitors.
Collapse
Affiliation(s)
- Zhouling Xie
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Shuzeng Hou
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Xiaoxiao Yang
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Yajun Duan
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Jihong Han
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Qin Wang
- Department of Otolaryngology─Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P. R. China
| | - Chenzhong Liao
- Department of Pharmaceutical Sciences and Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| |
Collapse
|
10
|
Buglioni L, Raymenants F, Slattery A, Zondag SDA, Noël T. Technological Innovations in Photochemistry for Organic Synthesis: Flow Chemistry, High-Throughput Experimentation, Scale-up, and Photoelectrochemistry. Chem Rev 2022; 122:2752-2906. [PMID: 34375082 PMCID: PMC8796205 DOI: 10.1021/acs.chemrev.1c00332] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Indexed: 02/08/2023]
Abstract
Photoinduced chemical transformations have received in recent years a tremendous amount of attention, providing a plethora of opportunities to synthetic organic chemists. However, performing a photochemical transformation can be quite a challenge because of various issues related to the delivery of photons. These challenges have barred the widespread adoption of photochemical steps in the chemical industry. However, in the past decade, several technological innovations have led to more reproducible, selective, and scalable photoinduced reactions. Herein, we provide a comprehensive overview of these exciting technological advances, including flow chemistry, high-throughput experimentation, reactor design and scale-up, and the combination of photo- and electro-chemistry.
Collapse
Affiliation(s)
- Laura Buglioni
- Micro
Flow Chemistry and Synthetic Methodology, Department of Chemical Engineering
and Chemistry, Eindhoven University of Technology, Het Kranenveld, Bldg 14—Helix, 5600 MB, Eindhoven, The Netherlands
- Flow
Chemistry Group, van ’t Hoff Institute for Molecular Sciences
(HIMS), Universiteit van Amsterdam (UvA), Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Fabian Raymenants
- Flow
Chemistry Group, van ’t Hoff Institute for Molecular Sciences
(HIMS), Universiteit van Amsterdam (UvA), Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Aidan Slattery
- Flow
Chemistry Group, van ’t Hoff Institute for Molecular Sciences
(HIMS), Universiteit van Amsterdam (UvA), Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Stefan D. A. Zondag
- Flow
Chemistry Group, van ’t Hoff Institute for Molecular Sciences
(HIMS), Universiteit van Amsterdam (UvA), Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Timothy Noël
- Flow
Chemistry Group, van ’t Hoff Institute for Molecular Sciences
(HIMS), Universiteit van Amsterdam (UvA), Science Park 904, 1098 XH, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Patel S, Globisch C, Pulugu P, Kumar P, Jain A, Shard A. Novel imidazopyrimidines-based molecules induce tetramerization of tumor pyruvate kinase M2 and exhibit potent antiproliferative profile. Eur J Pharm Sci 2021; 170:106112. [PMID: 34971746 DOI: 10.1016/j.ejps.2021.106112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/15/2021] [Accepted: 12/26/2021] [Indexed: 12/22/2022]
Abstract
Discovery of novel and potent lead molecules for the specific therapeutic targets by de novo drug design is still in infancy. Here, we disclose the unprecedented development of imidazopyri(mi)dine-based tumor pyruvate kinase M2 (PKM2) modulators by subsequent link and grow strategy. The most potent modulator 15n acts as a PKM2 activator with an AC50 of 90 nM, with considerable cancer cell-selectivity and membrane-permeability. NMR metabolomics studies also revealed that treatment with 15n results in diminution in lactate concentrations in MCF-7 cells. 15n binds to a previously reported site at PKM2 adjacent to the interface of two monomers. In molecular dynamics (MD) simulation studies, it was observed that 15n stabilizes the PKM2 at the dimeric interface, assisting in the formation of a biologically active tetramer conformation. 15n was also screened on MCF-7 breast cancer cell lines grown on 3-D scaffolds, and the results exhibited better anticancer potential compared to control, paving the way for future clinical studies.
Collapse
Affiliation(s)
- Sagarkumar Patel
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj-Basan Road, Gandhinagar, 382355, Gujarat, India
| | | | - Priyanka Pulugu
- Department of Medical Devices National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj-Basan Road, Gandhinagar, 382355, Gujarat, India
| | - Prasoon Kumar
- Department of Medical Devices National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj-Basan Road, Gandhinagar, 382355, Gujarat, India
| | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj-Basan Road, Gandhinagar, 382355, Gujarat, India; Department of Bioengineering, BIT Mesra, Ranchi, India.
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad, Opposite Air Force Station, Palaj-Basan Road, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
12
|
Nath V, Rohini A, Kumar V. Identification of M pro inhibitors of SARS-CoV-2 using structure based computational drug repurposing. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2021; 37:102178. [PMID: 34611467 PMCID: PMC8483991 DOI: 10.1016/j.bcab.2021.102178] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/03/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022]
Abstract
The recent outbreak of COVID-19, caused by the novel pathogen SARS-coronavirus 2 (SARS-CoV-2) is a severe health emergency. In this pandemic, drug repurposing seems to be the most promising alternative to identify effective therapeutic agents for immediate treatment of infected patients. The present study aimed to evaluate all the drugs present in drug bank as potential novel SARS-CoV-2 inhibitors, using computational drug repurposing studies. Docking-based virtual screening and binding energy prediction were performed, followed by Absorption Distribution Metabolism Excretion calculation. Hydroxychloroquine and Nelfinavir have been identified as the best potential inhibitor against the SARS-CoV-2, therefore, they were used as reference compounds in computational DR studies. The docking study revealed 13 best compounds based on their highest binding affinity, binding energy, and dock score concerning the other screened compounds. Out of 13, only 4 compounds were further shortlisted based on their binding energy and best ADME properties. The hierarchical virtual screening yielded the best 04 drugs, DB07042 (compound 2), DB13035 (compound 3), DB13604 (compound 5) and DB08253 (compound 6), with commendable binding energies in kcal/mol, i.e. −65.45, −62.01, −52.09 and −51.70 respectively. Further, Molecular dynamics simulation with 04 best-retrieved hits has confirmed stable trajectories in protein in terms of root mean square deviation and root mean square fluctuation. During 30 ns simulation, the interactions were also found similar to the docking-based studies. However, clinical studies are necessary to investigate their therapeutic use against this outbreak.
Collapse
Key Words
- ACE, Angiotensin-Converting Enzyme
- ADME, Absorption Distribution Metabolism Excretion
- Binding energy
- CDR, Computational Drug Repurposing
- COVID
- CoV, Corona Virus
- Docking
- Drug repurposing
- HTVS, High-throughput virtual screening
- MMGBSA, Molecular mechanics generalized born surface area
- OPLS, Optimized Potentials for Liquid Simulations
- PDB, Protein data bank
- SARS, Severe Acute Respiratory Syndrome
- SP, Standard Precision
- Virtual screening
- XP, Extra precision
Collapse
Affiliation(s)
- Virendra Nath
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, India
| | - A Rohini
- College of Pharmacy, JSS Academy of Technical Sciences, Noida, Uttar Pradesh, India
| | - Vipin Kumar
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
13
|
Iusupov IR, Curreli F, Spiridonov EA, Markov PO, Ahmed S, Belov DS, Manasova EV, Altieri A, Kurkin AV, Debnath AK. Design of gp120 HIV-1 entry inhibitors by scaffold hopping via isosteric replacements. Eur J Med Chem 2021; 224:113681. [PMID: 34246921 DOI: 10.1016/j.ejmech.2021.113681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
We present the development of alternative scaffolds and validation of their synthetic pathways as a tool for the exploration of new HIV gp120 inhibitors based on the recently discovered inhibitor of this class, NBD-14136. The new synthetic routes were based on isosteric replacements of the amine and acid precursors required for the synthesis of NBD-14136, guided by molecular modeling and chemical feasibility analysis. To ensure that these synthetic tools and new scaffolds had the potential for further exploration, we eventually tested few representative compounds from each newly designed scaffold against the gp120 inhibition assay and cell viability assays.
Collapse
Affiliation(s)
- Ildar R Iusupov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Evgeniy A Spiridonov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Pavel O Markov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Shahad Ahmed
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States
| | - Dmitry S Belov
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Ekaterina V Manasova
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Campus, Moscow State University, Leninskie Gory Bld. 75, 77-101b, Moscow, 119992, Russia.
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute, New York Blood Center, 310 E 67th Street, New York, 10065, New York, United States.
| |
Collapse
|
14
|
Nath V, Ramchandani M, Kumar N, Agrawal R, Kumar V. Computational identification of potential dipeptidyl peptidase (DPP)-IV inhibitors: Structure based virtual screening, molecular dynamics simulation and knowledge based SAR studies. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
15
|
Allnutt AB, Waters AK, Kesari S, Yenugonda VM. Physiological and Pathological Roles of Cdk5: Potential Directions for Therapeutic Targeting in Neurodegenerative Disease. ACS Chem Neurosci 2020; 11:1218-1230. [PMID: 32286796 DOI: 10.1021/acschemneuro.0c00096] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine (ser)/threonine (Thr) kinase that has been demonstrated to be one of the most functionally diverse kinases within neurons. Cdk5 is regulated via binding with its neuron-specific regulatory subunits, p35 or p39. Cdk5-p35 activity is critical for a variety of developmental and cellular processes in the brain, including neuron migration, memory formation, microtubule regulation, and cell cycle suppression. Aberrant activation of Cdk5 via the truncated p35 byproduct, p25, is implicated in the pathogenesis of several neurodegenerative diseases. The present review highlights the importance of Cdk5 activity and function in the brain and demonstrates how deregulation of Cdk5 can contribute to the development of neurodegenerative conditions such as Alzheimer's and Parkinson's disease. Additionally, we cover past drug discovery attempts at inhibiting Cdk5-p25 activity and discuss which types of targeting strategies may prove to be the most successful moving forward.
Collapse
|
16
|
Loganathan L, Natarajan K, Muthusamy K. Computational study on cross-talking cancer signalling mechanism of ring finger protein 146, AXIN and Tankyrase protein complex. J Biomol Struct Dyn 2019; 38:5173-5185. [PMID: 31760854 DOI: 10.1080/07391102.2019.1696707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is distinguished by uncontrolled cell growth and it is regulated by several environmental and genetic factors. The Wnt β-Catenin signaling pathway has been considered as the most significant colon cancer-targeted pathway. AXIN plays a major regulatory role in the Wnt signaling mechanism. The AXIN after PARsylated by TNKS is ubiqutinated by RNF146 through its WWE domain that leads to degradation of AXIN protein. Several studies have been proposed highlighting the inhibition of the PARsylation mechanism that mediates the degradation of AXIN and improves β-catenin stability. The present study focused on the identification of potential inhibitors for the inhibition of RNF146-TNKS complex through identifying potential RNF146 inhibitors to prevent ubiquitination of AXIN, further to confirm the regulatory role and inhibition mechanism of RNF146-AXIN and RNF146-TNKS. The docked complex was then evaluated using various computational analysis. Molecular interactions analysis was performed to observe the interacting residues between the protein complex. The compounds from various databases were docked with the RNF146 and complex proteins. Both the protein complex and ligand were analyzed for the confirmation of structural stability using molecular dynamics simulations. Selected compounds' atomic configuration and electron profile were analyzed through DFT calculations and ADME/T (Physico-chemical) properties. As a result, we found several common lead compounds for RNF146, TNKS protein inhibition. Therefore, the docked compounds may act as a better antagonist molecule for RNF146, TNKS and associated signaling molecules. Further, experimental validations are required to prove the potency of the identified compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Keerthana Natarajan
- Department of Bioinformatics, Alagappa University, Karaikudi, Tamilnadu, India
| | | |
Collapse
|
17
|
Computational and experimental elucidation of Plasmodium falciparum phosphoethanolamine methyltransferase inhibitors: Pivotal drug target. PLoS One 2019; 14:e0221032. [PMID: 31437171 PMCID: PMC6705855 DOI: 10.1371/journal.pone.0221032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/29/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Plasmodium falciparum synthesizes phosphatidylcholine for the membrane development through serine decarboxylase-phosphoethanolamine methyltransferase pathway for growth in human host. Phosphoethanolamine-methyltransferase (PfPMT) is a crucial enzyme for the synthesis of phosphocholine which is a precursor for phosphatidylcholine synthesis and is considered as a pivotal drug target as it is absent in the host. The inhibition of PfPMT may kill malaria parasite and hence is being considered as potential target for rational antimalarial drug designing. METHODS In this study, we have used computer aided drug designing (CADD) approaches to establish potential PfPMT inhibitors from Asinex compound library virtually screened for ADMET and the docking affinity. The selected compounds were tested for in-vitro schizonticidal, gametocidal and cytotoxicity activity. Nontoxic compounds were further studied for PfPMT enzyme specificity and antimalarial efficacy for P. berghei in albino mice model. RESULTS Our results have identified two nontoxic PfPMT competitive inhibitors ASN.1 and ASN.3 with better schizonticidal and gametocidal activity which were found to inhibit PfPMT at IC50 1.49μM and 2.31μM respectively. The promising reduction in parasitaemia was found both in orally (50 & 10 mg/kg) and intravenous (IV) (5& 1 mg/kg) however, the better growth inhibition was found in intravenous groups. CONCLUSION We report that the compounds containing Pyridinyl-Pyrimidine and Phenyl-Furan scaffolds as the potential inhibitors of PfPMT and thus may act as promising antimalarial inhibitor candidates which can be further optimized and used as leads for template based antimalarial drug development.
Collapse
|
18
|
Ganeshpurkar A, Swetha R, Kumar D, Gangaram GP, Singh R, Gutti G, Jana S, Kumar D, Kumar A, Singh SK. Protein-Protein Interactions and Aggregation Inhibitors in Alzheimer's Disease. Curr Top Med Chem 2019; 19:501-533. [PMID: 30836921 DOI: 10.2174/1568026619666190304153353] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/31/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alzheimer's Disease (AD), a multifaceted disorder, involves complex pathophysiology and plethora of protein-protein interactions. Thus such interactions can be exploited to develop anti-AD drugs. OBJECTIVE The interaction of dynamin-related protein 1, cellular prion protein, phosphoprotein phosphatase 2A and Mint 2 with amyloid β, etc., studied recently, may have critical role in progression of the disease. Our objective has been to review such studies and their implications in design and development of drugs against the Alzheimer's disease. METHODS Such studies have been reviewed and critically assessed. RESULTS Review has led to show how such studies are useful to develop anti-AD drugs. CONCLUSION There are several PPIs which are current topics of research including Drp1, Aβ interactions with various targets including PrPC, Fyn kinase, NMDAR and mGluR5 and interaction of Mint2 with PDZ domain, etc., and thus have potential role in neurodegeneration and AD. Finally, the multi-targeted approach in AD may be fruitful and opens a new vista for identification and targeting of PPIs in various cellular pathways to find a cure for the disease.
Collapse
Affiliation(s)
- Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Rayala Swetha
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Devendra Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Gore P Gangaram
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Gopichand Gutti
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Srabanti Jana
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Dileep Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | - Sushil K Singh
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| |
Collapse
|
19
|
Khair N, Lenjisa JL, Tadesse S, Kumarasiri M, Basnet SKC, Mekonnen LB, Li M, Diab S, Sykes MJ, Albrecht H, Milne R, Wang S. Discovery of CDK5 Inhibitors through Structure-Guided Approach. ACS Med Chem Lett 2019; 10:786-791. [PMID: 31098000 PMCID: PMC6511963 DOI: 10.1021/acsmedchemlett.9b00029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/20/2019] [Indexed: 12/18/2022] Open
Abstract
Specific abrogation of cyclin-dependent kinase 5 (CDK5) activity has been validated as a viable approach for the development of anticancer agents. However, no selective CDK5 inhibitor has been reported to date. Herein, a structure-based in silico screening was employed to identify novel scaffolds from a library of compounds to identify potential CDK5 inhibitors that would be relevant for drug discovery. Hits, representatives of three chemical classes, were identified as inhibitors of CDK5. Structural modification of hit-1 resulted in 29 and 30. Compound 29 is a dual inhibitor of CDK5 and CDK2, whereas 30 preferentially inhibits CDK5. Both leads exhibited anticancer activity against acute myeloid leukemia (AML) cells via a mechanism consistent with targeting cellular CDK5. This study provides an effective strategy for discovery of CDK5 inhibitors as potential antileukemic agents.
Collapse
Affiliation(s)
| | | | - Solomon Tadesse
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Malika Kumarasiri
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Sunita K. C. Basnet
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Laychiluh B. Mekonnen
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Manjun Li
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Sarah Diab
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Matthew J. Sykes
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Hugo Albrecht
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Robert Milne
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Shudong Wang
- Centre for Drug Discovery
and Development, School of Pharmacy and Medical Sciences, Cancer Research
Institute, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
20
|
Shukla R, Singh TR. Virtual screening, pharmacokinetics, molecular dynamics and binding free energy analysis for small natural molecules against cyclin-dependent kinase 5 for Alzheimer's disease. J Biomol Struct Dyn 2019; 38:248-262. [PMID: 30688165 DOI: 10.1080/07391102.2019.1571947] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and characterized by brain cell death, memory loss and is the most common form of dementia. Although AD has devastating effects, however, drugs which can treat the AD remain limited. The cyclin-dependent kinase 5 (CDK5) has been recognized as being involved in the pathological hyperphosphorylation of tau protein, which leads to the formation of neurofibrillary tangles (NFTs). We utilized the structure-based virtual screening (SBVS) approach to find the potential inhibitors against HsCDK5. The natural compound subset from the ZINC database (n = 167,741) was retrieved and screened by using SBVS method. From here, we have predicted 297 potent inhibitors. These 297 compounds were evaluated through their pharmacokinetic properties by ADMET (absorption, distribution, metabolism, elimination/excretion and toxicity) descriptors. Finally, 17 compounds were selected and used for re-docking. After the refinement by molecular docking and by using drug-likeness analysis, we have identified four potential inhibitors (ZINC85877721, ZINC96114862, ZINC96115616 and ZINC96116231). All these four ligands were employed for 100 ns MDS study. From the root mean square deviation (RMSD), root mean square fluctuation (RMSF), Rg, number of hydrogen bonds, solvent accessible surface area (SASA), principal component analysis (PCA) and binding free energy analysis we have found that out of four inhibitors ZINC85877721 and ZINC96116231 showed good binding free energy of -198.84 and -159.32 kJ.mol-1, respectively, and also good in other structural analyses. Both compounds displayed excellent pharmacological and structural properties to be the drug candidates. Collectively, these findings recommend that two compounds have great potential to be a promising agent against AD to reduce the CDK5 induced hyperphosphorylation and could be considered as therapeutic agents for the AD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, India
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, India
| |
Collapse
|
21
|
Latek D, Rutkowska E, Niewieczerzal S, Cielecka-Piontek J. Drug-induced diabetes type 2: In silico study involving class B GPCRs. PLoS One 2019; 14:e0208892. [PMID: 30650080 PMCID: PMC6334951 DOI: 10.1371/journal.pone.0208892] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/27/2018] [Indexed: 01/10/2023] Open
Abstract
A disturbance of glucose homeostasis leading to type 2 diabetes mellitus (T2DM) is one of the severe side effects that may occur during a prolonged use of many drugs currently available on the market. In this manuscript we describe the most common cases of drug-induced T2DM, discuss available pharmacotherapies and propose new ones. Among various pharmacotherapies of T2DM, incretin therapies have recently focused attention due to the newly determined crystal structure of incretin hormone receptor GLP1R. Incretin hormone receptors: GLP1R and GIPR together with the glucagon receptor GCGR regulate food intake and insulin and glucose secretion. Our study showed that incretin hormone receptors, named also gut hormone receptors as they are expressed in the gastrointestinal tract, could potentially act as unintended targets (off-targets) for orally administrated drugs. Such off-target interactions, depending on their effect on the receptor (stimulation or inhibition), could be beneficial, like in the case of incretin mimetics, or unwanted if they cause, e.g., decreased insulin secretion. In this in silico study we examined which well-known pharmaceuticals could potentially interact with gut hormone receptors in the off-target way. We observed that drugs with the strongest binding affinity for gut hormone receptors were also reported in the medical information resources as the least disturbing the glucose homeostasis among all drugs in their class. We suggested that those strongly binding molecules could potentially stimulate GIPR and GLP1R and/or inhibit GCGR which could lead to increased insulin secretion and decreased hepatic glucose production. Such positive effect on the glucose homeostasis could compensate for other, adverse effects of pharmacotherapy which lead to drug-induced T2DM. In addition, we also described several top hits as potential substitutes of peptidic incretin mimetics which were discovered in the drug repositioning screen using gut hormone receptors structures against the ZINC15 compounds subset.
Collapse
Affiliation(s)
- Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | | | | | - Judyta Cielecka-Piontek
- Department of Pharmacognosy, Faculty of Pharmacy, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
22
|
Kumar S, Singh J, Narasimhan B, Shah SAA, Lim SM, Ramasamy K, Mani V. Reverse pharmacophore mapping and molecular docking studies for discovery of GTPase HRas as promising drug target for bis-pyrimidine derivatives. Chem Cent J 2018; 12:106. [PMID: 30345469 PMCID: PMC6768019 DOI: 10.1186/s13065-018-0475-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/09/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Pyrimidine is an important pharmacophore in the field of medicinal chemistry and exhibit a broad spectrum of biological potentials. A study was carried out to identify the target protein of potent bis-pyrimidine derivatives using reverse docking program. PharmMapper, a robust online tool was used for identifying the target proteins based on reverse pharmacophore mapping. The murine macrophage (RAW 264.7) and human embryonic kidney (HEK-293) cancer cell line used for selectivity and safety study. METHODS An open web server PharmMapper was used to identify the possible target of the developed compounds through reverse pharmacophore mapping. The results were analyzed and validated through docking with Schrodinger v9.6 using 10 protein GTPase HRas selected as possible target. The docking studies with Schrödinger validated the binding behavior of bis-pyrimidine compounds within GTP binding pocket. MTT and sulforhodamine assay were used as antiproliferative activity. RESULTS AND DISCUSSION The protein was found one of the top scored targets of the compound 18, hence, the GTPase HRas protein was found crucial to be targeted for competing cancer. Toxicity study demonstrated the significant selectivity of most active compounds, 12, 16 and 18 showed negligible cell toxicity at their IC50 concentration. CONCLUSION From the results, we may conclude that GTPase HRas as a possible target of studied bis-pyrimidine derivatives where the retrieved information may be quite useful for rational drug designing.
Collapse
Affiliation(s)
- Sanjiv Kumar
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Jagbir Singh
- Faculty of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | | | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns), Universiti Teknologi MARA, 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Kalavathy Ramasamy
- Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), 42300, Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia.,Collaborative Drug Discovery Research (CDDR) Group, Pharmaceutical Life Sciences Community of Research, Universiti Teknologi MARA (UiTM), 40450, Shah Alam, Selangor Darul Ehsan, Malaysia
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, 51452, Saudi Arabia
| |
Collapse
|
23
|
Kant V, Vijayakumar S, Sahoo GC, Ali V, Singh K, Chaudhery SS, Das P. In-silico screening and validation of high-affinity tetra-peptide inhibitor of Leishmania donovani O-acetyl serine sulfhydrylase (OASS). J Biomol Struct Dyn 2018; 37:481-492. [PMID: 29415627 DOI: 10.1080/07391102.2018.1429315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OASS is a specific enzyme that helps Leishmania parasite to survive the oxidative stress condition in human macrophages. SAT C-terminal peptides in several organisms, including Leishmania, were reported to inhibit or reduce the activity of OASS. Small peptide and small molecules mimicking the SAT C-terminal residues are designed and tested for the inhibition of OASS in different organisms. Hence, in this study, all the possible tetra-peptide combinations were designed and screened based on the docking ability with Leishmania donovani OASS (Ld-OASS). The top ranked peptides were further validated for the stability using 50 ns molecular dynamic simulation. In order to identify the better binding capability of the peptides, the top peptides complexed with Ld-OASS were also subjected to molecular dynamic simulation. The docking and simulation results favored the peptide EWSI to possess greater advantage than previously reported peptide (DWSI) in binding with Ld-OASS active site. Also, screening of non-peptide inhibitor of Asinex Biodesign library based on the shape similarity of EWSI and DWSI was performed. The top similar molecules of each peptides were docked on to Ld-OASS active site and subsequently simulated for 20 ns. The results suggested that the ligand that shares high shape similarity with EWSI possess better binding capability than the ligand that shares high shape similarity with DWSI. This study revealed that the tetra-peptide EWSI had marginal advantage over DWSI in binding with Ld-OASS, thereby providing basis for defining a pharmacophoric scaffold for the design of peptidomimetic inhibitors as well as non-peptide inhibitors of Ld-OASS. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vishnu Kant
- a Department of Pharmacoinformatics , National Institute of Pharmaceutical Education and Research , Hajipur , Bihar 844102 , India
| | - Saravanan Vijayakumar
- b Department of Statistics/Bioinformatics Centre , Rajendra Memorial Research Institute of Medical Science, ICMR , Agamkuan, Patna , Bihar 800007 , India
| | - Ganesh Chandra Sahoo
- c Bioinformatics Division , Rajendra Memorial Research Institute of Medical Science, ICMR , Agamkuan, Patna , Bihar 800007 , India
| | - Vahab Ali
- d Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India
| | - Kuljit Singh
- d Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India
| | - Shailendra S Chaudhery
- a Department of Pharmacoinformatics , National Institute of Pharmaceutical Education and Research , Hajipur , Bihar 844102 , India
| | - Pradeep Das
- e Department of Molecular Biology , Rajendra Memorial Research Institute of Medical Science, ICMR , Agamkuan, Patna , Bihar 800007 , India
| |
Collapse
|
24
|
Abstract
Selective abrogation of cyclin-dependent kinases (CDK) activity is a highly promising strategy in cancer treatment. The atypical CDK, CDK5 has long been known for its role in neurodegenerative diseases, and is becoming an attractive drug target for cancer therapy. Myriads of recent studies have uncovered that aberrant expression of CDK5 contributes to the oncogenic initiation and progression of multiple solid and hematological malignancies. CDK5 is also implicated in the regulation of cancer stem cell biology. In this review, we present the current state of knowledge of CDK5 as a druggable target for cancer treatment. We also provide a detailed outlook of designing selective and potent inhibitors of this enzyme.
Collapse
|
25
|
Park SJ, Kim E, Yoo M, Lee JY, Park CH, Hwang JY, Ha JD. Synthesis and biological evaluation of N9-cis-cyclobutylpurine derivatives for use as cyclin-dependent kinase (CDK) inhibitors. Bioorg Med Chem Lett 2017; 27:4399-4404. [DOI: 10.1016/j.bmcl.2017.08.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/08/2017] [Accepted: 08/10/2017] [Indexed: 12/17/2022]
|
26
|
Fang Y, Tranmer GK. Expedited access to thieno[3,2-c]quinolin-4(5H)-ones and benzo[h]-1,6-naphthyridin-5(6H)-ones via a continuous flow photocyclization method. Org Biomol Chem 2016; 14:10799-10803. [PMID: 27824202 DOI: 10.1039/c6ob02279k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A single-step continuous flow method has been developed that gives expedited access to complex heterocycles via an intramolecular photochemical cyclization. Herein we report the first examples of the photochemically-induced generation of thieno[3,2-c]quinolin-4(5H)-ones, 16 examples, and benzo[h]-1,6-naphthyridin-5(6H)-ones, 6 examples. Overall, the continuous flow method provides access to complex heterocycles in two steps from commercially available starting materials in good yields and with greater atom efficiency than traditional batch reactions.
Collapse
Affiliation(s)
- Y Fang
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB, CanadaR3E 0T5.
| | - G K Tranmer
- College of Pharmacy, Faculty of Health Science, University of Manitoba, Winnipeg, MB, CanadaR3E 0T5. and Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, CanadaR3T 2N2
| |
Collapse
|
27
|
Synthesis of novel S-acyl and S-alkylpyrimidinone derivatives as potential cytotoxic agents. RESEARCH ON CHEMICAL INTERMEDIATES 2016. [DOI: 10.1007/s11164-016-2487-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|