1
|
Gong Y, Tian X, Ma X, Xing J, Ji Z, Gao X, Chen Z, Hu F, Yin D, Li F, Sheng C, Liu Y, Shi S. New insights into the pharmacological mechanisms of Jinqi Jiangtang Tablets in the treatment of type 2 diabetes mellitus: A multi-omics approach combined with experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025:120020. [PMID: 40449695 DOI: 10.1016/j.jep.2025.120020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) is a prevalent chronic metabolic disorder characterized by impaired glucose homeostasis and insulin dysfunction. Despite ongoing efforts, the management of T2DM remains challenging due to its complex pathogenesis and the limited effectiveness of current treatments. Jinqi Jiangtang Tablets (JQJTT), a well-established Traditional Chinese Medicine (TCM) formula, have shown promising clinical benefits in regulating blood glucose levels and improving metabolic health in T2DM patients. However, the specific pharmacological mechanisms behind its efficacy are still not fully understood, prompting the need for deeper exploration into its therapeutic actions. AIM OF THE STUDY This study aims to elucidate the pharmacologically active ingredients and mechanisms of JQJTT in T2DM using multi-omics methods combined with experimental verification. MATERIALS AND METHODS Liquid chromatography-mass spectrometry (LC-MS) identified the chemical profile of JQJTT, and targets were screened via SwissTargetPrediction database. T2DM-related targets were acquired from GeneCards and Gene Expression Omnibus (GEO) databases. Therapeutic targets were ascertained by intersecting JQJTT and T2DM targets, followed by Protein-Protein Interaction (PPI) network analysis. GO and KEGG pathway enrichment was performed, and the compound-target-pathway network was built using Cytoscape. Mendelian randomization, single-sample Gene Set Enrichment Analysis (ssGSEA), and CIBERSORT analyses were executed to explore immune cell involvement. Single-cell RNA sequencing (scRNA-seq) identified affected cell types, while molecular docking and molecular dynamics (MD) simulation assessed compound-target affinities. Metabolomics and 16S rDNA sequencing analyzed JQJTT's effects on metabolites and gut microbiota. RESULTS LC-MS identified 161 compounds in JQJTT. Through network pharmacology, 6 core targets (AKT1, MMP9, HSP90AA1, CASP3, IL-6, PTGS2) were screened, and PI3K/AKT, NFκB, and lipid metabolism were pinpointed as key regulatory pathways. Mendelian randomization linked 39 immune cell types to T2DM, while ssGSEA and CIBERSORT suggested JQJTT potentially modulated the proportion of immune cells. scRNA-seq indicated JQJTT may affect macrophage polarization. Tangeritin, isorhamnetin, and kaempferol were determined as the main active compounds with glucose-lowering effects confirmed in vitro. In vivo experiments validated the molecular mechanisms of JQJTT against T2DM. Metabolomics unearthed 56 differentially abundant metabolites, primarily in linoleic acid, arachidonic acid, and pyruvaldehyde metabolisms. JQJTT also improved gut microbiota diversity in T2DM. CONCLUSION JQJTT treated T2DM through a multi-target, multi-pathway approach, including attenuating inflammation, reshaping macrophage polarization, enhancing insulin signaling, regulating metabolites, and restoring intestinal flora balance.
Collapse
Affiliation(s)
- Yining Gong
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| | - Xiaoqing Ma
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Jin Xing
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Zongwen Ji
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Xueying Gao
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Zhi Chen
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Fangzhi Hu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Dandan Yin
- Department of Clinical Medicine, Jining Medical University, Jining 272067, China.
| | - Feng Li
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Can Sheng
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining 272000, China.
| | - Yaping Liu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Shulong Shi
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China; Cisen Pharmaceutical Co., Ltd, Jining 272000, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
2
|
Zhou Y, Li S, Hong B, Wang Z, Shao Y, Wu M, Wang J. Mechanisms of isorhamnetin inhibition of osteoclast differentiation: insights from molecular dynamics simulations and in vitro/ in vivo experiments. Front Pharmacol 2025; 16:1551257. [PMID: 40356982 PMCID: PMC12066772 DOI: 10.3389/fphar.2025.1551257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Background Osteoporosis (OP) represents a widespread bone remodeling disorder within the domain of orthopedics, markedly compromising the quality of life in the elderly population. The need to develop more efficient therapeutic approaches to attenuate bone resorption by suppressing the excessive activation of osteoclasts (OCs) remains urgent. The plant flavonoid Isorhamnetin (Iso), recognized for its potent antioxidant properties, has been the subject of extensive research regarding its potential in treating bone-related conditions. Method This study adopts a comprehensive methodology to evaluate Iso's impact on bone metabolism and its therapeutic possibilities for treating OP. By integrating network pharmacology, molecular dynamics simulations, and surface plasmon resonance (SPR), we performed in vitro phenotypic analyses to systematically evaluate the inhibitory effect of Iso on OC differentiation. The mechanisms behind Iso's inhibition of OC differentiation were further elucidated. In vivo testing was also performed to substantiate the therapeutic effects of Iso in an OP animal model. Results At low concentrations, Iso showed no cytotoxicity and did not interfere with cell proliferation in RAW 264.7 cells. Iso effectively inhibited RANKL-induced osteoclast differentiation in these cells, while downregulating related genes levels (Nfatc1, Ctsk, Trap, c-Fos). Molecular dynamics simulations and surface plasmon resonance confirmed Iso's dual binding to both RANKL and RANK. KEGG pathway enrichment analysis results indicated that Iso modulates the MAPK, NF-κB/PI3K-AKT, and calcium signaling pathways. Western blot analysis revealed that Iso treatment targeting the RANKL/RANK binding pathway significantly downregulated phosphorylation levels of JNK, P38, AKT, and p65. Concurrently, Iso stimulation markedly increased IκBα expression, thereby rescuing its degradation. Furthermore, Iso demonstrated a robust inhibitory effect on reactive oxygen species levels in vitro. Furthermore, in OVX mice, Iso treatment increased bone density, modulated serum bone metabolism markers, and downregulated transcriptional levels of OC marker genes. Conclusion Iso exhibits therapeutic potential for OP by selectively targeting and disrupting the RANKL-RANK interaction. This intervention modulates the expression of intracellular transcription factors and multiple signaling pathways, thereby inhibiting the maturation of OCs. Through mitigating OC-mediated bone loss, Iso holds significant promise as a potent therapeutic agent for OP.
Collapse
Affiliation(s)
- Yi Zhou
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaoshuo Li
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Bowen Hong
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zihan Wang
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Shao
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Mao Wu
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Graduate School, Nanjing University of Chinese Medicine, Nanjing, China
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
3
|
Shi S, Tian X, Gong Y, Sun M, Liu J, Zhang J, Liu Y, Li L, Jiang S. Pivotal role of JNK protein in the therapeutic efficacy of parthenolide against breast cancer: Novel and comprehensive evidences from network pharmacology, single-cell RNA sequencing and metabolomics. Int J Biol Macromol 2024; 279:135209. [PMID: 39244135 DOI: 10.1016/j.ijbiomac.2024.135209] [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: 06/12/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
This study aimed to evaluate the efficacy and therapeutic mechanism of parthenolide (PTL) in breast cancer (BC) through a comprehensive strategy integrating network pharmacology, single-cell RNA sequencing (scRNA-seq) and metabolomics. In network pharmacology, 70 therapeutic targets were identified, of which 16 core targets were filtered out through seven classical algorithms of Cytohubba plugin. Additionally, the hub module of PPI network was extracted using MCODE plugin. Molecular docking and molecular dynamics simulation showed a potent binding affinity between PTL and JNK, subsequently validated by MST and SPR assays. Further, Mendelian randomization analysis indicated that JNK was causally associated with BC. GO and KEGG enrichment analyses revealed that PTL counteracted BC via promoting ROS generation, inducing apoptosis and suppressing proliferation, which potentially involved the coordinated regulation of MAPK and FoxO1 pathways. Moreover, ssGSEA and scRNA-seq analysis suggested that PTL may act on T cell immune microenvironment of BC. Subsequently, these bioinformatics-based predictions were experimentally validated using in-vitro and in-vivo models. Finally, metabolome profiling unveiled that PTL remodeled the glycine, serine and threonine metabolism as well as biosynthesis of unsaturated fatty acids, and thereby contributed to BC inhibition. From molecular, immune and metabolic perspectives, this study not only provided a unique insight into the mechanistic details of PTL against BC, but also proposed a novel promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Shulong Shi
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China; Department of Clinical Medicine, Jining Medical University, Jining 272013, China; Cisen Pharmaceutical Co., Ltd, Jining 272000, China; School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China
| | - Yining Gong
- Department of Clinical Medicine, Jining Medical University, Jining 272013, China
| | - Mingliang Sun
- Department of Endocrinology, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan 250000, China
| | - Juan Liu
- Shandong Rehabilitation Hospital, Jinan 250000, China
| | - Jiaqi Zhang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China
| | - Yaping Liu
- Department of Endocrinology, Jining First People's Hospital, Jining 272000, China.
| | - Luning Li
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining 272000, China.
| |
Collapse
|
4
|
Shende SU, Al-Shar'i NA, Saini SM, Mohanlall V, Gleiser RM, Deb PK, Morsy MA, Venugopala KN, Chandrashekharappa S. Synthesis, characterization and larvicidal studies of ethyl 3-benzoyl-7-(piperidin-1-yl)indolizine-1-carboxylate analogues against Anopheles arabiensis and cheminformatics approaches. J Biomol Struct Dyn 2024:1-13. [PMID: 38315452 DOI: 10.1080/07391102.2024.2311881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
According to WHO, in 2021, there was an estimation of 247 million malaria cases from 84 malaria-endemic countries. Globally an estimated count of 2 billion malaria cases and 11.7 million deaths due to malaria were recorded in the past two decades. Further, the emergence of drug-resistant mosquitos threatens mankind. Therefore, the development of newer larvicidal agents is the need of the hour. This research identifies a new series of variably substituted indolizines for their effectiveness in controlling Anopheles arabiensis larvae through larvicidal activity. The series of Ethyl 3-benzoyl-7-(piperidin-1-yl)indolizine-1-carboxylate analogues (4a-j) were synthesized by reacting 4-(piperidin-1-yl)pyridine, phenacyl bromides with ethyl propiolate via 1, 3-dipolar cycloaddition and the green metrics of the process are reported. All the newly synthesized compounds were characterized by spectroscopic techniques such as 1H NMR,13C NMR, FT-IR, and HRMS. The larvicidal effectiveness of the newly synthesized compounds was assessed against Anopheles arabiensis. Among the compounds studied, namely 4c, 4d, 4e, and 4f, displayed the most notable larval mortality rates within the series, reaching 73%, 81%, 76%, and 71% respectively, in contrast with the negative control acetone. In comparison, the standard Temephos exhibited a mortality rate of 99% at the same concentration. Furthermore, computational approaches including molecular docking and molecular dynamics simulations identified the potential targets of the series compounds as the larval Acetylcholinesterase (AChE) enzyme and the Sterol Carrier Protein-2 (SCP-2) protein. However, it is essential for these computational predictions to undergo experimental validation.
Collapse
Affiliation(s)
- Sondarya Uttam Shende
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Raebareli (NIPER-R), Lucknow, UP, India
| | - Nizar A Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Surbhi Mahender Saini
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Raebareli (NIPER-R), Lucknow, UP, India
| | - Viresh Mohanlall
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Raquel M Gleiser
- CREAN-IMBIV (UNC-CONICET), Av. Valparaiso s.n., and FCEFyN, Av. V. Sarsfield 299, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Pran Kishore Deb
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology (BIT), Mesra, Ranchi, Jharkhand, India
| | - Mohamed A Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| | - Katharigatta N Venugopala
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Sandeep Chandrashekharappa
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Raebareli (NIPER-R), Lucknow, UP, India
| |
Collapse
|
5
|
Yuan P, Chen W, Wang X, Li L, Peng Z, Mu S, You M, Xu H. RAGE: a potential target for Epimedium's anti-neuroinflammation role in vascular dementia-insights from network pharmacology and molecular simulation. J Biomol Struct Dyn 2023; 42:10856-10875. [PMID: 37732621 DOI: 10.1080/07391102.2023.2259480] [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: 06/19/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Vascular dementia (VaD), a cognitive impairment resulting from cerebrovascular issues, could be mitigated by Epimedium. This study investigates Epimedium's efficacy in VaD management through a systematic review, network pharmacology, molecular docking, and molecular dynamic simulations (MDS). Comprehensive literature searches were conducted across various databases. Epimedium's pharmacological properties were analyzed using the TCMSP database. Integration with the Aging Atlas database enabled the identification of shared targets between Epimedium and VaD. A protein-protein interaction (PPI) network was constructed, and central targets' topological attributes were analyzed using Cytoscape 3.9.1. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted using "ClusterProfiler" R package. The interactions between Epimedium and central targets were assessed by Molecular docking and MDS. Epimedium and its 23 bioactive components counteracted oxidative stress, neuroinflammation, and neuronal damage, thereby attenuating cognitive deterioration in VaD. A total of 78 common targets were identified, with 22 being significantly related to aging. Enrichment analysis identified 1769 GO terms and 139 KEGG pathways, highlighting the AGE-RAGE signaling pathway. Molecular docking revealed that 23 bioactive components, except Linoleyl acetate, effectively interacted with top central targets (JUN, MAPK14, IL6, FOS, TNF). MDS demonstrated that flavonoids Icariin, Kaempferol, Luteolin, and Quercetin formed stable complexes with RAGE. The study identifies RAGE as a novel therapeutic target for Epimedium in the mitigation of VaD via its anti-inflammatory properties.
Collapse
Affiliation(s)
- Ping Yuan
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Wei Chen
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Xiaohu Wang
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Liangqian Li
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Zijun Peng
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Song Mu
- Department of Colorectal Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Mingyao You
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Hongbei Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| |
Collapse
|
6
|
Nwaefulu ON, Al-Shar'i NA, Owolabi JO, Sagineedu SR, Woei LC, Wai LK, Islam MK, Jayanthi S, Stanslas J. The impact of cycleanine in cancer research: a computational study. J Mol Model 2022; 28:340. [PMID: 36194315 DOI: 10.1007/s00894-022-05326-1] [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: 04/08/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022]
Abstract
Cancer is imposing a global health burden because of the steady increase in new cases. Moreover, current anticancer therapeutics are associated with many drawbacks, mainly the emergence of resistance and the severe adverse effects. Therefore, there is a continuous need for developing new anticancer agents with novel mechanisms of action and lower side effects. Natural products have been a rich source of anticancer medication. Cycleanine, a natural product, was reported to exert an antiproliferative effect on ovarian cancer cells by causing apoptosis through activation of caspases 3/7 and cleavage of poly (ADP-ribose) polymerase to form poly (ADP-ribose) polymerase-1 (PARP1). It is well-established that PARP1 is associated with carcinogenesis, and different PARP1 inhibitors are approved as anticancer drugs. In this study, the cytotoxic activity of cycleanine was computationally investigated to determine whether it is a PARP1 inhibitor or a caspase activator. Molecular docking and molecular dynamics (MD) simulations were utilized for this purpose. The results showed that cycleanine has a good binding affinity to PARP1; moreover, MD simulation showed that it forms a stable complex with the enzyme. Consequently, the results showed that cycleanine is a potential inhibitor of the PARP1 enzyme.
Collapse
Affiliation(s)
- Ogochukwu Ngozi Nwaefulu
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Nizar A Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Josephine Omonkhelin Owolabi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, Edo State, Nigeria
| | - Sreenivasa Rao Sagineedu
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Lim Chee Woei
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Lam Kok Wai
- Centre for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, 50300, Kuala Lumpur, Selangor, Malaysia
| | - Mohammad Kaisarul Islam
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Sivaraman Jayanthi
- Computational Drug Design Lab, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore-632014, Tamil Nadu, India
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| |
Collapse
|
7
|
Cao JF, Gong Y, Wu M, Yang X, Xiong L, Chen S, Xiao Z, Li Y, Zhang L, Zan W, Zhang X. Exploring the mechanism of action of licorice in the treatment of COVID-19 through bioinformatics analysis and molecular dynamics simulation. Front Pharmacol 2022; 13:1003310. [PMID: 36120307 PMCID: PMC9481297 DOI: 10.3389/fphar.2022.1003310] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/11/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose: The rapid worldwide spread of Corona Virus Disease 2019 (COVID-19) has become not only a global challenge, but also a lack of effective clinical treatments. Studies have shown that licorice can significantly improve clinical symptoms such as fever, dry cough and shortness of breath in COVID-19 patients with no significant adverse effects. However, there is still a lack of in-depth analysis of the specific active ingredients of licorice in the treatment of COVID-19 and its mechanism of action. Therefore, we used molecular docking and molecular dynamics to explore the mechanism of action of licorice in the treatment of COVID-19.Methods: We used bioinformatics to screen active pharmaceutical ingredients and potential targets, the disease-core gene target-drug network was established and molecular docking was used for verification. Molecular dynamics simulations were carried out to verify that active ingredients were stably combined with protein targets. The supercomputer platform was used to measure and analyze stability of protein targets at the residue level, solvent accessible surface area, number of hydrogen bonds, radius of gyration and binding free energy.Results: Licorice had 255 gene targets, COVID-19 had 4,628 gene targets, the intersection gene targets were 101. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene ontology (GO) analysis showed that licorice played an important role mainly through the signaling pathways of inflammatory factors and oxidative stress. Molecular docking showed that Glycyrol, Phaseol and Glyasperin F in licorice may playe a role in treating COVID-19 by acting on STAT3, IL2RA, MMP1, and CXCL8. Molecular dynamics were used to demonstrate and analyze the binding stability of active ingredients to protein targets.Conclusion: This study found that Phaseol in licorice may reduce inflammatory cell activation and inflammatory response by inhibiting the activation of CXCL8 and IL2RA; Glycyrol may regulate cell proliferation and survival by acting on STAT3. Glyasperin F may regulate cell growth by inhibiting the activation of MMP1, thus reducing tissue damage and cell death caused by excessive inflammatory response and promoting the growth of new tissues. Therefore, licorice is proposed as an effective candidate for the treatment of COVID-19 through STAT3, IL2RA, MMP1, and CXCL8.
Collapse
Affiliation(s)
- Jun-Feng Cao
- Clinical Medicine, Chengdu Medical College, Chengdu, China
- Chengdu Medical College of Basic Medical Sciences, Chengdu, China
| | - Yunli Gong
- Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Mei Wu
- Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Xingyu Yang
- Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Li Xiong
- Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Shengyan Chen
- Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Zixuan Xiao
- Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Yang Li
- Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Lixin Zhang
- Yunnan Academy of Forestry Sciences, Kunming, China
- *Correspondence: Lixin Zhang, , orcid.org/0086-135-2919-4157; Wang Zan, , orcid.org/0086-186-2812-0209; Xiao Zhang, , orcid.org/0086-130-8661-6376
| | - Wang Zan
- Chengdu Medical College of Pharmacy, Chengdu, China
- *Correspondence: Lixin Zhang, , orcid.org/0086-135-2919-4157; Wang Zan, , orcid.org/0086-186-2812-0209; Xiao Zhang, , orcid.org/0086-130-8661-6376
| | - Xiao Zhang
- Chengdu Medical College of Basic Medical Sciences, Chengdu, China
- *Correspondence: Lixin Zhang, , orcid.org/0086-135-2919-4157; Wang Zan, , orcid.org/0086-186-2812-0209; Xiao Zhang, , orcid.org/0086-130-8661-6376
| |
Collapse
|
8
|
Antitubercular, Cytotoxicity, and Computational Target Validation of Dihydroquinazolinone Derivatives. Antibiotics (Basel) 2022; 11:antibiotics11070831. [PMID: 35884084 PMCID: PMC9311641 DOI: 10.3390/antibiotics11070831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 02/04/2023] Open
Abstract
A series of 2,3-dihydroquinazolin-4(1H)-one derivatives (3a–3m) was screened for in vitro whole-cell antitubercular activity against the tubercular strain H37Rv and multidrug-resistant (MDR) Mycobacterium tuberculosis (MTB) strains. Compounds 3l and 3m with di-substituted aryl moiety (halogens) attached to the 2-position of the scaffold showed a minimum inhibitory concentration (MIC) of 2 µg/mL against the MTB strain H37Rv. Compound 3k with an imidazole ring at the 2-position of the dihydroquinazolin-4(1H)-one also showed significant inhibitory action against both the susceptible strain H37Rv and MDR strains with MIC values of 4 and 16 µg/mL, respectively. The computational results revealed the mycobacterial pyridoxal-5′-phosphate (PLP)-dependent aminotransferase (BioA) enzyme as the potential target for the tested compounds. In vitro, ADMET calculations and cytotoxicity studies against the normal human dermal fibroblast cells indicated the safety and tolerability of the test compounds 3k–3m. Thus, compounds 3k–3m warrant further optimization to develop novel BioA inhibitors for the treatment of drug-sensitive H37Rv and drug-resistant MTB.
Collapse
|
9
|
Design, synthesis, and biological evaluation of SMYD3 inhibitors possessing N-thiazole benzenesulfonamide moiety as potential anti-cancer agents. JOURNAL OF SAUDI CHEMICAL SOCIETY 2022. [DOI: 10.1016/j.jscs.2022.101482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
10
|
Díaz-Muñoz M, Hernández-Muñoz R, Butanda-Ochoa A. Structure-activity features of purines and their receptors: implications in cell physiopathology. MOLECULAR BIOMEDICINE 2022; 3:5. [PMID: 35079944 PMCID: PMC8789959 DOI: 10.1186/s43556-022-00068-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/19/2022] [Indexed: 11/21/2022] Open
Abstract
The purine molecular structure consists of fused pyrimidine and imidazole rings. Purines are main pieces that conform the structure of nucleic acids which rule the inheritance processes. Purines also work as metabolic intermediates in different cell functions and as messengers in the signaling pathways throughout cellular communication. Purines, mainly ATP and adenosine (ADO), perform their functional and pharmacological properties because of their structural/chemical characteristics that make them either targets of mutagenesis, mother frameworks for designing molecules with controlled effects (e.g. anti-cancer), or chemical donors (e.g., of methyl groups, which represent a potential chemoprotective action against cancer). Purines functions also come from their effect on specific receptors, channel-linked and G-protein coupled for ATP, and exclusively G-coupled receptors for ADO (also known as ADORAs), which are involved in cell signaling pathways, there, purines work as chemical messengers with autocrine, paracrine, and endocrine actions that regulate cell metabolism and immune response in tumor progression which depends on the receptor types involved in these signals. Purines also have antioxidant and anti-inflammatory properties and participate in the cell energy homeostasis. Therefore, purine physiology is important for a variety of functions relevant to cellular health; thus, when these molecules present a homeostatic imbalance, the stability and survival of the cellular systems become compromised.
Collapse
Affiliation(s)
- Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM, Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular Y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Ciudad Universitaria/Circuito Exterior, C.P. 04510, Ciudad de México, México
| | - Armando Butanda-Ochoa
- Departamento de Biología Celular Y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Ciudad Universitaria/Circuito Exterior, C.P. 04510, Ciudad de México, México.
| |
Collapse
|
11
|
Fakhouri LI, Al-Shar'i NA. The design of TOPK inhibitors using structure-based pharmacophore modeling and molecular docking based on an MD-refined homology model. Mol Divers 2022; 26:2679-2702. [PMID: 35031933 DOI: 10.1007/s11030-021-10361-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 12/01/2021] [Indexed: 11/27/2022]
Abstract
The TOPK enzyme (also known as PBK) is a serine-threonine protein kinase that is rarely detected in normal tissues yet is found to be overexpressed and activated in a variety of cancers such as lung, colorectal, breast, and esophageal cancer. Its prevalence in cancerous cells is associated with their poor prognosis and responsiveness to treatment. This enzyme plays a vital role in cell division, specifically in regulating cytokinesis. Unlike drugs targeting early phases in mitosis, inhibition of cytokinesis by targeting biomolecules that are unique to multiplying cells poses no threat to the normal function of non-multiplying cells. Studies have shown that inhibition of cytokinesis is promising in suppressing the growth of proliferating cancerous cells as exemplified by the complete tumor regression seen with the suppression of TOPK. Herein, we report the identification of potent TOPK inhibitors with anticancer potential using a structure-based drug design approach. The only available crystal structure of TOPK corresponds to a double mutant (T9E and T198E) dimer with a distorted N-lobe conformation, thus 3D homology modeling was implemented to rebuild the enzyme's native conformation. The resulting refined model was used to generate 3D pharmacophore models for the virtual screening of small molecules databases. Retrieved hits were filtered, docked into the ATP binding site of the enzyme, rescored, and the binding free energies for the top consensually scoring hits were calculated. Consequently, 45 compounds were selected and their in vitro inhibitory activity against TOPK was tested revealing four potential hits with the most active compound having an IC50 of 3.85 µM. This compound will be chosen as a lead compound to synthesize analogs aiming to enhance potency and drug-like properties and to enrich the SAR data.
Collapse
Affiliation(s)
- Lara I Fakhouri
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Nizar A Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| |
Collapse
|
12
|
Deb PK, Al-Shar’i NA, Venugopala KN, Pillay M, Borah P. In vitro anti-TB properties, in silico target validation, molecular docking and dynamics studies of substituted 1,2,4-oxadiazole analogues against Mycobacterium tuberculosis. J Enzyme Inhib Med Chem 2021; 36:869-884. [PMID: 34060396 PMCID: PMC8172222 DOI: 10.1080/14756366.2021.1900162] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
The alarming increase in multi- and extensively drug-resistant (MDR and XDR) strains of Mycobacterium tuberculosis (MTB) has triggered the scientific community to search for novel, effective, and safer therapeutics. To this end, a series of 3,5-disubstituted-1,2,4-oxadiazole derivatives (3a-3i) were tested against H37Rv, MDR and XDR strains of MTB. Of which, compound 3a with para-trifluorophenyl substituted oxadiazole showed excellent activity against the susceptible H37Rv and MDR-MTB strain with a MIC values of 8 and 16 µg/ml, respectively.To understand the mechanism of action of these compounds (3a-3i) and identify their putative drug target, molecular docking and dynamics studies were employed against a panel of 20 mycobacterial enzymes reported to be essential for mycobacterial growth and survival. These computational studies revealed polyketide synthase (Pks13) enzyme as the putative target. Moreover, in silico ADMET predictions showed satisfactory properties for these compounds, collectively, making them, particularly compound 3a, promising leads worthy of further optimisation.
Collapse
Affiliation(s)
- Pran Kishore Deb
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, Amman, Jordan
| | - Nizar A. Al-Shar’i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
- Department of Biotechnology and Food Technology, Durban University of Technology, Durban, South Africa
| | - Melendhran Pillay
- Department of Microbiology, National Health Laboratory Services, KZN Academic Complex, Inkosi Albert Luthuli Central Hospital, Durban, South Africa
| | - Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, India
| |
Collapse
|
13
|
Methodological approaches for the analysis of transmembrane domain interactions: A systematic review. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183712. [PMID: 34331948 DOI: 10.1016/j.bbamem.2021.183712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/28/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022]
Abstract
The study of protein-protein interactions (PPI) has proven fundamental for the understanding of the most relevant cell processes. Any protein domain can participate in PPI, including transmembrane (TM) segments that can establish interactions with other TM domains (TMDs). However, the hydrophobic nature of TMDs and the environment they occupy complicates the study of intramembrane PPI, which demands the use of specific approaches and techniques. In this review, we will explore some of the strategies available to study intramembrane PPI in vitro, in vivo, and, in silico, focusing on those techniques that could be carried out in a standard molecular biology laboratory regarding its previous experience with membrane proteins.
Collapse
|
14
|
Salmaso V, Jain S, Jacobson KA. Purinergic GPCR transmembrane residues involved in ligand recognition and dimerization. Methods Cell Biol 2021; 166:133-159. [PMID: 34752329 PMCID: PMC8620127 DOI: 10.1016/bs.mcb.2021.06.001] [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] [Indexed: 09/25/2023]
Abstract
We compare the GPCR-ligand interactions and highlight important residues for recognition in purinergic receptors-from both X-ray crystallographic and cryo-EM structures. These include A1 and A2A adenosine receptors, and P2Y1 and P2Y12 receptors that respond to ADP and other nucleotides. These receptors are important drug discovery targets for immune, metabolic and nervous system disorders. In most cases, orthosteric ligands are represented, except for one allosteric P2Y1 antagonist. This review catalogs the residues and regions that engage in contacts with ligands or with other GPCR protomers in dimeric forms. Residues that are in proximity to bound ligands within purinergic GPCR families are correlated. There is extensive conservation of recognition motifs between adenosine receptors, but the P2Y1 and P2Y12 receptors are each structurally distinct in their ligand recognition. Identifying common interaction features for ligand recognition within a receptor class that has multiple structures available can aid in the drug discovery process.
Collapse
Affiliation(s)
- Veronica Salmaso
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
15
|
CHK1 kinase inhibition: identification of allosteric hits using MD simulations, pharmacophore modeling, docking and MM-PBSA calculations. Mol Divers 2021; 26:903-921. [PMID: 33686514 DOI: 10.1007/s11030-021-10202-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/16/2021] [Indexed: 12/09/2022]
Abstract
The CHK1 kinase plays a pivotal role in the DNA damage response pathway. Hence, inhibition of CHK1 appeared as a promising strategy to overcome the resistance problem of chemotherapeutic agents resulting from the overexpression of CHK1 that enables cancerous cells to repair their chemotherapy-induced DNA damage. In this study, different computational drug design techniques were employed to identify new CHK1 inhibitors targeting its allosteric pocket. A 1 μs MD simulation of the apo form of the enzyme was run to study its native dynamics. The resulting trajectory was analyzed to select a frame where the ATP binding pocket is most occluded while its allosteric counterpart is most exposed to be used in the design of potential allosteric inhibitors that could trap the enzyme in such nearly inactive state. Besides the selected frame, another three crystal structures of CHK1 complexed with allosteric inhibitors were utilized to generate structure-based pharmacophore models. Seven pharmacophores were generated and utilized in virtual screening of different databases. The retrieved hits were filtered and then docked into the allosteric pocket. Finally, the binding energies of the top-ranked docked hits were calculated. Twenty compounds were selected as candidates for biological evaluation against CHK1 enzyme. The biological screening results showed moderate activities where the percentage of CHK1 inhibition ranged from zero to 28.26%. Four of the tested compounds showed percentage of CHK1 inhibition greater than 20%, of which, two compounds were identified as allosteric hits that upon further optimization could be converted into lead-like compounds.
Collapse
|
16
|
Shakya AK, Naik RR, Almasri IM, Kaur A. Role and Function of Adenosine and its Receptors in Inflammation, Neuroinflammation, IBS, Autoimmune Inflammatory Disorders, Rheumatoid Arthritis and Psoriasis. Curr Pharm Des 2020; 25:2875-2891. [PMID: 31333103 DOI: 10.2174/1381612825666190716145206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
The physiological effects of endogenous adenosine on various organ systems are very complex and numerous which are elicited upon activation of any of the four G-protein-coupled receptors (GPCRs) denoted as A1, A2A, A2B and A3 adenosine receptors (ARs). Several fused heterocyclic and non-xanthine derivatives are reported as a possible target for these receptors due to physiological problems and lack of selectivity of xanthine derivatives. In the present review, we have discussed the development of various new chemical entities as a target for these receptors. In addition, compounds acting on adenosine receptors can be utilized in treating diseases like inflammation, neuroinflammation, autoimmune and related diseases.
Collapse
Affiliation(s)
- Ashok K Shakya
- Medicinal Chemistry, Drug Design and Drug Metabolism, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al- Ahliyya Amman University, PO Box 263, Amman 19328, Jordan
| | - Rajashri R Naik
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ihab M Almasri
- Medicinal Chemistry and Drug Design, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Al Azhar University Gaza, Gaza Strip, Palestinian Territory, Occupied
| | - Avneet Kaur
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Mehrauli-Badarpur Road, Pushp Vihar, Sector-3, New Delhi-110017, India
| |
Collapse
|
17
|
Choudhury H, Chellappan DK, Sengupta P, Pandey M, Gorain B. Adenosine Receptors in Modulation of Central Nervous System Disorders. Curr Pharm Des 2020; 25:2808-2827. [PMID: 31309883 DOI: 10.2174/1381612825666190712181955] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
The ubiquitous signaling nucleoside molecule, adenosine is found in different cells of the human body to provide its numerous pharmacological role. The associated actions of endogenous adenosine are largely dependent on conformational change of the widely expressed heterodimeric G-protein-coupled A1, A2A, A2B, and A3 adenosine receptors (ARs). These receptors are well conserved on the surface of specific cells, where potent neuromodulatory properties of this bioactive molecule reflected by its easy passage through the rigid blood-brainbarrier, to simultaneously act on the central nervous system (CNS). The minimal concentration of adenosine in body fluids (30-300 nM) is adequate to exert its neuromodulatory action in the CNS, whereas the modulatory effect of adenosine on ARs is the consequence of several neurodegenerative diseases. Modulatory action concerning the activation of such receptors in the CNS could be facilitated towards neuroprotective action against such CNS disorders. Our aim herein is to discuss briefly pathophysiological roles of adenosine on ARs in the modulation of different CNS disorders, which could be focused towards the identification of potential drug targets in recovering accompanying CNS disorders. Researches with active components with AR modulatory action have been extended and already reached to the bedside of the patients through clinical research in the improvement of CNS disorders. Therefore, this review consist of recent findings in literatures concerning the impact of ARs on diverse CNS disease pathways with the possible relevance to neurodegeneration.
Collapse
Affiliation(s)
- Hira Choudhury
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Dinesh K Chellappan
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Pallav Sengupta
- Department of Physiology, Faculty of Medicine, MA`HSA University, Kuala Lumpur, Malaysia
| | - Manisha Pandey
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
18
|
Borah P, Deka S, Mailavaram RP, Deb PK. P1 Receptor Agonists/Antagonists in Clinical Trials - Potential Drug Candidates of the Future. Curr Pharm Des 2020; 25:2792-2807. [PMID: 31333097 DOI: 10.2174/1381612825666190716111245] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Adenosine mediates various physiological and pathological conditions by acting on its four P1 receptors (A1, A2A, A2B and A3 receptors). Omnipresence of P1 receptors and their activation, exert a wide range of biological activities. Thus, its modulation is implicated in various disorders like Parkinson's disease, asthma, cardiovascular disorders, cancer etc. Hence these receptors have become an interesting target for the researchers to develop potential therapeutic agents. Number of molecules were designed and developed in the past few years and evaluated for their efficacy in various disease conditions. OBJECTIVE The main objective is to provide an overview of new chemical entities which have crossed preclinical studies and reached clinical trials stage following their current status and future prospective. METHODS In this review we discuss current status of the drug candidates which have undergone clinical trials and their prospects. RESULTS Many chemical entities targeting various subtypes of P1 receptors are patented; twenty of them have crossed preclinical studies and reached clinical trials stage. Two of them viz adenosine and regadenoson are approved by the Food and Drug Administration. CONCLUSION This review is an attempt to highlight the current status, progress and probable future of P1 receptor ligands which are under clinical trials as promising novel therapeutic agents and the direction in which research should proceed with a view to come out with novel therapeutic agents.
Collapse
Affiliation(s)
- Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Panikhaiti, Chandrapur Road, Guwahati, Assam, India
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Panikhaiti, Chandrapur Road, Guwahati, Assam, India
| | - Raghu Prasad Mailavaram
- Department of Pharmaceutical Chemistry, Shri Vishnu College of Pharmacy, Vishnupur (Affiliated to Andhra University), Bhimavaram, W.G. Dist., AP, India
| | - Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, Amman, PO Box-1, 19392, Jordan
| |
Collapse
|
19
|
Deb PK, Deka S, Borah P, Abed SN, Klotz KN. Medicinal Chemistry and Therapeutic Potential of Agonists, Antagonists and Allosteric Modulators of A1 Adenosine Receptor: Current Status and Perspectives. Curr Pharm Des 2020; 25:2697-2715. [PMID: 31333094 DOI: 10.2174/1381612825666190716100509] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 12/28/2022]
Abstract
Adenosine is a purine nucleoside, responsible for the regulation of a wide range of physiological and pathophysiological conditions by binding with four G-protein-coupled receptors (GPCRs), namely A1, A2A, A2B and A3 adenosine receptors (ARs). In particular, A1 AR is ubiquitously present, mediating a variety of physiological processes throughout the body, thus represents a promising drug target for the management of various pathological conditions. Agonists of A1 AR are found to be useful for the treatment of atrial arrhythmia, angina, type-2 diabetes, glaucoma, neuropathic pain, epilepsy, depression and Huntington's disease, whereas antagonists are being investigated for the treatment of diuresis, congestive heart failure, asthma, COPD, anxiety and dementia. However, treatment with full A1 AR agonists has been associated with numerous challenges like cardiovascular side effects, off-target activation as well as desensitization of A1 AR leading to tachyphylaxis. In this regard, partial agonists of A1 AR have been found to be beneficial in enhancing insulin sensitivity and subsequently reducing blood glucose level, while avoiding severe CVS side effects and tachyphylaxis. Allosteric enhancer of A1 AR is found to be potent for the treatment of neuropathic pain, culminating the side effects related to off-target tissue activation of A1 AR. This review provides an overview of the medicinal chemistry and therapeutic potential of various agonists/partial agonists, antagonists and allosteric modulators of A1 AR, with a particular emphasis on their current status and future perspectives in clinical settings.
Collapse
Affiliation(s)
- Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, PO Box - 1, 19392, Amman, Jordan
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Pobitra Borah
- Pratiksha Institute of Pharmaceutical Sciences, Chandrapur Road, Panikhaiti, Guwahati-26, Assam, India
| | - Sara N Abed
- Faculty of Pharmacy, Philadelphia University, PO Box - 1, 19392, Amman, Jordan
| | - Karl-Norbert Klotz
- University of Würzburg, Department of Pharmacology and Toxicology Versbacher Str. 9, D-97078 Würzburg, Germany
| |
Collapse
|
20
|
Chandrasekaran B, Samarneh S, Jaber AMY, Kassab G, Agrawal N. Therapeutic Potentials of A2B Adenosine Receptor Ligands: Current Status and Perspectives. Curr Pharm Des 2020; 25:2741-2771. [PMID: 31333084 DOI: 10.2174/1381612825666190717105834] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adenosine receptors (ARs) are classified as A1, A2A, A2B, and A3 subtypes belong to the superfamily of G-protein coupled receptors (GPCRs). More than 40% of modern medicines act through either activation or inhibition of signaling processes associated with GPCRs. In particular, A2B AR signaling pathways are implicated in asthma, inflammation, cancer, ischemic hyperfusion, diabetes mellitus, cardiovascular diseases, gastrointestinal disorders, and kidney disease. METHODS This article reviews different disease segments wherein A2B AR is implicated and discusses the potential role of subtype-selective A2B AR ligands in the management of such diseases or disorders. All the relevant publications on this topic are reviewed and presented scientifically. RESULTS This review provides an up-to-date highlight of the recent advances in the development of novel and selective A2B AR ligands and their therapeutic role in treating various disease conditions. A special focus has been given to the therapeutic potentials of selective A2B AR ligands in the management of airway inflammatory conditions and cancer. CONCLUSIONS This systematic review demonstrates the current status and perspectives of A2B AR ligands as therapeutically useful agents that would assist medicinal chemists and pharmacologists in discovering novel and subtype-selective A2B AR ligands as potential drug candidates.
Collapse
Affiliation(s)
- Balakumar Chandrasekaran
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Sara Samarneh
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Abdul Muttaleb Yousef Jaber
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Ghadir Kassab
- Faculty of Pharmacy, Philadelphia University-Jordan, P. O. Box: 1, Philadelphia University-19392, Amman, Jordan
| | - Nikhil Agrawal
- College of Health Sciences, University of KwaZulu-Natal, P. O. Box: 4000, Westville, Durban, South Africa
| |
Collapse
|
21
|
Pal Y, Bandyopadhyay N, Pal RS, Ahmed S, Bandopadhyay S. Perspective and Potential of A2A and A3 Adenosine Receptors as Therapeutic Targets for the Treatment of Rheumatoid Arthritis. Curr Pharm Des 2019; 25:2859-2874. [DOI: 10.2174/1381612825666190710111658] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 01/10/2023]
Abstract
Adenosine is a purine nucleoside which is an effective controller of inflammation. The inflammatory effect of adenosine is expressed via its four receptor subtypes viz. A1, A2A, A2B and A3. The various inflammatory conditions including rheumatoid arthritis (RA) are initiated by adenosine receptors of which A2A and A3 play a vital role. RA primarily is an auto-immune disorder which is manifested as chronic inflammation in the synovial lining of joints. In order to develop an effective treatment, the role of cytokines, IL–1, TNF-α and IL–6 is crucial. Besides, the knowledge of PI3K-PKB/Akt and NF-kB signaling pathway is also important to understand the antiinflammatory targets. Methotrexate along with various other molecules like, NSAIDs and DMARDs are presently used as treatment lines for controlling RA. The enhanced knowledge of the preclinical stages and pathogenesis along with recent potent therapeutics raises the hopes that RA can be prevented in the near future.
Collapse
Affiliation(s)
- Yogendra Pal
- Department of Pharmacy, Pranveer Singh Institute of Technology, Bhauti, Kanpur, Uttar Pradesh 209305, India
| | - Nabamita Bandyopadhyay
- Molecular Biology Division, National Institute of Malarial Research (NIMR), Dwarka, New Delhi, Delhi 110077, India
| | - Rashmi S. Pal
- Department of Pharmacy, Pranveer Singh Institute of Technology, Bhauti, Kanpur, Uttar Pradesh 209305, India
| | - Sarfaraz Ahmed
- Global Institute of Pharmaceutical Education and Research, Kashipur, Udham Singh Nagar, Uttarakhand 244713, India
| | - Shantanu Bandopadhyay
- Faculty of Pharmacy, Naraina Vidya Peeth Group of Institutions, Panki, Kanpur, Uttar Pradesh 208020, India
| |
Collapse
|
22
|
Gorain B, Choudhury H, Yee GS, Bhattamisra SK. Adenosine Receptors as Novel Targets for the Treatment of Various Cancers. Curr Pharm Des 2019; 25:2828-2841. [DOI: 10.2174/1381612825666190716102037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/01/2019] [Indexed: 01/28/2023]
Abstract
Adenosine is a ubiquitous signaling nucleoside molecule, released from different cells within the body
to act on vasculature and immunoescape. The physiological action on the proliferation of tumour cell has been
reported by the presence of high concentration of adenosine within the tumour microenvironment, which results
in the progression of the tumour, even leading to metastases. The activity of adenosine exclusively depends upon
the interaction with four subtypes of heterodimeric G-protein-coupled adenosine receptors (AR), A1, A2A, A2B,
and A3-ARs on the cell surface. Research evidence supports that the activation of those receptors via specific
agonist or antagonist can modulate the proliferation of tumour cells. The first category of AR, A1 is known to play
an antitumour activity via tumour-associated microglial cells to prevent the development of glioblastomas.
A2AAR are found in melanoma, lung, and breast cancer cells, where tumour proliferation is stimulated due to
inhibition of the immune response via inhibition of natural killer cells cytotoxicity, T cell activity, and tumourspecific
CD4+/CD8+ activity. Alternatively, A2BAR helps in the development of tumour upon activation via
upregulation of angiogenin factor in the microvascular endothelial cells, inhibition of MAPK and ERK 1/2 phosphorylation
activity. Lastly, A3AR is expressed in low levels in normal cells whereas the expression is upregulated
in tumour cells, however, agonists to this receptor inhibit tumour proliferation through modulation of Wnt
and NF-κB signaling pathways. Several researchers are in search for potential agents to modulate the overexpressed
ARs to control cancer. Active components of A2AAR antagonists and A3AR agonists have already entered
in Phase-I clinical research to prove their safety in human. This review focused on novel research targets towards
the prevention of cancer progression through stimulation of the overexpressed ARs with the hope to protect lives
and advance human health.
Collapse
Affiliation(s)
- Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Science, Taylor’s University, Subang Jaya, Selangor, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Gan Sook Yee
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Deb PK. Progress in the Development of Agonists, Antagonists and Allosteric Modulators of Adenosine Receptors. Curr Pharm Des 2019; 25:2695-2696. [DOI: 10.2174/138161282525190916100149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, PO Box 1, 19392, Amman, Jordan
| |
Collapse
|
24
|
Al-Attraqchi OH, Attimarad M, Venugopala KN, Nair A, Al-Attraqchi NH. Adenosine A2A Receptor as a Potential Drug Target - Current Status and Future Perspectives. Curr Pharm Des 2019; 25:2716-2740. [DOI: 10.2174/1381612825666190716113444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
Abstract
Adenosine receptors (ARs) are a class of G-protein coupled receptors (GPCRs) that are activated by
the endogenous substance adenosine. ARs are classified into 4 subtype receptors, namely, the A1, A2A, A2B and A3
receptors. The wide distribution and expression of the ARs in various body tissues as well as the roles they have
in controlling different functions in the body make them potential drug targets for the treatment of various pathological
conditions, such as cardiac diseases, cancer, Parkinson’s disease, inflammation and glaucoma. Therefore,
in the past decades, there have been extensive investigations of ARs with a high number of agonists and antagonists
identified that can interact with these receptors. This review shall discuss the A2A receptor (A2AAR) subtype
of the ARs. The structure, properties and the recent advances in the therapeutic potential of the receptor are discussed
with an overview of the recent advances in the methods of studying the receptor. Also, molecular modeling
approaches utilized in the design of A2AAR ligands are highlighted with various recent examples.
Collapse
Affiliation(s)
- Omar H.A. Al-Attraqchi
- Faculty of Pharmacy, Philadelphia University-Jordan, P.O BOX (1), Philadelphia University-19392, Amman, Jordan
| | - Mahesh Attimarad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Katharigatta N. Venugopala
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Anroop Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | | |
Collapse
|
25
|
Mailavaram RP, Al-Attraqchi OH, Kar S, Ghosh S. Current Status in the Design and Development of Agonists and Antagonists of Adenosine A3 Receptor as Potential Therapeutic Agents. Curr Pharm Des 2019; 25:2772-2787. [DOI: 10.2174/1381612825666190716114056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022]
Abstract
Adenosine receptors (ARs) belongs to the family of G-protein coupled receptors (GPCR) that are responsible
for the modulation of a wide variety of physiological functions. The ARs are also implicated in many
diseases such as cancer, arthritis, cardiovascular and renal diseases. The adenosine A3 receptor (A3AR) has
emerged as a potential drug target for the progress of new and effective therapeutic agents for the treatment of
various pathological conditions. This receptor’s involvement in many diseases and its validity as a target has been
established by many studies. Both agonists and antagonists of A3AR have been extensively investigated in the last
decade with the goal of developing novel drugs for treating diseases related to immune disorders, inflammation,
cancer, and others. In this review, we shall focus on the medicinal chemistry of A3AR ligands, exploring the
diverse chemical classes that have been projected as future leading drug candidates. Also, the recent advances in
the therapeuetic applications of A3AR ligands are highlighted.
Collapse
Affiliation(s)
- Raghu P. Mailavaram
- Department of Pharmaceutical Chemistry, Shri Vishnu College of Pharmacy, Vishnupur (Affiliated to Andhra University), Bhimavaram, W.G. Dist., AP, India
| | - Omar H.A. Al-Attraqchi
- Faculty of Pharmacy, Philadelphia University-Jordan, P.O BOX (1), Philadelphia University- 19392, Amman, Jordan
| | - Supratik Kar
- Interdisciplinary Center for Nanotoxicity, Department of Chemistry, Physics and Atmospheric Sciences, Jackson State University, Jackson, MS 39217, United States
| | - Shinjita Ghosh
- School of Public Health, Jackson State University, Jackson, MS 39217, United States
| |
Collapse
|
26
|
Deb PK. Recent Updates in the Computer Aided Drug Design Strategies for the Discovery of Agonists and Antagonists of Adenosine Receptors. Curr Pharm Des 2019; 25:747-749. [DOI: 10.2174/1381612825999190515120510] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Pran Kishore Deb
- Faculty of Pharmacy, Philadelphia University, PO Box 1, 19392, Amman, Jordan
| |
Collapse
|