1
|
Xiang G, Yang L, Qin J, Wang S, Zhang Y, Yang S. Revealing the potential bioactive components and mechanism of Qianhua Gout Capsules in the treatment of gouty arthritis through network pharmacology, molecular docking and pharmacodynamic study strategies. Heliyon 2024; 10:e30983. [PMID: 38770346 PMCID: PMC11103544 DOI: 10.1016/j.heliyon.2024.e30983] [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: 01/08/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
Recent clinical studies have confirmed the effectiveness of Qianhua Gout Capsules (QGC) in the treatment of gouty arthritis (GA). However, the specific regulatory targets and mechanisms of action of QGC are still unclear. To address this gap, we utilized network pharmacology, molecular docking, and pharmacodynamic approaches to investigate the bioactive components and associated mechanisms of QGC in the treatment of GA. By employing UPLC-Q Exactive-MS, we identified the compounds present in QGC, with active ingredients defined as those with oral bioavailability ≥30 % and drug similarity ≥0.18. Subsequently, the targets of these active compounds were determined using the TCMSP database, while GA-related targets were identified from DisGeNET, GeneCards, TTD, OMIM, and DrugBank databases. Further analysis including PPI analysis, GO analysis, and KEGG pathway enrichment was conducted on the targets. Validation of the predicted results was performed using a GA rat model, evaluating pathological changes, inflammatory markers, and pathway protein expression. Our results revealed a total of 130 components, 44 active components, 16 potential shared targets, GO-enriched terms, and 47 signaling pathways related to disease targets. Key active ingredients included quercetin, kaempferol, β-sitosterol, luteolin, and wogonin. The PPI analysis highlighted five targets (PPARG, IL-6, MMP-9, IL-1β, CXCL-8) with the highest connectivity, predominantly enriched in the IL-17 signaling pathway. Molecular docking experiments demonstrated strong binding of CXCL8, IL-1β, IL-6, MMP9, and PPARG targets with the top five active compounds. Furthermore, animal experiments confirmed the efficacy of QGC in treating GA in rats, showing reductions in TNF-α, IL-6, and MDA levels, and increases in SOD levels in serum. In synovial tissues, QGC treatment upregulated CXCL8 and PPARG expression, while downregulating IL-1β, MMP9, and IL-6 expression. In conclusion, this study applied a network pharmacology approach to uncover the composition of QGC, predict its pharmacological interactions, and demonstrate its in vivo efficacy, providing insights into the anti-GA mechanisms of QGC. These findings pave the way for future investigations into the therapeutic mechanisms underlying QGC's effectiveness in the treatment of GA.
Collapse
Affiliation(s)
- Gelin Xiang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luyin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Jing Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaohui Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, China
| | - Yi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, China
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Wen M, Chen Q, Chen W, Yang J, Zhou X, Zhang C, Wu A, Lai J, Chen J, Mei Q, Yang S, Lan C, Wu J, Huang F, Wang L. A comprehensive review of Rubia cordifolia L.: Traditional uses, phytochemistry, pharmacological activities, and clinical applications. Front Pharmacol 2022; 13:965390. [PMID: 36160419 PMCID: PMC9500525 DOI: 10.3389/fphar.2022.965390] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Rubia cordifolia (family: Rubiaceae) L (R. cordifolia) is a perennial botanical drug climbing vine. As the main part of the traditional Chinese medicine, the rhizome has a long history. A great number of literary studies have reported that it can be used for the improvement of blood circulation, hemostasis, activation of collaterals, etc. When it comes to the wide application of R. cordifolia in traditional medicine, we systematically review its traditional uses, phytochemistry and pharmacological effects. Literatures were systematically searched using several scientific databases, including China National Knowledge Infrastructure (CNKI), Baidu Scholar, PubMed, Web of Science, and other professional websites. Kew Botanical Garden and the iPlant were used for obtaining the scientific names and plant images of R. cordifolia. In addition, other information was also gathered from books including traditional Chinese herbal medicine, the Chinese Pharmacopoeia, and Chinese Materia Medica. So far, many prescriptions containing R. cordifolia have been widely used in the clinical treatment of abnormal uterine bleeding, primary dysmenorrhea and other gynecological diseases, allergic purpura, renal hemorrhage and other diseases. The phytochemistry studies have reported that more than 100 compounds are found in R. cordifolia, such as bicyclic peptides, terpenes, polysaccharides, trace elements, flavonoids, and quinones. Among them, quinones and peptides are the types of components with the highest contents in R. cordifolia. The modern pharmacological studies have revealed that R. cordifolia and its derived components have anti-tumor, anti-oxidative, anti-platelet aggregation, and anti-inflammatory effects. However, most studies are preclinical. The pharmacological mechanism of R. cordifolia has not been thoroughly studied. In addition, there are few pharmacokinetic and toxicity studies of R. cordifolia, therefore the clinical safety data for R. cordifolia is lacking. To sum up, this review for the first time summarizes a systemic and integrated traditional uses, chemical compositions, pharmacological actions and clinical applications of R. cordifolia, which provides the novel and full-scale insight for the drug development, medicinal value, and application of R. cordifolia in the future.
Collapse
Affiliation(s)
- Min Wen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qi Chen
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wang Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaogang Zhou
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chunxiang Zhang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jia Lai
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianping Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Qibing Mei
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shuo Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Cai Lan
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
3
|
Therapeutic potential of biogenic and optimized silver nanoparticles using Rubia cordifolia L. leaf extract. Sci Rep 2022; 12:8831. [PMID: 35614187 PMCID: PMC9133087 DOI: 10.1038/s41598-022-12878-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/12/2022] [Indexed: 12/24/2022] Open
Abstract
Rubia cordifolia L. is a widely used traditional medicine in the Indian sub-continent and Eastern Asia. In the present study, the aqueous leaf extract of the R. Cordifolia was used to fabricate silver nanoparticles (RC@AgNPs), following a green synthesis approach. Effect of temperature (60 °C), pH (8), as well the concentration of leaf extract (2 ml) and silver nitrate (2 mM) were optimized for the synthesis of stable RC@AgNPs. The phytofabrication of nanosilver was validated by UV–visible spectral analysis, which displayed a distinctive surface plasmon resonance peak at 432 nm. The effective functional molecules as capping and stabilizing agents, and responsible for the conversion of Ag+ to nanosilver (Ag0) were identified using the FTIR spectra. The spherical RC@AgNPs with an average size of ~ 20.98 nm, crystalline nature, and 61% elemental composition were revealed by TEM, SEM, XRD, and. EDX. Biogenic RC@AgNPs displayed a remarkable anticancer activity against B16F10 (melanoma) and A431 (carcinoma) cell lines with respective IC50 of 36.63 and 54.09 µg/mL, respectively. Besides, RC@AgNPs showed strong antifungal activity against aflatoxigenic Aspergillus flavus, DNA-binding properties, and DPPH and ABTS free radical inhibition. The presented research provides a potential therapeutic agent to be utilized in various biomedical applications.
Collapse
|
4
|
Kagisha V, Djang'eing'a RM, Muganga R, Bonnet O, Tchinda AT, Jansen O, Tomani JC, Njakarinala R, Ledoux A, Nyirimigabo A, Frederich M. Pentas longiflora Oliv. (Rubiaceae), a plant used in the treatment of Pityriasis Versicolor in Rwanda: Chemical composition and standardization of leaves and roots. Fitoterapia 2021; 153:104974. [PMID: 34166706 DOI: 10.1016/j.fitote.2021.104974] [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: 04/21/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 11/24/2022]
Abstract
In Rwanda, the roots of Pentas longiflora Oliv. (Rubiaceae) have been used for a long time to treat Pityriasis versicolor. However, many people reported the use of leaves instead of roots. This research was conducted to compare the phytochemical composition and establish chromatographic methods for the standardization of roots and leaves extracts of P. longiflora. During this process, three new pentalongin glycosides (pentalonginoside A, pentalonginoside B, and pentalonginoside C) and two known glycosides of the same type (harounoside and clarinoside), as well as rutin, luteolin-7-rutinoside were isolated from methanol extract of leaves. In addition, pentalongin and psychorubrin, previously isolated from ethylacetate roots extract, were also identified in Pentas longiflora ethylacetate leaves extract. The presence of the antifungal compound pentalongin in leaves may explain the traditional use of leaves in the treatment of Pytiriasis versicolor. Furthermore, harounoside, psychorubrin, and pentalongin were selected as markers for HPLC fingerprints of MeOH extract. The accuracy and risk profile demonstrated the reliability of the validated method. In general, considerable variations of concentration in plant metabolites, including pentalongin, were observed between samples from different sites. The content in pentalongin (expressed as juglone) in collected samples ranged between 1.7 and 70.0 mg/100 g. The highest concentration (70.0 ± 17 mg/100 g) was registered in the cultivated samples from Mukoni. This important variation of pentalongin concentrations according to sampling sites, shows that in order to guarantee equivalent efficacy, finished products with P. longiflora should be standardized based on their pentalongin content.
Collapse
Affiliation(s)
- Vedaste Kagisha
- Department of Pharmacy, School of Medicine and Pharmacy, Huye Biotechnology Laboratory Complex, University of Rwanda, Gikondo, KK 737 Street, P.O. Box 4285, Kigali, Rwanda; Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium.
| | - Roland Marini Djang'eing'a
- University of Liege, Laboratory of Analytical Pharmaceutical Chemistry, Center for Interdisciplinary Research on Medicines, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| | - Raymond Muganga
- Department of Pharmacy, School of Medicine and Pharmacy, Huye Biotechnology Laboratory Complex, University of Rwanda, Gikondo, KK 737 Street, P.O. Box 4285, Kigali, Rwanda
| | - Olivier Bonnet
- Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| | - Alembert Tiabou Tchinda
- Laboratory of Phytochemistry, Institute of Medical Research and Medicinal Plants Studies (IMPM), PO Box 13033, Yaoundé, Cameroon
| | - Olivia Jansen
- Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| | - Jean Claude Tomani
- Department of Chemistry, College of Science and technology, Huye Biotechnology Laboratory Complex, University of Rwanda, Gikondo, KK 737 Street, P.O. Box 4285, Kigali, Rwanda
| | - Ranarivelo Njakarinala
- Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium; Centre National d'Application de Recherches Pharmaceutiques (CNARP), Ambodivoanjo - Ambohijatovo, BP 702, 101, Antananarivo, Madagascar
| | - Allison Ledoux
- Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| | - Alain Nyirimigabo
- Department of Pharmacy, School of Medicine and Pharmacy, Huye Biotechnology Laboratory Complex, University of Rwanda, Gikondo, KK 737 Street, P.O. Box 4285, Kigali, Rwanda; Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| | - Michel Frederich
- Laboratory of Pharmacognosy, Center for Interdisciplinary Research on Medicines, University of Liege, Avenue Hippocrate 15, B36, B4000 Liège, Belgium
| |
Collapse
|
5
|
Singh J, Hussain Y, Luqman S, Meena A. Purpurin: A natural anthraquinone with multifaceted pharmacological activities. Phytother Res 2021; 35:2418-2428. [PMID: 33254282 DOI: 10.1002/ptr.6965] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/09/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022]
Abstract
Purpurin is a naturally occurring anthraquinone isolated from the roots of Rubia cordifolia. Historically, it has been used as a red dye. However, its photosensitizing property and biological effects have deciphered its novel application. Purpurin shows antigenotoxic, anticancer, neuromodulatory, and antimicrobial potential associated with antioxidant action in in vivo and in vitro experiments. A robust antioxidant nature of purpurin is responsible for the majority of its pharmacological effects. It produces anti-inflammatory activity by reducing oxidative stress, which is a fundamental property to target diseases involving endoplasmic reticulum and mitochondrial stress. It can cross the blood-brain barrier and produce neuroprotective effects, including antidepressant and anti-Alzheimer action. It shows antimutagenic property via inhibiting essential CYP-450 enzymes. Interestingly, it gets photosensitized by UV-light and produces target-specific ROS-dependent apoptosis in cancer cells. Therefore, it owns cell killing and cell survival potential subject to the influence of external conditions. Hitherto, limited research studies are performed with purpurin to understand its therapeutic potential. Hence, this review describes and discusses different in vivo, in vitro, and in silico studies performed using purpurin. It also covers physicochemical, pharmacokinetics, and toxicology aspects of purpurin. Moreover, in the end, the prospect of purpurin in the management of cancer has also been proposed.
Collapse
Affiliation(s)
- Jyoti Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Jawaharlal Nehru University, New Delhi, India
| | - Yusuf Hussain
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, Uttar Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
6
|
Wang D, Wang XH, Yu X, Cao F, Cai X, Chen P, Li M, Feng Y, Li H, Wang X. Pharmacokinetics of Anthraquinones from Medicinal Plants. Front Pharmacol 2021; 12:638993. [PMID: 33935728 PMCID: PMC8082241 DOI: 10.3389/fphar.2021.638993] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/03/2021] [Indexed: 12/23/2022] Open
Abstract
Anthraquinones are bioactive natural products, some of which are active components in medicinal medicines, especially Chinese medicines. These compounds exert actions including purgation, anti-inflammation, immunoregulation, antihyperlipidemia, and anticancer effects. This study aimed to review the pharmacokinetics (PKs) of anthraquinones, which are importantly associated with their pharmacological and toxicological effects. Anthraquinones are absorbed mainly in intestines. The absorption rates of free anthraquinones are faster than those of their conjugated glycosides because of the higher liposolubility. A fluctuation in blood concentration and two absorption peaks of anthraquinones may result from the hepato-intestinal circulation, reabsorption, and transformation. Anthraquinones are widely distributed throughout the body, mainly in blood-flow rich organs and tissues, such as blood, intestines, stomach, liver, lung, kidney, and fat. The metabolic pathways of anthraquinones are hydrolysis, glycuronidation, sulfation, methylation/demethylation, hydroxylation/dehydroxylation, oxidation/reduction (hydrogenation), acetylation and esterification by intestinal flora and liver metabolic enzymes, among which hydrolysis, glycuronidation and sulfation are dominant. Of note, anthraquinones can be transformed into each other. The main excretion routes for anthraquinones are the kidney, recta, and gallbladder. Conclusion: Some anthraquinones and their glycosides, such as aloe-emodin, chrysophanol, emodin, physcion, rhein and sennosides, have attracted the most PK research interest due to their more biological activities and/or detectability. Anthraquinones are mainly absorbed in the intestines and are mostly distributed in blood flow-rich tissues and organs. Transformation into another anthraquinone may increase the blood concentration of the latter, leading to an increased pharmacological and/or toxicological effect. Drug-drug interactions influencing PK may provide insights into drug compatibility theory to enhance or reduce pharmacological/toxicological effects in Chinese medicine formulae and deserve deep investigation.
Collapse
Affiliation(s)
- Dongpeng Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Biomedical Research Institute, Hubei Key Laboratory of Wudang Local Chinese Medicine Research and School of Pharmacy, Hubei University of Medicine, Shiyan, China
| | - Xian-He Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiongjie Yu
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Fengjun Cao
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaojun Cai
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Ping Chen
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Minglun Li
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Biomedical Research Institute, Hubei Key Laboratory of Wudang Local Chinese Medicine Research and School of Pharmacy, Hubei University of Medicine, Shiyan, China
| | - Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital, Hubei University of Medicine, Shiyan, China.,Biomedical Research Institute, Hubei Key Laboratory of Wudang Local Chinese Medicine Research and School of Pharmacy, Hubei University of Medicine, Shiyan, China.,Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Balachandran P, Ibrahim MA, Zhang J, Wang M, Pasco DS, Muhammad I. Crosstalk of Cancer Signaling Pathways by Cyclic Hexapeptides and Anthraquinones from Rubia cordifolia. Molecules 2021; 26:molecules26030735. [PMID: 33572569 PMCID: PMC7866972 DOI: 10.3390/molecules26030735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 12/03/2022] Open
Abstract
The anticancer activities of Rubia cordifolia and its constituents have been reported earlier, but their influence on the crosstalk of complex cancer-related signaling metabolic pathways (i.e., transcription factors; TF) has not yet been fully investigated. In this study, R. cordifolia root extract was subjected to the cancer signaling assay based bioactivity-guided fractionation, which yielded the following compounds viz., three anthraquinones, namely alizarin (1), purpurin (2), and emodin (3); two lignans, namely eudesmin (4) and compound 5; and two cyclic hexapeptides, namely deoxybouvardin RA-V (6), and a mixture of 6+9 (RA-XXI). The structures of the isolated compounds were determined by NMR spectroscopy and HRESIMS. The isolated compounds 1, 2, 3, 6, and a mixture of 6+9 were tested against a panel of luciferase reporter genes that assesses the activity of a wide-range of cancer-related signaling pathways. In addition, reference anthraquinones viz., chrysophanol (11), danthron (12), quinizarin (13), aloe-emodin (14), and α-lapachone (15) were also tested. Among the tested compounds, the cyclic hexapeptide 6 was found to be very active against several signaling pathways, notably Wnt, Myc, and Notch with IC50 values of 50, 75, and 93 ng/mL, respectively. Whereas, the anthraquinones exhibited very mild or no inhibition against these signaling pathways. Compound 6 being the most active, we tested it for stability in simulated intestinal (SIF) and gastric fluids (SGF), since the stability in biological fluid is a key short-coming of cyclic hexapeptides. The anticancer activity of 6 was found to remain unchanged before and after the treatment of simulated gastric/intestinal fluids, indicating that RA-V was stable. As a result, it could be bioavailable when orally used in therapeutics and possibly a drug candidate for cancer treatment. The mechanism for the preferential inhibition of these pathways and the possible crosstalk effect with other previously reported signaling pathways has been discussed.
Collapse
Affiliation(s)
- Premalatha Balachandran
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
- Correspondence: (P.B.); (M.A.I.); (I.M.); Tel.: +1-662-915-3463 (P.B.); +1-662-915-1147 (M.A.I.); +1-662-915-1051 (I.M.)
| | - Mohamed Ali Ibrahim
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
- Correspondence: (P.B.); (M.A.I.); (I.M.); Tel.: +1-662-915-3463 (P.B.); +1-662-915-1147 (M.A.I.); +1-662-915-1051 (I.M.)
| | - Jin Zhang
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
| | - Mei Wang
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
- Natural Products Utilization Research Unit, Agricultural Research Service, United States Department of Agriculture, Oxford, MS 38677, USA
| | - David S. Pasco
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
| | - Ilias Muhammad
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (J.Z.); (M.W.); (D.S.P.)
- Correspondence: (P.B.); (M.A.I.); (I.M.); Tel.: +1-662-915-3463 (P.B.); +1-662-915-1147 (M.A.I.); +1-662-915-1051 (I.M.)
| |
Collapse
|
8
|
Chen DB, Gao HW, Peng C, Pei SQ, Dai AR, Yu XT, Zhou P, Wang Y, Cai B. Quinones as preventive agents in Alzheimer's diseases: focus on NLRP3 inflammasomes. J Pharm Pharmacol 2020; 72:1481-1490. [PMID: 32667050 DOI: 10.1111/jphp.13332] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Alzheimer's disease (AD) is a hidden neurological degenerative disease, which main clinical manifestations are cognitive dysfunction, memory impairment and mental disorders. Neuroinflammation is considered as a basic response of the central nervous system. NLRP3 (Nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3) inflammasome is closely related to the occurrence of neuroinflammation. Activation of the NLRP3 inflammasome results in the release of cytokines, pore formation and ultimately pyroptosis, which has demonstrated one of the critical roles in AD pathogenesis. Inhibition of the activity of NLRP3 is one of the focuses of the research. Therefore, NLRP3 represents an attractive pharmacological target, and discovery compounds with good NLRP3 inhibitory activity are particularly important. KEY FINDINGS Quinones have good neuroprotective effects and prevent AD, which may be related to their regulation of inflammatory response. The molecular docking was used to explore 12 quinones with AD prevention and treatment and NLRP3. Docking results showed that the combination of anthraquinones and NLRP3 were the best, and the top two chemical compounds were Purpurin and Rhein, which are the most promising NLRP3 inhibitors. SUMMARY These quinones may provide the theoretical basis for finding lead compounds for novel neuroprotective agents.
Collapse
Affiliation(s)
- Da-Bao Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hua-Wu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Cheng Peng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Shao-Qiang Pei
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - An-Ran Dai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xue-Ting Yu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
9
|
Haider K, Haider MR, Neha K, Yar MS. Free radical scavengers: An overview on heterocyclic advances and medicinal prospects. Eur J Med Chem 2020; 204:112607. [PMID: 32721784 DOI: 10.1016/j.ejmech.2020.112607] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/08/2020] [Accepted: 06/19/2020] [Indexed: 12/22/2022]
Abstract
In the present scenario, there has been a lot of consideration toward the field of free radical chemistry. Free radicals responsive oxygen species are produced by different endogenous frameworks, exposure to various physicochemical conditions, radiation, toxins, metabolized drug by-product, and pathological states. On the off chance that free radical overpowers the body's capacity, it generates a condition known as oxidative stress, which can alter physiological conditions of the body and results in several diseases. For appropriate physiological function, it is necessary to have a proper balance between free radicals and antioxidants. Antioxidants chemically inhibit the oxidation process; they are also known as free radical scavengers. For tackling the problem of oxidative stress application of an external source of antioxidant is helpful. A lot of antioxidants of natural, semi-synthetic and synthetic origin are in use, with time search of more effective, nontoxic, safe antioxidant is intensified. The present review, discuss different synthetic derivatives bearing various heterocyclic scaffolds as radical scavengers.
Collapse
Affiliation(s)
- Kashif Haider
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Md Rafi Haider
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kumari Neha
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - M Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
10
|
Chen Y, Chen PD, Bao BH, Shan MQ, Zhang KC, Cheng FF, Cao YD, Zhang L, Ding AW. Anti-thrombotic and pro-angiogenic effects of Rubia cordifolia extract in zebrafish. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:152-160. [PMID: 29126989 DOI: 10.1016/j.jep.2017.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 11/02/2017] [Accepted: 11/05/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rubia cordifolia is a common traditional Chinese medicine that promotes blood circulation and eliminates blood stasis, and has been used to cure diseases related to blood stasis syndrome (BSS) clinically for many years. It has been previously demonstrated that anti-thrombosis and pro-angiogenesis can improve BSS. However, the anti-thrombotic and pro-angiogenic activities of Rubia cordifolia have not been well investigated. AIM OF STUDY To determine the potential anti-thrombotic and pro-angiogenic activities of Rubia cordifolia and to elucidate the underlying mechanisms. In addition, the major chemical constituents of Rubia cordifolia extract (QC) were qualitatively analysed by UPLC-Q-TOF/MS to explore the association between pharmacological activity and chemical constituents. MATERIAL AND METHODS The QC samples were composed of a 95% ethanol extract and an aqueous extract following extraction using 95% ethanol. UPLC-Q-TOF/MS was used to analyse the major chemical constituents of QC. For the anti-thrombotic experiment of QC, a phenylhydrazine (PHZ)-induced AB strain zebrafish thrombosis model was used. The zebrafish larvae were stained using O-dianisidine, and the heart and caudal vein of the zebrafish were observed and imaged with a fluorescence microscope. The staining intensity of erythrocytes in the heart (SI) of each group and the morphology of thrombus in the caudal vein were used to assess the anti-thrombotic effect of QC. For the pro-angiogenic assay of QC, the intersegmental blood vessel (ISV) insufficiency model of Tg(fli-1: EGFP)y1 transgenic zebrafish (Flik zebrafish), which was induced by the VEGF receptor tyrosine kinase inhibitor II (VRI), was used. The morphology of the intact ISVs and defective ISVs was observed to evaluate the pro-angiogenic activity of QC. The mechanism involved in promoting angiogenesis was studied with real-time PCR. RESULTS A total of 12 components in QC were identified based on standard compounds and references, including nine anthraquinones and three naphthoquinones. After treatment with QC, the PHZ-induced thrombosis in AB strain zebrafish larvae decreased to a certain degree, which we believe was related to its dosages, and the therapeutic effect within the 50-200 µg/mL QC treatment groups was especially prominent (P < 0.01, P < 0.001) compared to that in the PHZ model group. Similarly, QC also recovered the loss of the ISVs, which was induced by VRI in Flik zebrafish larvae, which have a certain dose-effect relationship. The pro-angiogenic activity of QC was also conspicuous (P < 0.01, P < 0.001) compared to that of the VRI model group. The following real-time PCR assay proved that QC significantly restored the VRI-induced downregulation of vWF, VEGF-A, kdrl, and flt-1 in Flik zebrafish (P < 0.05, P < 0.01, P < 0.001). CONCLUSIONS A total of 12 compounds from QC were analysed by UPLC-Q-TOF/MS. The data of the pharmacological experiments demonstrated that QC presented anti-thrombotic and pro-angiogenic activities in zebrafish, and the principal active components were likely anthraquinones and naphthoquinones. Thus, the current study provided a theoretical basis for the clinical use of Rubia cordifolia as a traditional Chinese medicine in promoting blood circulation and eliminating stasis.
Collapse
Affiliation(s)
- Yi Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Pei-Dong Chen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Bei-Hua Bao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Ming-Qiu Shan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Kai-Cheng Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Fang-Fang Cheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Yu-Dan Cao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China
| | - An-Wei Ding
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138#, Nanjing 210023, China.
| |
Collapse
|
11
|
Gao M, Wang Z, Yang J, Wang L, Wu C, Cui B, Yang C. Simultaneous determination and pharmacokinetics study of four quinones in rat plasma by ultra high performance liquid chromatography with electrospray ionization tandem mass spectrometry after the oral administration of Qianzhi capsules. J Sep Sci 2018; 41:2161-2168. [DOI: 10.1002/jssc.201700981] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/29/2018] [Accepted: 02/03/2018] [Indexed: 01/24/2023]
Affiliation(s)
- Mingjie Gao
- College of Pharmacy; Harbin Medical University; Harbin P.R. China
| | - Zhibin Wang
- Key Laboratory of Chinese Materia Medical (Ministry of Education); Heilongjiang University of Chinese Medicine; Harbin P.R. China
| | - Jing Yang
- Department of Pathology, School of Basic Medical Science; Heilongjiang University of Chinese Medicine; Harbin P.R. China
| | - Liqian Wang
- College of Pharmacy; Harbin Medical University; Harbin P.R. China
| | - Chengcui Wu
- College of Pharmacy; Harbin Medical University; Harbin P.R. China
| | - Binbin Cui
- College of Pharmacy; Harbin Medical University; Harbin P.R. China
| | - Chunjuan Yang
- College of Pharmacy; Harbin Medical University; Harbin P.R. China
| |
Collapse
|
12
|
Nam W, Kim SP, Nam SH, Friedman M. Structure-Antioxidative and Anti-Inflammatory Activity Relationships of Purpurin and Related Anthraquinones in Chemical and Cell Assays. Molecules 2017; 22:E265. [PMID: 28208613 PMCID: PMC6155578 DOI: 10.3390/molecules22020265] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 01/09/2023] Open
Abstract
Anthraquinone (9,10-anthraquinone) and several hydroxy derivatives, including purpurin (1,2,4-trihydroxyanthraquinone), anthrarufin (1,5-dihydroxyanthraquinone), and chrysazin (1,8-dihydroxyanthraquinone), were evaluated for antioxidative and anti-inflammatory activities in chemical assays and mammalian cells (murine macrophage RAW 264.7 cells). Several tests were used to assess their activities: 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radical; ABTS radical cation; hydrogen peroxide scavenging; reduction of potassium ferricyanide; chelation of ferrous ions; inhibition of lipid peroxidation; inhibition of nitric oxide generation; scavenging of the intracellular hydroxyl radical; expression of NLRP3 polypeptide for inflammasome assembly; and quantitation of proinflammatory cytokine interleukin 1β (IL-1β) for inflammasome activation. The results show that purpurin, from the root of the madder plant (Rubia tinctorum L.), exhibited the highest antioxidative activity in both chemical and cultured cell antioxidant assays. The antioxidative activities of the other three anthraquinones were lower than that of purpurin. In addition, purpurin could down-regulate NLRP3 inflammasome assembly and activation, suggesting that it might protect foods against oxidative damage and prevent in vivo oxidative stress and inflammation. Structure-activity relationships and the significance of the results for food quality and human health are discussed.
Collapse
Affiliation(s)
- Woo Nam
- Department of Biological Science, Ajou University, Suwon 16499, Korea.
| | - Sung Phil Kim
- Research Institute of Basic Sciences, Ajou University, Suwon 16499, Korea.
- STR Biotech. Ltd., Chuncheon 24232, Korea.
| | - Seok Hyun Nam
- Department of Biological Science, Ajou University, Suwon 16499, Korea.
| | - Mendel Friedman
- Western Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA 94710, USA.
| |
Collapse
|