1
|
Misra A, Chaudhary MK, Rawat P, Tripathi D, Barik SK, Srivastava S. Benzyl-isoquinoline alkaloids rich extract of Coptis teeta Wall., exhibit potential efficacy in calcium-oxalate and uric-acid linked metabolic disorders. Fitoterapia 2024; 177:106050. [PMID: 38838823 DOI: 10.1016/j.fitote.2024.106050] [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: 01/31/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Coptis teeta Wall., an endangered but valuable medicinal species having various folklore uses in Indian and Chinese Traditional system of medicine. Its distribution is restricted to India, China and Tibet. In India, C. teeta is traditionally used in joint disorders, urinary infections and inflammatory diseases, however the scientific validation is missing. Thus, the present study aims to validate the anti-lithiatic and anti-gout activity of C. teeta rhizome extract (CTME) through in-vitro biological assays. The metabolic fingerprinting of CTME through reverse phase-high performance liquid chromatography-photodiode array (RP-HPLC-PDA) showed the presence of five benzyl-isoquinoline alkaloids, namely berberine (2.59%), coptisine (0.746%) jatrorrhizine (0.133%), palmatine (0.03%) and tetrahydropalmatine (0.003%). The anti-gout potency analysed via in-vitro xanthine oxidase (XOD) inhibition assay, followed by HPTLC (High performance thin layer chromatography) mediated bio-autographic inhibition of XOD signifies that CTME exhibit strong inhibition of XOD (IC50: 3.014 μg/ml), insignificantly different (p > 0.05) from allopurinol (IC50: 2.47 μg/ml). The XOD bioautographic assay advocates that the efficacy is primarily due to berberine and coptisine alkaloids. The CTME has significant anti-lithiatic activity, and thereby limiting the progression of crystal nidus formation, mediated via inhibition of calcium oxalate crystals nucleation and aggregation. Additionally, the extract also exhibits potential effect on inhibition of oxidative stress associated inflammation, which plays crucial role in alleviating urolithiasis and gouty conditions. Validating the traditional claims of C. teeta will not only confirm its medicinal benefits for targeted pathological conditions but also enhance its industrial demand.
Collapse
Affiliation(s)
- Ankita Misra
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, UP, India
| | - Mridul Kant Chaudhary
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, UP, India
| | - Poonam Rawat
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, UP, India
| | - Deepali Tripathi
- FEST Division, CSIR-Indian Institute of Toxicological Research, Lucknow, UP, India
| | - Saroj Kanta Barik
- Department of Botany, North-Eastern Hill University, Shillong, India
| | - Sharad Srivastava
- Pharmacognosy Division, CSIR-National Botanical Research Institute, Lucknow, UP, India.
| |
Collapse
|
2
|
Sun Q, Liang J, Zhang Q, Wang X, Zhao N, Meng F. Pharmacokinetics and Tissue Distribution of Itampolin A following Intragastric and Intravenous Administration in Rats Using Ultra-High-Performance Liquid Chromatography-Tandem Mass Spectrometry. Molecules 2024; 29:2652. [PMID: 38893526 PMCID: PMC11173508 DOI: 10.3390/molecules29112652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Itampolin A, a natural brominated tyrosine alkaloid isolated from the sponge Iotrochota purpurea, has been shown to have good inhibitory effects in lung cancer cells as a p38α inhibitor. A simple, sensitive, and reliable ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method has been established, validated, and applied to the study of the pharmacokinetics and tissue distribution of itampolin A following intragastric and intravenous administration. Itampolin A and theophylline (internal standard, IS) were extracted by the simple protein precipitation technique using methanol as the precipitating solvent. Chromatographic separation was achieved by using the optimized mobile phase of a 0.1% formic acid aqueous solution and acetonitrile in the gradient elution mode. Itampolin A and IS were detected and quantified using positive electrospray ionization in the multiple reaction monitoring mode with transitions of m/z 863.9 → 569.1 for itampolin A and m/z 181.1 → 124.1 for IS, respectively. The assay exhibited a linear dynamic range of 1-1600 ng/mL for itampolin A in biological samples and the low limit of quantification was 1 ng/mL. Non-compartmental pharmacokinetic parameters indicated that itampolin A was well-absorbed into the systemic circulation and rapidly eliminated after administration. The apparent distribution volume of itampolin A was much higher after intragastric administration than that after intravenous administration. A tissue distribution study showed that itampolin A could be detected in different tissues and maintained a high concentration in the lung, which provided a material basis for its effective application in lung cancer. The pharmacokinetic process and tissue distribution characteristics of imtapolin A were expounded in this study, which can provide beneficial information for the further research and clinical application of itampolin A.
Collapse
Affiliation(s)
- Qi Sun
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Q.S.); (Q.Z.); (X.W.)
| | - Jingwei Liang
- School of Pharmacy, Hainan Medical University, Haikou 570100, China;
| | - Qingyu Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Q.S.); (Q.Z.); (X.W.)
| | - Xuezhen Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Q.S.); (Q.Z.); (X.W.)
| | - Nan Zhao
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Q.S.); (Q.Z.); (X.W.)
| | - Fanhao Meng
- School of Pharmacy, China Medical University, Shenyang 110122, China; (Q.S.); (Q.Z.); (X.W.)
| |
Collapse
|
3
|
Zhang S, Zhang Y, Wen Z, Chen Y, Bu T, Yang Y, Ni Q. Enhancing β-cell function and identity in type 2 diabetes: The protective role of Coptis deltoidea C. Y. Cheng et Hsiao via glucose metabolism modulation and AMPK signaling activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155396. [PMID: 38547617 DOI: 10.1016/j.phymed.2024.155396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Abnormalities in glucose metabolism may be the underlying cause of β-cell dysfunction and identity impairment resulting from high glucose exposure. In China, Coptis deltoidea C. Y. Cheng et Hsiao (YL) has demonstrated remarkable hypoglycemic effects. HYPOTHESIS/PURPOSE To investigate the hypoglycemic effect of YL and determine the mechanism of YL in treating diabetes. METHODS A type 2 diabetes mouse model was used to investigate the pharmacodynamics of YL. YL was administrated once daily for 8 weeks. The hypoglycemic effect of YL was assessed by fasting blood glucose, an oral glucose tolerance test, insulin levels, and other indexes. The underlying mechanism of YL was examined by targeting glucose metabolomics, western blotting, and qRT-PCR. Subsequently, the binding capacity between predicted AMP-activated protein kinase (AMPK) and important components of YL (Cop, Ber, and Epi) were validated by molecular docking and surface plasmon resonance. Then, in AMPK knockdown MIN6 cells, the mechanisms of Cop, Ber, and Epi were inversely confirmed through evaluations encompassing glucose-stimulated insulin secretion, markers indicative of β-cell identity, and the examination of glycolytic genes and products. RESULTS YL (0.9 g/kg) treatment exerted notable hypoglycemic effects and protected the structural integrity and identity of pancreatic β-cells. Metabolomic analysis revealed that YL inhibited the hyperactivated glycolysis pathway in diabetic mice, thereby regulating the products of the tricarboxylic acid cycle. KEGG enrichment revealed the intimate relationship of this process with the AMPK signaling pathway. Cop, Ber, and Epi in YL displayed high binding affinities for AMPK protein. These compounds played a pivotal role in preserving the identity of pancreatic β-cells and amplifying insulin secretion. The mechanism underlying this process involved inhibition of glucose uptake, lowering intracellular lactate levels, and elevating acetyl coenzyme A and ATP levels through AMPK signaling. The use of a glycolytic inhibitor corroborated that attenuation of glycolysis restored β-cell identity and function. CONCLUSION YL demonstrates significant hypoglycemic efficacy. We elucidated the potential mechanisms underlying the protective effects of YL and its active constituents on β-cell function and identity by observing glucose metabolism processes in pancreatic tissue and cells. In this intricate process, AMPK plays a pivotal regulatory role.
Collapse
Affiliation(s)
- Shan Zhang
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yueying Zhang
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhige Wen
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yupeng Chen
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Tianjie Bu
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanan Yang
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qing Ni
- Department of Endocrinology, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
4
|
Lu H, Huang Y, Ni X, Ma T, Chang T, Liu M, Li N, Lu P, Yuan P, Liu L, Shi F, Xiao J, Xiao H, Duan Q, Zhu F. TOPK promotes the development of psoriasis and worenine alleviates psoriasiform dermatitis by inhibiting TOPK activity. J Eur Acad Dermatol Venereol 2024; 38:851-863. [PMID: 38131517 DOI: 10.1111/jdv.19724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/24/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Psoriasis is an inflammatory skin disease. The pathogenesis of psoriasis has not been fully elucidated. T-lymphokine-activated killer cell-originated protein kinase (TOPK) activity increases in a proinflammatory environment, and inhibiting TOPK blocks inflammation. However, whether TOPK is involved in the pathogenesis of psoriasis remains to be identified. OBJECTIVES We aimed to study the role of TOPK in psoriasis and attempted to find a drug targeting TOPK for the prevention and treatment of psoriasis. METHOD Firstly, the expressions of TOPK in psoriatic patients, psoriatic cell and animal model were analysed by Gene Expression Omnibus database, immunohistochemistry (IHC) staining and western blot (WB). After inhibiting TOPK by chemical or gene knockout, the effect of TOPK on the development of psoriasis was verified in cell and animal model by WB, qRT-PCR, ELISA, haematoxylin-eosin (H&E) and IHC staining. Moreover, phosphoproteomic analysis was performed to explore the signalling pathways regulated by TOPK in the occurrence and development of psoriasis. Then, an in vitro kinase assay was performed to prove TOPK kinase activity was inhibited by worenine. Ultimately, WB, qRT-PCR, ELISA, H&E and IHC staining were used to verify the anti-psoriasis effect of worenine by inhibiting TOPK was in cell and animal model. RESULTS In this study, we found that TOPK was highly expressed in psoriasis patients, psoriatic cell and animal model, which suggests that TOPK might be associated with psoriasis pathogenesis. Interestingly, chemical or genetic inhibition of TOPK alleviated M5- and imiquimod (IMQ)-induced psoriasis-like dermatitis, which further confirmed the role of TOPK in promoting the development of psoriasis. Moreover, we determined that worenine inhibited TOPK kinase activity. In addition, worenine relieved M5- and IMQ-induced psoriasiform dermatitis by inhibiting TOPK activity. CONCLUSIONS T-lymphokine-activated killer cell-originated protein kinase promotes the development of psoriasis. Therefore, TOPK might be a promising drug target for the prevention and treatment of psoriasis. Worenine alleviates psoriasiform dermatitis by inhibiting TOPK activity, providing new strategies for clinical intervention.
Collapse
Affiliation(s)
- Hui Lu
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, China
| | - Yingze Huang
- Cancer Research Institute, the Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Xiaofang Ni
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tengfei Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Teding Chang
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Man Liu
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nijie Li
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peijiang Lu
- Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ping Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fei Shi
- Department of Dermatology, the General Hospital of Air Force, Beijing, Beijing, China
| | - Juanjuan Xiao
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, China
- Cancer Research Institute, the Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Han Xiao
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuhong Duan
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feng Zhu
- Translational Medical Center, Huaihe Hospital, Henan University, Kaifeng, Henan, China
- Cancer Research Institute, the Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
5
|
Yang C, Zhao Y, Jiang S, Sun X, Wang X, Wang Z, Wu Y, Wu J, Li Y. A breakthrough in phytochemical profiling: ultra-sensitive surface-enhanced Raman spectroscopy platform for detecting bioactive components in medicinal and edible plants. Mikrochim Acta 2024; 191:286. [PMID: 38652378 DOI: 10.1007/s00604-024-06360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
A perennial challenge in harnessing the rich biological activity of medicinal and edible plants is the accurate identification and sensitive detection of their active compounds. In this study, an innovative, ultra-sensitive detection platform for plant chemical profiling is created using surface-enhanced Raman spectroscopy (SERS) technology. The platform uses silver nanoparticles as the enhancing substrate, excess sodium borohydride prevents substrate oxidation, and methanol enables the tested molecules to be better adsorbed onto the silver nanoparticles. Subsequently, nanoparticle aggregation to form stable "hot spots" is induced by Ca2+, and the Raman signal of the target molecule is strongly enhanced. At the same time, deuterated methanol was used as the internal standard for quantitative determination. The method has excellent reproducibility, RSD ≤ 1.79%, and the enhancement factor of this method for the detection of active ingredients in the medicinal plant Coptis chinensis was 1.24 × 109, with detection limits as low as 3 fM. The platform successfully compared the alkaloid distribution in different parts of Coptis chinensis: root > leaf > stem, and the difference in content between different batches of Coptis chinensis decoction was successfully evaluated. The analytical technology adopted by the platform can speed up the determination of Coptis chinensis and reduce the cost of analysis, not only making better use of these valuable resources but also promoting development and innovation in the food and pharmaceutical industries. This study provides a new method for the development, evaluation, and comprehensive utilization of both medicinal and edible plants. It is expected that this method will be extended to the modern rapid detection of other medicinal and edible plants and will provide technical support for the vigorous development of the medicinal and edible plants industry.
Collapse
Affiliation(s)
- Chunjuan Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yue Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shuang Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaomeng Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaotong Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zhibin Wang
- Key Laboratory of Chinese Materia Medical (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Yanli Wu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Jing Wu
- School of Science, Nantong University, No. 9, Seyuan Road, Nantong, Jiangsu, 226019, China
| | - Yang Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
- Research Center for Innovative Technology of Pharmaceutical Analysis, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
- Research Unit of Health Sciences and Technology (HST), Faculty of Medicine University of Oulu, Oulu, Finland.
| |
Collapse
|
6
|
Xiang ZD, Guan HD, Zhao X, Xie Q, Cai FJ, Xie ZJ, Dang R, Li ML, Wang CH. Protoberberine alkaloids: A review of the gastroprotective effects, pharmacokinetics, and toxicity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155444. [PMID: 38367423 DOI: 10.1016/j.phymed.2024.155444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Stomach diseases have become global health concerns. Protoberberine alkaloids (PBAs) are a group of quaternary isoquinoline alkaloids from abundant natural sources and have been shown to improve gastric disorders in preclinical and clinical studies. The finding that PBAs exhibit low oral bioavailability but potent pharmacological activity has attracted great interest. PURPOSE This review aims to provide a systematic review of the molecular mechanisms of PBAs in the treatment of gastric disorders and to discuss the current understanding of the pharmacokinetics and toxicity of PBAs. METHODS The articles related to PBAs were collected from the Web of Science, Pubmed, and China National Knowledge Infrastructure databases using relevant keywords. The collected articles were screened and categorized according to their research content to focus on the gastroprotective effects, pharmacokinetics, and toxicity of PBAs. RESULTS Based on the results of preclinical studies, PBAs have demonstrated therapeutic effects on chronic atrophic gastritis and gastric cancer by activating interleukin-4 (IL-4)/signal transducer and activator of transcription 6 (STAT6) pathway and suppressing transforming growth factor-beta 1 (TGF-β1)/phosphoinositide 3-kinase (PI3K), Janus kinase-2 (JAK2)/signal transducers and activators of transcription 3 (STAT3), and mitogen-activated protein kinase (MAPK) pathways. The major PBAs exhibit similar pharmacokinetic properties, including rapid absorption, slow elimination, and low bioavailability. Notably, the natural organ-targeting property of PBAs may account for the finding of their low blood levels and high pharmacological activity. PBAs interact with other compounds, including conventional drugs and natural products, by modulation of metabolic enzymes and transporters. The potential tissue toxicity of PBAs should be emphasized due to their high tissue accumulation. CONCLUSION This review highlights the gastroprotective effects, pharmacokinetics, and toxicity of PBAs and will contribute to the evaluation of drug properties and clinical translational studies of PBAs, accelerating their transfer from the laboratory to the bedside.
Collapse
Affiliation(s)
- Ze-Dong Xiang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Hui-Da Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Xiang Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Qi Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Fu-Jie Cai
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Zhe-Jun Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Rui Dang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Man-Lin Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China.
| | - Chang-Hong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Laboratory of Standardization of Chinese Medicines, Shanghai R&D Center for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
7
|
Wang H, Yang D, Jiang S, Ren Y, Wu L, Wang Z, Kuang H, Wang Z. Simultaneous determination of four phytoecdysteroids by LC-MS/MS: application to a comparative pharmacokinetic study in normal and adjuvant arthritis rats after oral administration of C. officinalis Kuan phytoecdysteroids extract. Xenobiotica 2023; 53:634-643. [PMID: 38053346 DOI: 10.1080/00498254.2023.2270741] [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: 08/04/2023] [Accepted: 10/10/2023] [Indexed: 12/07/2023]
Abstract
C. officinalis Kuan is the dry root of Cyathula officinalis Kuan. Clinically, it is used for fall and flutter injury, rheumatism and arthralgia. Phytoecdysteroids have significant anti-inflammatory effects, and the phytoecdysteroids present in C. officinalis Kuan exhibit potential for treating rheumatoid arthritis.This study first developed a selective, accurate and efficient LC-MS/MS method for 12-day pharmacokinetic studies regarding the simultaneous determination of cyasterone, 25-epi-28-epi-cyasterone, precyasterone and capitasterone from C. officinalis Kuan phytoecdysteroids extract in normal and adjuvant arthritis rats.An Agilent Eclipse Plus C18 RRHD column (1.8 µm, 50mm × 2.1 mm) with a gradient mobile phase consisting of water (A) and acetonitrile (B) was used for analysis. The mass analysis was performed in an Agilent 6430 QQQ-MS mass spectrometer with positive mode multiple reaction monitoring (MRM).The results indicated that the AUC0-t and AUC0-∞ values of the four phytoecdysteroids in adjuvant arthritis rats were different from those in normal rats on the first day, which could provide a helpful reference for pharmacological and toxicological studies, as well as clinical applications of C. officinalis Kuan in the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Haiqiang Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Deqiang Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shuang Jiang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yixuan Ren
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lihong Wu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhenyue Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhibin Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese medicine), Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
8
|
Chen AQ, Wu HL, Sun HB, Wang XZ, Yan XQ, Wang T, Yu RQ. Chemometrics-enhanced high-performance liquid chromatography-diode array detection strategy to quantify protoberberine alkaloids in varying Coptidis Rhizoma-related medicines. J Chromatogr A 2022; 1681:463426. [DOI: 10.1016/j.chroma.2022.463426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/06/2022] [Accepted: 08/16/2022] [Indexed: 10/15/2022]
|
9
|
Feng X, Wang K, Cao S, Ding L, Qiu F. Pharmacokinetics of Five Alkaloids and their Metabolites in Normal and Diabetic Rats after Oral Administration of Rhizoma coptidis. PLANTA MEDICA 2022; 88:921-932. [PMID: 34111890 DOI: 10.1055/a-1506-1627] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Rhizoma coptidis has been clinically used for a long time for the treatment of various diseases in China, such as hypertension, diabetes, and inflammation. Previous studies have shown that alkaloid components of Rhizoma coptidis extract could be extensively metabolized and the metabolites were also considered to be the therapeutic material basis. However, until now, pharmacokinetic studies of the in vivo metabolites have not been revealed yet. The aim of the present study was to characterize the pharmacokinetics and excretions of five main alkaloids (berberine, jatrorrhizine, palmatine, epiberberine, and coptisine) and their seven metabolites (berberrubine, demethyleneberberine, jatrorrhizine-3-O-β-D-glucuronide, thalifendine-10-O-β-D-glucuronide, berberrubine-9-O-β-D-glucuronide, demethyleneberberine-2-O-sulfate, and demethyleneberberine-2-O-β-D-glucuronide) in rats after oral administration of Rhizoma coptidis extract. Meanwhile, comparative pharmacokinetics and excretions of these analytes in diabetic model rats were also investigated, since Rhizoma coptidis is widely used for the treatment of diabetes. Our results showed that the in vivo existing forms of alkaloid components were phase II metabolites, highlighting the glucuronidation metabolic pathway. In diabetic model rats, the utilization of Rhizoma coptidis alkaloids was significantly increased and the biotransformation of berberine into berberrubine was significantly inhibited.
Collapse
Affiliation(s)
- Xinchi Feng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kun Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Liqin Ding
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
10
|
Jiang S, Yang X, Wang Z, Gan C, Huang J, Sun J, Peng H, Wei F, Wang Z, Yang C. Biotransformation and pharmacokinetic studies of four alkaloids from Uncaria rhynchophylla in rat plasma by ultra-performance liquid chromatography with tandem mass spectrometry. J Pharm Biomed Anal 2022; 218:114858. [DOI: 10.1016/j.jpba.2022.114858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
|
11
|
Liu CS, Hu YN, Luo ZY, Xia T, Chen FL, Tang QF, Tan XM. Comparative pharmacokinetics, intestinal absorption and urinary excretion of six alkaloids from herb pair Phellodendri Chinensis cortex-Atractylodis Rhizoma. Biomed Chromatogr 2021; 36:e5254. [PMID: 34605575 DOI: 10.1002/bmc.5254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/15/2021] [Accepted: 09/26/2021] [Indexed: 11/07/2022]
Abstract
Phellodendri Chinensis Cortex (PCC) and Atractylodis Rhizoma (AR) are frequently used as herb pair to treat eczema and gout owing to their synergistic effects. Alkaloids are the major ingredients from PCC and the effect of their combination on the in vivo processing of alkaloids remains unclear. In this study, a simple and reliable UPLC-MS/MS method for simultaneous determination of six alkaloids in rat plasma was developed. This method was applied to a comparative pharmacokinetic study between PCC and PCC-AR in rats. Effect of AR on absorption of alkaloids was investigated by a single-pass intestinal perfusion study. The effect of AR on urinary excretion of alkaloids was studied. Pharmacokinetic studies showed that the values of rea under the concentration-time curve of phellodendrine, magnoflorine and palmatine were greater in the PCC-AR group than in the PCC group. The intestinal absorptive parameters absorption rate constant and effective permeability of phellodendrine and jatrorrhizine in PCC-AR groups were higher than those in the PCC group. Urinary excretion studies revealed that the excreted amount of alkaloids in the PCC-AR group was lower than that in the PCC group. The results revealed that the combination of PCC and AR improves intestinal absorption of alkaloids and reduces their urinary excretion, which enhances their systemic exposure. This study may explain the synergetic effects of PCC and AR in clinical applications.
Collapse
Affiliation(s)
- Chang-Shun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Yan-Nan Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Ting Xia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Fei-Long Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Qing-Fa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, China
| |
Collapse
|
12
|
Pharmacokinetics, tissue distribution and plasma protein binding rate of palmatine following intragastric and intravenous administration in rats using liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal 2021; 203:114226. [PMID: 34182412 DOI: 10.1016/j.jpba.2021.114226] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 10/21/2022]
Abstract
Palmatine is a natural isoquinoline alkaloid widely found in traditional Chinese medicines. In this study, a simple, sensitive and rapid ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method was developed and validated for the quantification of palmatine in the plasma and tissue samples in rats. Sample preparation involved a simple protein precipitation extraction technique using acetonitrile as the precipitating solvent. Chromatographic separation was accomplished on an ACQUITY UPLC BEH C18 column with a mobile phase of acetonitrile-5 mM ammonium acetate solution (70:30, v/v) at a flow rate of 0.3 mL/min. Coptisine was selected as the internal standard. The protonated analytes were determined with MRM in the positive ion mode. The assay exhibited a linear dynamic range of 1.0-1000 ng/mL for palmatine in each biological matrix and the low limit of quantification was 1.0 ng/mL. Non-compartmental pharmacokinetic parameters indicated that there is a significant difference in the apparent distribution volume and half-life between intragastric and intravenous administration modes. Palmatine could be detected in different tissues and the content in liver and kidney is relatively high, suggesting that liver and kidney might be the targeting organs of palmatine. The plasma protein binding rate test showed that the percent binding of palmatine is medium, and was found to be higher in human than in rats.
Collapse
|
13
|
Xiao Y, Liu Y, Lai Z, Huang J, Li C, Zhang Y, Gong X, Deng J, Ye X, Li X. An integrated network pharmacology and transcriptomic method to explore the mechanism of the total Rhizoma Coptidis alkaloids in improving diabetic nephropathy. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113806. [PMID: 33444721 DOI: 10.1016/j.jep.2021.113806] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhizoma Coptidis (RC) is a traditional Chinese medicine (TCM) used for treating diabetes (Xiao Ke Zheng), which is firstly recorded in Shennong Bencao Jing. Modern pharmacological studies have confirmed that RC has beneficial effects on diabetes and its complications. Alkaloids are the main active pharmacological component of RC. However, the effect and molecular mechanism of total Rhizoma Coptidis alkaloids (TRCA) in improving diabetic nephropathy (DN) are still unclear. AIM OF THE STUDY To verify the effect of TRCA in the treatment of DN and clarify the molecular mechanism by combining network pharmacology and transcriptomic. MATERIALS AND METHODS Eight-week-old db/db mice were orally administered with normal saline, 100 mg/kg TRCA, and 100 mg/kg berberine (BBR) for 8 weeks. Serum, urine, and kidney samples were collected to measure biological indicators and observe renal pathological changes. Then, the molecular mechanism of TRCA improving DN was predicted by the network pharmacology. Briefly, the main active alkaloids components of TRCA and their targets were collected from the database, as well as the potential targets of DN. Using the Cytoscape software to visualize the interactive network diagram of "ingredient-target". The GO and KEGG pathways enrichment analysis of the core targets were executed by Metascape. Furthermore, RNA-seq was used to get whole transcriptomes from the kidneys of db/m mice, db/db mice, and db/db mice treated with TRCA. The key differentially expressed genes (DEGs) were gathered to conduct the GO and KEGG pathways enrichment analysis. Finally, the potential pathways were validated by western blotting. RESULTS The administration of BBR or TRCA for 8 weeks significantly reduced the fasting blood glucose (FBG) and body weight of db/db mice, and improved their renal function and lipid disorders. According to H&E, PAS, and Masson staining, both the BBR and TRCA could alleviate renal damage and fibrosis. The Venn diagram had shown that seven alkaloids ingredients collected from TRCA regulated 85 common targets merged in the TRCA and DN. The results of RNA-seq indicated that there are 121 potential targets for TRCA treatment on DN. Intriguingly, both the AGE-RAGE signaling pathway and the PI3k-Akt signaling pathway were included in the KEGG pathways enrichment results of network pharmacology and RNA-seq. Moreover, we verified that TRCA down-regulated the expression of related proteins in the AGEs-RAGE-TGFβ/Smad2 and PI3K-Akt pathways in the kidney tissues. CONCLUSIONS In summary, the renal protection of TRCA on DN may be related to activation of the AGEs-RAGE-TGFβ/Smad2 and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Yaping Xiao
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Yan Liu
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Zhihui Lai
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Jieyao Huang
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Chunming Li
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Yaru Zhang
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Xiaobao Gong
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Jianling Deng
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China
| | - Xiaoli Ye
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Xuegang Li
- College of Pharmaceutical Sciences, Translational Pharmacy Center of Medical Research Institute, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
14
|
Ai G, Huang Z, Cheng J, Xie J, Zeng H, Liu Y, Li Y, Huang X, Chen J, Su Z. Gut Microbiota-Mediated Transformation of Coptisine Into a Novel Metabolite 8-Oxocoptisine: Insight Into Its Superior Anti-Colitis Effect. Front Pharmacol 2021; 12:639020. [PMID: 33859564 PMCID: PMC8042337 DOI: 10.3389/fphar.2021.639020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/15/2021] [Indexed: 01/22/2023] Open
Abstract
Coptisine (COP) is a bioactive isoquinoline alkaloid derived from Coptis Chinemsis Franch, which is traditionally applied for the management of colitis. However, the blood concentration of COP was extremely low, and its gut microbiota-mediated metabolites were thought to contribute to its prominent bioactivities. To comparatively elucidate the protective effect and underlying mechanism of COP and its novel gut microbiota metabolite (8-oxocoptisine, OCOP) against colitis, we used dextran sulfate sodium (DSS) to induce colitis in mice. Clinical symptoms, microscopic alternation, immune-inflammatory parameters for colitis were estimated. The results indicated that OCOP dramatically ameliorated disease activity index (DAI), the shortening of colon length and colonic histopathological deteriorations. OCOP treatment also suppressed the mRNA expression and release of inflammatory mediators (TGF-β, TNF-α, IL-6, IL-18, IL-1β and IFN-γ) and elevated the transcriptional and translational levels of anti-inflammatory cytokine (IL-10) as well as the mRNA expression levels of adhesion molecules (ICAM-1 and VCAM-1). Besides, the activation of NF-κB pathway and NLRP3 inflammasome was markedly inhibited by OCOP. Furthermore, OCOP displayed superior anti-colitis effect to COP, and was similar to MSZ with much smaller dosage. Taken together, the protective effect of OCOP against DSS-induced colitis might be intimately related to inhibition of NF-κB pathway and NLRP3 inflammasome. And the findings indicated that OCOP might have greater potential than COP to be further exploited as a promising candidate in the treatment of colitis.
Collapse
Affiliation(s)
- Gaoxiang Ai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziwei Huang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juanjuan Cheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huifang Zeng
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Miao L, Yun X, Yang X, Jia S, Jiao C, Shao R, Hao J, Chang Y, Fan G, Zhang J, Geng Q, Wichai N, Gao X. An inhibitory effect of Berberine from herbal Coptis chinensis Franch on rat detrusor contraction in benign prostatic hyperplasia associated with lower urinary tract symptoms. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113666. [PMID: 33301912 DOI: 10.1016/j.jep.2020.113666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coptis chinensis Franch (CCF), also known as Huang Lian in China, is a traditional Chinese medicine that commonly used for more than 2000 years. Clinically, CCF often used as anti-inflammatory, immune regulation and other effects. It has been reported that the decoction containing CCF can be used for the treatment of benign prostatic hyperplasia (BPH) or lower urinary tract symptoms (LUTS). AIM OF THE STUDY This research aims to investigate the effect of CCF on inhibition of BPH development in vivo and in vitro, and further identify the active compound (s) and the possible mechanism involved in BPH-related bladder dysfunction. MATERIALS AND METHODS Oestrodial/testosterone-induced BPH rat model was established as the in vivo model. The prostate index (PI) was calculated, the pathogenesis was analyzed and the micturition parameters were determined in the shamed-operated, BPH model and BPH + CCF groups after 4-week administration. The tension in detrusor strips was then assessed upon KCl or ACh stimulation with or without incubation of CCF or active compounds. To further investigate the signaling involved, rat detrusor cells were cultured as the in vitro models, the instantaneous calcium influx was measured and the ROCK-1 expression was detected. RESULTS Increased PI value and the aggravated prostatic pathology were observed with voiding dysfunction in BPH rats, which were significantly blocked by oral CCF taken. ACh or KCl-induced contractile responses in detrusor strips were significantly inhibited and the micturition parameters were improved when incubation with CCF or its active compounds such as berberine. Both CCF and berberine suppressed the cellular calcium influx and ROCK-1 expression upon ACh stimulation, demonstrating that berberine was one of the active compounds that contributed to CCF-improved micturition symptoms and function. CONCLUSIONS Taken together, our findings give evidence that CCF and its active compound berberine inhibited BPH and bladder dysfunction via Ca2+ and ROCK signaling, supporting their clinical use for BPH and BPH-related LUTS treatment.
Collapse
Affiliation(s)
- Lin Miao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China.
| | - Xiaoting Yun
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaohua Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China
| | - Sitong Jia
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China
| | - Chanyuan Jiao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rui Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China
| | - Jia Hao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yanxu Chang
- Tianjin Key Laboratory of Phytochemistry and Pharmaceutical Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Guanwei Fan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ju Zhang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, And Bioactive Materials Key Lab of Ministry of Education (J.Z.), Nankai University, Tianjin, 300071, China
| | - Qiang Geng
- Department of Andrology, The First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Nuttapong Wichai
- Faculty of Pharmacy, Mahasarakham University, Mahasarakham, 44150, Thailand
| | - Xiumei Gao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin Health Industry Park, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
16
|
Liu CS, Xia T, Luo ZY, Wu YY, Hu YN, Chen FL, Tang QF, Tan XM. Network pharmacology and pharmacokinetics integrated strategy to investigate the pharmacological mechanism of Xianglian pill on ulcerative colitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153458. [PMID: 33486267 DOI: 10.1016/j.phymed.2020.153458] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/20/2020] [Accepted: 12/28/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease with high morbidity, which leads to poor quality of life. The Xianglian pill (XLP) is a classical Chinese patent medicine and has been clinically proven to be an effective treatment for UC. PURPOSE The pharmacological mechanism of the key bioactive ingredients of XLP for the treatment of UC was investigated by a network pharmacology and pharmacokinetics integrated strategy. STUDY DESIGN AND METHODS Network pharmacology was used to analyze the treatment effect of nine quantified XLP ingredients on UC. Key pathways were enriched and analyzed by protein-protein interaction and Kyoto Encyclopedia of Genes and Genomes analyses. The effect of XLP on Th17 cell differentiation was validated using a mouse model of UC. The binding of nine compounds with JAk2, STAT3, HIF-1α, and HSP90AB1 was assessed using molecular docking. A simple and reliable ultra-high-performance liquid chromatography-tandem mass spectrometry method was developed for the simultaneous quantification of nine ingredients from XLP in plasma and applied to a pharmacokinetic study following oral administration. RESULTS Nine compounds of XLP, including coptisine, berberine, magnoflorine,berberrubine, jatrorrhizine, palmatine, evodiamine, rutaecarpine, and dehydrocostus lactone, were detected. Network pharmacology revealed 50 crossover genes between the nine compoundsand UC. XLP treats UC mainly by regulating key pathways of the immune system, including Th17 cell differentiation, Jak-Stat, and PI3K-Akt signaling pathways. An in vivo validation in mice found that XLP inhibits Th17 cell differentiation by suppressing the Jak2-Stat3 pathway, which alleviates mucosal inflammation in UC. Molecular docking confirmed that eight compounds are capable of binding with JAk2, HIF-1α, and HSP90AB1, further confirming the inhibitory effect of XLP on the Jak2-Stat3 pathway. Moreover, apharmacokinetic study revealed that the nine ingredients of XLP are exposed in the plasma and colon tissue, which demonstrates its pharmacological effect on UC. CONCLUSION This study evaluates the clinical treatment efficacy of XLP for UC. The network pharmacology and pharmacokinetics integrated strategy evaluation paradigm is efficient in discovering the key pharmacological mechanism of herbal formulae.
Collapse
Affiliation(s)
- Chang-Shun Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Ting Xia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China
| | - Yuan-Yuan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China
| | - Yan-Nan Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China
| | - Fei-Long Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China
| | - Qing-Fa Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou 510515, PR China.
| |
Collapse
|
17
|
Sun F, Yang X, Ma C, Zhang S, Yu L, Lu H, Yin G, Liang P, Feng Y, Zhang F. The Effects of Diosgenin on Hypolipidemia and Its Underlying Mechanism: A Review. Diabetes Metab Syndr Obes 2021; 14:4015-4030. [PMID: 34552341 PMCID: PMC8450287 DOI: 10.2147/dmso.s326054] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
Hyperlipidemia is a disorder of lipid metabolism, which is a major cause of coronary heart disease. Although there has been considerable progress in hyperlipidemia treatment, morbidity and risk associated with the condition continue to rise. The first-line treatment for hyperlipidemia, statins, has multiple side effects; therefore, development of safe and effective drugs from natural products to prevent and treat hyperlipidemia is necessary. Diosgenin is primarily derived from fenugreek (Trigonella foenum graecum) seeds, and is also abundant in medicinal herbs such as Dioscorea rhizome, Dioscorea septemloba, and Rhizoma polygonati, is a well-known steroidal sapogenin and the active ingredient in many drugs to treat cardiovascular conditions. There is abundant evidence that diosgenin has potential for application in correcting lipid metabolism disorders. In this review, we evaluated the latest evidence related to diosgenin and hyperlipidemia from clinical and animal studies. Additionally, we elaborate the pharmacological mechanism underlying the activity of diosgenin in treating hyperlipidemia in detail, including its role in inhibition of intestinal absorption of lipids, regulation of cholesterol transport, promotion of cholesterol conversion into bile acid and its excretion, inhibition of endogenous lipid biosynthesis, antioxidation and lipoprotein lipase activity, and regulation of transcription factors related to lipid metabolism. This review provides a deep exploration of the pharmacological mechanisms involved in diosgenin-hyperlipidemia interactions and suggests potential routes for the development of novel drug therapies for hyperlipidemia.
Collapse
Affiliation(s)
- Fengcui Sun
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Xiufen Yang
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Chaoqun Ma
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Shizhao Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Lu Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Haifei Lu
- Hubei University of Traditional Chinese Medicine, Wuhan, 430065, People's Republic of China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Pengpeng Liang
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Yanan Feng
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, People’s Republic of China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People’s Republic of China
- Correspondence: Fengxia Zhang Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People’s Republic of ChinaTel +8653168616011 Email
| |
Collapse
|
18
|
Okon E, Kukula-Koch W, Jarzab A, Halasa M, Stepulak A, Wawruszak A. Advances in Chemistry and Bioactivity of Magnoflorine and Magnoflorine-Containing Extracts. Int J Mol Sci 2020; 21:ijms21041330. [PMID: 32079131 PMCID: PMC7072879 DOI: 10.3390/ijms21041330] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 01/09/2023] Open
Abstract
The review collects together some recent information on the identity and pharmacological properties of magnoflorine, a quaternary aporphine alkaloid, that is widely distributed within the representatives of several botanical families like Berberidaceae, Magnoliaceae, Papaveraceae, or Menispermaceae. Several findings published in the scientific publications mention its application in the treatment of a wide spectrum of diseases including inflammatory ones, allergies, hypertension, osteoporosis, bacterial, viral and fungal infections, and some civilization diseases like cancer, obesity, diabetes, dementia, or depression. The pharmacokinetics and perspectives on its introduction to therapeutic strategies will also be discussed.
Collapse
Affiliation(s)
- Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland
- Correspondence: (W.K.-K.); (A.W.); Tel.: +48-81448-6350 (W.K.-K.); +48-81448-7087 (A.W.)
| | - Agata Jarzab
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1 St., 20-093 Lublin, Poland; (E.O.); (A.J.); (M.H.); (A.S.)
- Correspondence: (W.K.-K.); (A.W.); Tel.: +48-81448-6350 (W.K.-K.); +48-81448-7087 (A.W.)
| |
Collapse
|