1
|
Wang F, Wu Q, Zhang Q, Ma S, Wang K, Jian H, Zhang Y. Gegen Qinlian Decoction Combined with Conventional Western Medicine for the Treatment of Infectious Diarrhea: A Systematic Review and Trial Sequential Analysis. Complement Med Res 2024; 31:461-476. [PMID: 39137735 DOI: 10.1159/000540793] [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: 10/30/2023] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Infectious diarrhea (ID) is a highly prevalent disease worldwide that poses a substantial risk to human well-being. In China, numerous clinical studies have investigated the efficacy of Gegen Qinlian decoction (GGQLD) in treating ID. However, there is a need for additional rigorous and evidence-based medical research to enhance physicians' confidence in their prescribing practices. METHODS Seven Chinese and English databases were systematically searched. The Cochrane Risk of Bias tool was used to assess the quality of the included studies. Meta-analysis was conducted using RevMan 5.3, and Stata 16.0 was used for the sensitivity analysis. Trial sequential analysis was performed using TSA v0.9, and GRADEprofiler was utilized to evaluate the quality of evidence. RESULTS A total of 12 randomized controlled trials (RCTs) involving 1,240 patients were included. The meta-analysis demonstrated that the combination of GGQLD with conventional Western medicine had better effects on clinical efficacy (relative risk [RR] = 1.15, 95% confidence interval [CI] [1.10, 1.20]), duration of diarrhea symptoms (weighted mean difference [WMD] = -10.96, 95% CI [-11.97, -9.96]), duration of abdominal pain symptoms (WMD = -12.01, 95% CI [-14.12, -9.90]), duration of fever symptoms (WMD = -11.91, 95% CI [-13.39, -10.43]), interleukin-6 levels (WMD = -113.59, 95% CI [-113.03, -108.14]), and tumor necrosis factor-α levels (WMD = -62.18, 95% CI [-65.25, -59.11]) and that no significant adverse reactions occurred (RR = 0.45, 95% CI [0.10, 1.97]). The sample size of the included studies reached the expected size. The quality of evidence for outcome indicators was rated as low or very low. CONCLUSIONS The combination of GGQLD with conventional Western medicine demonstrates promising efficacy and safety in treating ID. Nonetheless, more high-quality RCTs are required to confirm this conclusion.
Collapse
Affiliation(s)
- Fei Wang
- College of Graduate Studies, Jiangxi University of Chinese Medicine, Nanchang, China
- Academician Workstation, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qianyan Wu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qingyuan Zhang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Shuaishuai Ma
- Academician Workstation, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Kangyi Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Hui Jian
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Ying Zhang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
2
|
Alsharairi NA. The Role of Licorice Chalcones as Molecular Genes and Signaling Pathways Modulator-A Review of Experimental Implications for Nicotine-Induced Non-Small Cell Lung Cancer Treatment. Curr Issues Mol Biol 2024; 46:5894-5908. [PMID: 38921023 PMCID: PMC11202283 DOI: 10.3390/cimb46060352] [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: 05/27/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Lung cancer (LC) represents the leading cause of global cancer deaths, with cigarette smoking being considered a major risk factor. Nicotine is a major hazardous compound in cigarette smoke (CS), which stimulates LC progression and non-small cell lung cancer (NSCLC) specifically through activation of the nicotinic acetylcholine receptor (α7nAChR)-mediated cell-signaling pathways and molecular genes involved in proliferation, angiogenesis, and metastasis. Chalcones (CHs) and their derivatives are intermediate plant metabolites involved in flavonol biosynthesis. Isoliquiritigenin (ILTG), licochalcone A-E (LicoA-E), and echinatin (ECH) are the most common natural CHs isolated from the root of Glycyrrhiza (also known as licorice). In vitro and/or vivo experiments have shown that licorice CHs treatment exhibits a range of pharmacological effects, including antioxidant, anti-inflammatory, and anticancer effects. Despite advances in NSCLC treatment, the mechanisms of licorice CHs in nicotine-induced NSCLC treatment remain unknown. Therefore, the aim of this paper is to review experimental studies through the PubMed/Medline database that reveal the effects of licorice CHs and their potential mechanisms in nicotine-induced NSCLC treatment.
Collapse
Affiliation(s)
- Naser A Alsharairi
- Heart, Mind and Body Research Group, Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
3
|
Chlipała P, Tronina T, Dymarska M, Urbaniak M, Kozłowska E, Stępień Ł, Kostrzewa-Susłow E, Janeczko T. Multienzymatic biotransformation of flavokawain B by entomopathogenic filamentous fungi: structural modifications and pharmacological predictions. Microb Cell Fact 2024; 23:65. [PMID: 38402203 PMCID: PMC10893614 DOI: 10.1186/s12934-024-02338-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/16/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Flavokawain B is one of the naturally occurring chalcones in the kava plant (Piper methysticum). It exhibits anticancer, anti-inflammatory and antimalarial properties. Due to its therapeutic potential, flavokawain B holds promise for the treatment of many diseases. However, due to its poor bioavailability and low aqueous solubility, its application remains limited. The attachment of a sugar unit impacts the stability and solubility of flavonoids and often determines their bioavailability and bioactivity. Biotransformation is an environmentally friendly way to improve the properties of compounds, for example, to increase their hydrophilicity and thus affect their bioavailability. Recent studies proved that entomopathogenic filamentous fungi from the genera Isaria and Beauveria can perform O-methylglycosylation of hydroxyflavonoids or O-demethylation and hydroxylation of selected chalcones and flavones. RESULTS In the present study, we examined the ability of entomopathogenic filamentous fungal strains of Beauveria bassiana, Beauveria caledonica, Isaria farinosa, Isaria fumosorosea, and Isaria tenuipes to transform flavokawain B into its glycosylated derivatives. The main process occurring during the reaction is O-demethylation and/or hydroxylation followed by 4-O-methylglycosylation. The substrate used was characterized by low susceptibility to transformations compared to our previously described transformations of flavones and chalcones in the cultures of the tested strains. However, in the culture of the B. bassiana KCh J1.5 and BBT, Metarhizium robertsii MU4, and I. tenuipes MU35, the expected methylglycosides were obtained with high yields. Cheminformatic analyses indicated altered physicochemical and pharmacokinetic properties in the derivatives compared to flavokawain B. Pharmacological predictions suggested potential anticarcinogenic activity, caspase 3 stimulation, and antileishmanial effects. CONCLUSIONS In summary, the study provided valuable insights into the enzymatic transformations of flavokawain B by entomopathogenic filamentous fungi, elucidating the structural modifications and predicting potential pharmacological activities of the obtained derivatives. The findings contribute to the understanding of the biocatalytic capabilities of these microbial cultures and the potential therapeutic applications of the modified flavokawain B derivatives.
Collapse
Affiliation(s)
- Paweł Chlipała
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland.
| | - Tomasz Tronina
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland
| | - Monika Dymarska
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland
| | - Monika Urbaniak
- Institute of Plant Genetics, Polish Academy of Sciences, Poznań, Strzeszyńska 34, 60-479, Poland
| | - Ewa Kozłowska
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland
| | - Łukasz Stępień
- Institute of Plant Genetics, Polish Academy of Sciences, Poznań, Strzeszyńska 34, 60-479, Poland
| | - Edyta Kostrzewa-Susłow
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland
| | - Tomasz Janeczko
- Department of Food Chemistry and Biocatalysis, Wrocław University of Environmental and Life Sciences, Wrocław, Norwida 25, 50-375, Poland.
| |
Collapse
|
4
|
Gan J, Kong X, Wang K, Chen Y, Du M, Xu B, Xu J, Wang Z, Cheng Y, Yu T. Effect of fermentation using different lactic acid bacteria strains on the nutrient components and mineral bioavailability of soybean yogurt alternative. Front Nutr 2023; 10:1198456. [PMID: 37426196 PMCID: PMC10327429 DOI: 10.3389/fnut.2023.1198456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/29/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction Analysis of the composition of yogurt alternatives (YAs) during fermentation provides critical information for evaluating its quality and nutritional attributes. Method We investigated the effects of homotypic (HO) and heterotypic (HE) lactic acid bacteria on the nutritional and mineral bioavailabilities of soybean YA (SYA) during fermentation. Result The acidic amino acid (Glu, Asp) and organic acid contents in HO-fermented YA were increased from 2.93, 1.71, and 7.43 mg/100 g to 3.23, 1.82, and 73.47 mg/100 g, respectively. Moreover, both HO and HE lactic acid bacteria fermentation enhanced mineral absorptivity. They altered the molecular speciation of minerals from a large molecular type (2,866 Da) to a small molecular type (1,500 Da), which was manifested in a time-dependent manner. Furthermore, YA substantially increased the bone mass in a zebrafish osteoporosis model, further highlighting the potential of lactic acid bacterial fermentation for mineral bioavailability. Discussion This study provides a foundation for understanding the effects of fermentation conditions on the composition and bioavailability of minerals in YA and can assist in its production.
Collapse
Affiliation(s)
- Jing Gan
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Xiao Kong
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Kuaitian Wang
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Yuhang Chen
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Mengdi Du
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Bo Xu
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Jingru Xu
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| | - Yongqiang Cheng
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Tianying Yu
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong, China
| |
Collapse
|
5
|
Michalkova R, Kello M, Cizmarikova M, Bardelcikova A, Mirossay L, Mojzis J. Chalcones and Gastrointestinal Cancers: Experimental Evidence. Int J Mol Sci 2023; 24:ijms24065964. [PMID: 36983038 PMCID: PMC10059739 DOI: 10.3390/ijms24065964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal (CRC) and gastric cancers (GC) are the most common digestive tract cancers with a high incidence rate worldwide. The current treatment including surgery, chemotherapy or radiotherapy has several limitations such as drug toxicity, cancer recurrence or drug resistance and thus it is a great challenge to discover an effective and safe therapy for CRC and GC. In the last decade, numerous phytochemicals and their synthetic analogs have attracted attention due to their anticancer effect and low organ toxicity. Chalcones, plant-derived polyphenols, received marked attention due to their biological activities as well as for relatively easy structural manipulation and synthesis of new chalcone derivatives. In this study, we discuss the mechanisms by which chalcones in both in vitro and in vivo conditions suppress cancer cell proliferation or cancer formation.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martina Cizmarikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Annamaria Bardelcikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
6
|
Liu XY, Zhang YB, Yang XW, Wu XW, Yang YF, Xu W, Wan MQ, Gong Y, Liu NF, Zhang P. Biological analysis of constituents in Spatholobi Caulis by UFLC-MS/MS: Enhanced quantification and application to permeability properties study in Caco-2 cell monolayer model. J Pharm Biomed Anal 2023; 226:115235. [PMID: 36680806 DOI: 10.1016/j.jpba.2023.115235] [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: 09/06/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023]
Abstract
Major chemical constituents in medicinal materials are often used as the marker compounds of traditional Chinese medicine (TCM) for treating various diseases. For spatholobi caulis (SPC), it contains a variety of flavones, phenolic acid esters, and lignans which exert many pharmacological effects. However, the absorption and permeability properties of these constituents of SPC are still unclear and require further investigation. Different types and major compounds of SPC were chosen as representative constituents to study their absorption and transepithelial transport characteristics in the human intestinal epithelium-like Caco-2 cell monolayer model. 35 constituents of SPC were evaluated by using ultra fast liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry (UFLC-MS/MS) method, acetonitrile and water containing with 0.5 mM ammonium acetate were used as mobile phase, these analytes with good linear relationships (R2 was within 0.9967-0.9998), precision (CV values were less than 10.23 %, LLOQ was less than 13.69 %), accuracy (Mean of inter- and intra-day were within 85.02 %-111.61 % and 85.50-112.97 %, respectively) and stability (The mean was within 85.07 %-113.93 %), among which 16 analytes showed good permeability, 5 analytes were considered to be poorly permeable compounds, and the other 14 analytes were assigned for the moderately absorbed compounds in Caco-2 cell monolayer model. The further results showed that the absorption mechanism of 7 well absorbed compounds, 8-O-methylretusin (1), genistein (7), spasuberol B (16), naringenin (18), isoliquiritigenin (19), 4-hydroxy-3-methoxy cinnamic acid methyl ester (23) and (+)-epipinoresinol (31) in SPC was mainly passive diffusion, their bidirectional transport rate was correlated with the concentration and transport time. The chemical structures of these compounds could affect the permeability properties on the cell monolayer. This study demonstrated the utility of Caco-2 cell monolayer model for evaluating the absorption properties and initial mechanisms of compounds in SPC in vitro, and provided important basis for predicting oral bioavailability of SPC compounds.
Collapse
Affiliation(s)
- Xiao-Yan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
| | - You-Bo Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
| | - Xiu-Wei Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China.
| | - Xiu-Wen Wu
- School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Yan-Fang Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
| | - Wei Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Haidian District, Beijing 100191, China
| | - Mei-Qi Wan
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yun Gong
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412003, China
| | - Ni-Fu Liu
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412003, China
| | - Peng Zhang
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412003, China
| |
Collapse
|
7
|
da Silva ECS, Bernardo Guerra GC, de Araújo ERD, Schlamb J, da Silva VC, de Aragão Tavares E, Dantas-Medeiros R, Abreu LS, Fechine Tavares J, de Araújo Júnior RF, Esposito D, Moncada M, Maria Zucolotto S. Phenolic-rich extract of Nopalea cochenillifera attenuates gastric lesions induced in experimental models through inhibiting oxidative stress, modulating inflammatory markers and a cytoprotective effect. Food Funct 2023; 14:3242-3258. [PMID: 36928439 DOI: 10.1039/d2fo03735a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Nopalea cochenillifera (Cactaceae), popularly known as "palma" or "palma doce", is from Mexico, but it was widely introduced in Brazil through crops. It has been used as food and in traditional medicine and is a good source of phenolic compounds. In this study the phytochemical profile and gastroprotective activity of phenolic-rich extract of N. cochenillifera in acute gastric lesion models induced by ethanol and indomethacin were evaluated. High-performance liquid chromatography coupled with mass spectrometry (HPLC/ESI/MSn) allowed the characterization of 12 compounds such as sugars, phenolics and flavonoids. Among polyphenols, the main peak was assigned to isorhamnetin-3-O-(2'',3''-O-di-rhamnose)-glucoside. The TPC and TFC in the dry extract were 67.85 mg of gallic acid equivalent per g/extract and 46.16 mg quercetin equivalent per g/extract, respectively. In the in vitro MTT assay, the extract showed no cytotoxicity and suppressed ROS levels in LPS-treated RAW 264.7 cells. Preclinical models in rats showed that a dose of 100 mg kg-1 (p < 0.0001) in the ethanol model and doses of 100 mg kg-1 (p < 0.5) and 200 mg kg-1 (p < 0.01) in the indomethacin model reduced the gastric lesions. Also, the extract reduced the MPO, MDA, TNF-α and IL-1β levels and increased the GSH and IL-10 levels. The pre-treatment with the extract led to the upregulation of SOD and the downregulation of COX-2 by immunohistochemical analysis. It also showed a cytoprotective effect in the histopathological analysis and stimulated the restoration of the mucus content as observed in the periodic acid-Schiff analysis without modifying the pH, volume or total acidity of the gastric juice. Taken together, N. cochenillifera extract can be applied as a novel gastroprotective ingredient for food or pharmaceutical products.
Collapse
Affiliation(s)
| | - Gerlane Coelho Bernardo Guerra
- Graduate Program in Health Science, Federal University of Rio Grande do Norte, Natal, Brazil. .,Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil.
| | | | - Jade Schlamb
- Cancer and Inflammation Research Laboratory, Morphology Department, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil. .,Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC, 28081, USA.
| | - Valéria Costa da Silva
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Emanuella de Aragão Tavares
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Renato Dantas-Medeiros
- Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Lucas Silva Abreu
- Department of Organic Chemistry, Institute of Chemistry, Fluminense Federal University, Niterói, RJ, Brazil.
| | | | - Raimundo Fernandes de Araújo Júnior
- Cancer and Inflammation Research Laboratory, Morphology Department, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil.
| | - Debora Esposito
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC, 28081, USA. .,Department of Animal Science, NC State University, 120 Broughton Drive, Raleigh, North Carolina 27695, USA.
| | - Marvin Moncada
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC, 28081, USA. .,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC, 27695, USA.
| | - Silvana Maria Zucolotto
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil. .,Graduate Program in Health Science, Federal University of Rio Grande do Norte, Natal, Brazil. .,Graduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil. .,Research Group on Bioactive Natural Products, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil. .,Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC, 28081, USA.
| |
Collapse
|
8
|
Araújo ERD, Xavier-Santos JB, da Silva VC, de Lima JBF, Schlamb J, Fernandes-Pedrosa MDF, da Silva Júnior AA, de Araújo Júnior RF, Rathinasabapathy T, Moncada M, Esposito D, Guerra GCB, Zucolotto SM. Gel formulated with Bryophyllum pinnatum leaf extract promotes skin wound healing in vivo by increasing VEGF expression: A novel potential active ingredient for pharmaceuticals. Front Pharmacol 2023; 13:1104705. [PMID: 36712663 PMCID: PMC9877235 DOI: 10.3389/fphar.2022.1104705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Bryophyllum pinnatum (Crassulaceae) is used in traditional medicine for treating skin wounds. In our previous study, a topical gel containing B. pinnatum aqueous leaf extract showed a preclinical anti-inflammatory effect in in vivo acute edema models. In continuation, the present study aims to evaluate the phytochemical content and the stability of a formulation in gel containing B. pinnatum aqueous leaf extract and its healing properties and mechanism of action through an experimental model of induction of skin wounds in rats and in vitro assays. The animals were treated topically for 7 or 14 days with a formulation in gel containing extract at 5% or a placebo or Fibrinase® in cream. In addition, to establish some quality control parameters, the total phenolic content (TPC), total flavonoid content (TFC), and a study focusing on the phytochemical and biological stability of a gel for 30 days at two different conditions (room temperature and 40°C/75% RH) were performed. Gel formulation containing extract showed a TPC and TFC of 2.77 ± 0.06 mg of gallic acid/g and 1.58 ± 0.03 mg of quercetin/g, respectively. Regarding the stability study, the formulation in gel showed no significant change in the following parameters: pH, water activity, chromatographic profile, and the content of the major compound identified in the extract. The gel formulation containing extract stimulated skin wound healing while reducing the wound area, as well as decreasing the inflammatory infiltrate, reducing the levels of IL-1β and TNF-α, and stimulating angiogenesis with increased expression of VEGF, an effect similar to Fibrinase. In conclusion, the gel formulation containing extract exhibited relevant skin wound healing properties and, therefore, has the potential to be applied as a novel active ingredient for developing wound healing pharmaceuticals.
Collapse
Affiliation(s)
- Edilane Rodrigues Dantas Araújo
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil,Department of Pharmacy, Research Group on Bioactive Natural Products, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jacinthia Beatriz Xavier-Santos
- Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, Brazil,Biotechnology and Technology Laboratory, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Valéria Costa da Silva
- Department of Pharmacy, Research Group on Bioactive Natural Products, Federal University of Rio Grande do Norte, Natal, Brazil,Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Jade Schlamb
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Matheus de Freitas Fernandes-Pedrosa
- Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, Brazil,Biotechnology and Technology Laboratory, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil,Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Arnóbio Antônio da Silva Júnior
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil,Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, Brazil,Biotechnology and Technology Laboratory, Department of Pharmacy, Federal University of Rio Grande do Norte, Natal, Brazil,Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Raimundo Fernandes de Araújo Júnior
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil,Cancer and Inflammation Research Laboratory, Morphology Department, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Marvin Moncada
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States
| | - Debora Esposito
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States,Department of Animal Science, NC State University, Raleigh, NC, United States
| | - Gerlane Coelho Bernardo Guerra
- Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, Brazil,Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Silvana Maria Zucolotto
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil,Department of Pharmacy, Research Group on Bioactive Natural Products, Federal University of Rio Grande do Norte, Natal, Brazil,Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte, Natal, Brazil,Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil,Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States,*Correspondence: Silvana Maria Zucolotto,
| |
Collapse
|
9
|
Malekinejad M, Pashaee MR, Malekinejad H. 18β-Glycyrrhetinic acid altered the intestinal permeability in the human Caco-2 monolayer cell model. Eur J Nutr 2022; 61:3437-3447. [PMID: 35578042 DOI: 10.1007/s00394-022-02900-4] [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: 10/07/2021] [Accepted: 04/27/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE Glycyrrhizin (GL) and its metabolites 18α-glycyrrhetinic acid (18α-GA) and 18β-glycyrrhetinic acid (18β-GA) are used as traditional medicine and food sweeteners. As the major rout of their administration is oral way, therefore their impact on intestinal epithelial cells are investigated. METHODS The effects of GL and its metabolites on cell viability using MTT assay, on cytotoxicity using LDH release, on integrity of intestinal epithelial cells by measuring the transepithelial electrical resistance (TEER) and Luciferase permeability tests, on the expression of tight junction proteins at mRNA and protein level by qPCR and western blot techniques, and ultimately on the rate of test compounds absorption via Caco-2 cells monolayer were investigated. RESULTS MTT assay showed a concentration- and time-dependent decrease in metabolic activity of Caco-2 cells induced by GL, 18α-GA, and 18β-GA, while only 18β-GA increased the LDH leakage. The monolayer integrity of Caco-2 cells in TEER assay only was affected by 18β-GA. The permeability of paracellular transport marker was increased by 18α-GA and 18β-GA and not GL. In transport studies, only metabolites were able to cross from Caco-2 cells monolayer. qPCR analyses revealed that 18β-GA upregulated the expression of claudin-1 and -4, occludin, junctional adhesion molecules and zonula occludens-1, while 18α-GA upregulated only claudin-4. The expression of claudin-4 at protein level was downregulated non-significantly at 50 μM concentration of 18β-GA. CONCLUSION Our results suggest that 18β-GA may cause cellular damages at higher concentrations on gastrointestinal cells and requires a remarkable attention of the nutraceutical and pharmaceutical industries.
Collapse
Affiliation(s)
- Mojtaba Malekinejad
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran.,Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Reza Pashaee
- Department of Internal Medicine, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hassan Malekinejad
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran. .,Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
10
|
Bartley T, Reese P, Turfus S, Alexander-Lindo R. Validating the Hypoglycaemic and Hypotensive Roles of Salvia serotina (Chicken Weed) in Normal Healthy Sprague-Dawley Rats. ScientificWorldJournal 2022; 2022:6547734. [PMID: 35812002 PMCID: PMC9259359 DOI: 10.1155/2022/6547734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
Diabetes mellitus (DM) is an endocrine disease and is characterized by hyperglycaemia. Salvia serotina L. (chicken weed) has been used in traditional medicine to treat various ailments including DM. Aqueous, hexane, ethyl acetate, and methanol crude extracts of S. serotina L. were investigated for their anti-oxidant activities and hypoglycaemic and hypotensive effects in normal, healthy Sprague-Dawley rats using the 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay, oral glucose tolerance test (OGTT), and the CODA noninvasive blood pressure system to determine systolic blood pressure (SBP), diastolic blood pressure (DBP), mean arterial pressure (MAP), and heart rate (HR). The aqueous extract caused a free radical scavenging effect with an IC50 value of 10.2 ± 1.01 µg/mL versus vitamin C (9.42 ± 1.01 µg/mL). The extract lowered the blood glucose concentration at the 150 minute interval (5.00 ± 0.22 mM vs. 6.51 ± 0.33 mM; p = 0.004) and the 180 minute interval (4.77 ± 0.27 mM vs. 5.93 ± 0.0.30 mM; p = 0.015). The hexane extract gave significant hypoglycaemic activity at the 120 minute interval (4.54 ± 0.21 mmol/L vs. 5.50 ± 0.17 mmol/L; p = 0.005). The hexane extract also significantly lowered the SBP (132 ± 6 mm Hg; p = 0.014), DBP (106 ± 7 mm Hg; p = 0.034), and MAP (114 ± 7 mm Hg; p = 0.023) versus the controls SBP (156 ± 4 mm Hg), DBP (132 ± 8 mm Hg), and MAP (140 ± 6 mm Hg). Bioassay-directed purification of the hexane extract yielded 3,7,11-trimethyl-1,6,10-dodecatrien-3-ol (1), 3,7,11-trimethyl-2,6,10-dodecatrien-1-ol (2), and 5,22-stigmastadien-3β-ol (3) as active principles. Hence, S. serotina L. showed anti-oxidant, hypoglycaemic, and hypotensive effects in the rats and may have potential applications in the treatment of diabetes.
Collapse
Affiliation(s)
- Tameika Bartley
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica
| | - Paul Reese
- Department of Chemistry, Faculty of Science & Technology, The University of the West Indies, Kingston 7, Jamaica
| | | | - Ruby Alexander-Lindo
- Department of Basic Medical Sciences, Faculty of Medical Sciences, The University of the West Indies, Kingston 7, Jamaica
| |
Collapse
|
11
|
Zhang M, Geng T, Jing SH, Li L, Wang K, Zhang Q, Zhang Y, Zhou SK, Gao P, Ding AW, Zhang L. Chemical profile and miscarriage prevention evaluation of Jiao-ai decoction, a classical traditional Chinese formula. J Pharm Biomed Anal 2022; 217:114832. [DOI: 10.1016/j.jpba.2022.114832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 11/28/2022]
|
12
|
Cheng H, Jia X, Yuan D, Li H, Wang L, Fu T, Qiao H, Chen J, Wang Z, Cui X, Cheng J, Li J. Excipient-free nanodispersions dominated by amphiphilic glycosides for bioavailability enhancement of hydrophobic aglycones, a case of glycyrrhetinic acid with diammonium glycyrrhizinate. Int J Pharm 2022; 620:121770. [PMID: 35483618 DOI: 10.1016/j.ijpharm.2022.121770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022]
Abstract
Natural aglycones, a major ingredient accompanied by glycosides in plants, have played an important role in the treatment of various diseases. However, their bioavailability is limited by their poor water solubility. In contrast to previous efforts that required the use of new exotic materials which may raise concerns about biocompatibility, we report the first case of excipient-free nanodispersions in which an insoluble glycyrrhetinic acid (GA) assembled with its amphiphilic parent drug diammonium glycyrrhizinate (DG) into water-dispersible nanodispersions (130.8 nm for particle size and 91.74% for encapsulation efficiency). This strategy largely increased GA's water apparent solubility by hundreds of times to 549.0 μg/mL with a high cumulative dissolution percentage in vitro greater than 80% in 5 min. The study on the formation mechanism showed that the OH, C-O and C=O group stretching peaks shifted in the FTIR spectra of GA-DG nanodispersions, while the COOH peak (δ COOH 12.19 ppm) disappeared in the 1H NMR spectrum of GA-DG nanodispersions, indicating that carboxyl groups on GA may interact with the hydroxyl groups of DG in solution. Molecular dynamics simulations suggested that both hydrophobic interactions and hydrogen-bond interactions contribute to the coassembly of GA and DG molecules in aqueous solution. Oral pharmacokinetic studies in rats demonstrated that such nanodispersions have a significant increase in Cmax and AUC0-t of 2.45- and 3.45-fold compared with those for GA, respectively. Therefore, this strategy, employing amphiphilic glycosides as excipients to prepare nanodispersions, not using new materials, paves the way for the further application of hydrophobic aglycone drugs.
Collapse
Affiliation(s)
- Hongqing Cheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing 210023, PR China
| | - Xiaoshun Jia
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing 210023, PR China
| | - Dandan Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Huaning Li
- Medical Department, Weifang Medical College, Weifang 261042, PR China
| | - Lingchong Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing 210023, PR China
| | - Tingming Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Hongzhi Qiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing 210023, PR China
| | - Jing Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zengwu Wang
- Medical Department, Weifang Medical College, Weifang 261042, PR China
| | - Xiaobing Cui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Jianming Cheng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Province Engineering Research Center of Classical Prescription, Nanjing 210023, PR China.
| | - Junsong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System (DDS), Nanjing 210023, PR China.
| |
Collapse
|
13
|
Hao DC, Wang F, Xiao PG. Impact of Drug Metabolism/Pharmacokinetics and their Relevance Upon Traditional Medicine-based anti-COVID-19 Drug Research. Curr Drug Metab 2022; 23:374-393. [PMID: 35440304 DOI: 10.2174/1389200223666220418110133] [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: 12/30/2021] [Revised: 01/22/2022] [Accepted: 02/01/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND The representative anti-COVID-19 herbs, i.e. Poria cocos, Pogostemon, Prunus, and Glycyrrhiza plants, are commonly used in the prevention and treatment of COVID-19, a pandemic caused by SARS-CoV-2. Diverse medicinal compounds with favorable anti-COVID-19 activities are abundant in these plants, and their unique pharmacological/pharmacokinetic properties are being revealed. However, the current trends of drug metabolism/pharmacokinetic (DMPK) investigations of anti-COVID-19 herbs have not been systematically summarized. METHODS Here, the latest awareness, as well as the perception gaps of DMPK attributes, in the anti-COVID-19 drug development and clinical usage was elaborated and critically commented. RESULTS The extracts and compounds of P. cocos, Pogostemon, Prunus, and Glycyrrhiza plants show distinct and diverse absorption, distribution, metabolism, excretion and toxicity (ADME/T) properties. The complicated herb-herb interactions (HHIs) and herb-drug interactions (HDIs) of anti-COVID-19 Traditional Chinese Medicine (TCM) herb pair/formula dramatically influence the PK/pharmacodynamic (PD) performance of compounds thereof, which may inspire researchers to design innovative herbal/compound formulas for optimizing the therapeutic outcome of COVID-19 and related epidemic diseases. The ADME/T of some abundant compounds in anti-COVID-19 plants have been elucidated, but DMPK studies should be extended to more compounds of different medicinal parts, species and formulations, and would be facilitated by various omics platforms and computational analyses. CONCLUSION In the framework of systems pharmacology and pharmacophylogeny, the DMPK knowledge base would promote the translation of bench findings into the clinical practice of anti-COVID-19, and speed up the anti-COVID-19 drug discovery and development.
Collapse
Affiliation(s)
- Da-Cheng Hao
- Biotechnology Institute, School of Environment and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, China.,Institute of Molecular Plant Science, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Fan Wang
- Biotechnology Institute, School of Environment and Chemical Engineering, Dalian Jiaotong University, Dalian 116028, China
| | - Pei-Gen Xiao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
14
|
Fernández-Ochoa Á, Cádiz-Gurrea MDLL, Fernández-Moreno P, Rojas-García A, Arráez-Román D, Segura-Carretero A. Recent Analytical Approaches for the Study of Bioavailability and Metabolism of Bioactive Phenolic Compounds. Molecules 2022; 27:777. [PMID: 35164041 PMCID: PMC8838714 DOI: 10.3390/molecules27030777] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/14/2022] Open
Abstract
The study of the bioavailability of bioactive compounds is a fundamental step for the development of applications based on them, such as nutraceuticals, functional foods or cosmeceuticals. It is well-known that these compounds can undergo metabolic reactions before reaching therapeutic targets, which may also affect their bioactivity and possible applications. All recent studies that have focused on bioavailability and metabolism of phenolic and terpenoid compounds have been developed because of the advances in analytical chemistry and metabolomics approaches. The purpose of this review is to show the role of analytical chemistry and metabolomics in this field of knowledge. In this context, the different steps of the analytical chemistry workflow (design study, sample treatment, analytical techniques and data processing) applied in bioavailability and metabolism in vivo studies are detailed, as well as the most relevant results obtained from them.
Collapse
Affiliation(s)
- Álvaro Fernández-Ochoa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Berlin Institute of Health, Metabolomics Platform, 10178 Berlin, Germany
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| | - María de la Luz Cádiz-Gurrea
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| | - Patricia Fernández-Moreno
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| | - Alejandro Rojas-García
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| | - David Arráez-Román
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Fuentenueva s/n, E-18071 Granada, Spain; (M.d.l.L.C.-G.); (P.F.-M.); (A.R.-G.); (A.S.-C.)
| |
Collapse
|
15
|
van Dinteren S, Meijerink J, Witkamp R, van Ieperen B, Vincken JP, Araya-Cloutier C. Valorisation of liquorice ( Glycyrrhiza) roots: antimicrobial activity and cytotoxicity of prenylated (iso)flavonoids and chalcones from liquorice spent ( G. glabra, G. inflata, and G. uralensis). Food Funct 2022; 13:12105-12120. [DOI: 10.1039/d2fo02197h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prenylated phenolics are antimicrobials found in liquorice (Glycyrrhiza spp.).
Collapse
Affiliation(s)
- Sarah van Dinteren
- Laboratory of Food Chemistry, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - Jocelijn Meijerink
- Division of Human Nutrition and Health, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - Renger Witkamp
- Division of Human Nutrition and Health, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - Bo van Ieperen
- Division of Human Nutrition and Health, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - Jean-Paul Vincken
- Laboratory of Food Chemistry, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| | - Carla Araya-Cloutier
- Laboratory of Food Chemistry, Wageningen University, P.O. box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
16
|
De Araújo ERD, Guerra GCB, Andrade AWL, Fernandes JM, Da Silva VC, De Aragão Tavares E, De Araújo AA, de Araújo Júnior RF, Zucolotto SM. Gastric Ulcer Healing Property of Bryophyllum pinnatum Leaf Extract in Chronic Model In Vivo and Gastroprotective Activity of Its Major Flavonoid. Front Pharmacol 2021; 12:744192. [PMID: 34975468 PMCID: PMC8717929 DOI: 10.3389/fphar.2021.744192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Gastric ulcer is a common disease that develops complications such as hemorrhages and perforations when not properly treated. Extended use of drugs in the treatment of this pathology can provoke many adverse effects. Therefore, finding medicinal plants with gastroprotective and mucosal healing properties has gained increasing interest. Bryophyllum pinnatum (Crassulaceae), popularly known in Brazil as “saião” or “coirama,” has been used to treat inflammatory disorders. It is rich in flavonoids, and quercetin 3-O-α-L-arabinopyranosyl-(1→2)-O-α-L-rhamnopyranoside-Bp1 is its major compound. In this study, we aimed to investigate ulcer healing properties of B. pinnatum against an acetic acid–induced chronic ulcer model and the gastroprotective activity of Bp1 against gastric lesions induced by ethanol and indomethacin. Ultrafast liquid chromatography was used to quantify the main compounds (mg/g of the extract)—quercetin 3-O-α-L-arabinopyranosyl-(1→2)-O-α-L-rhamnopyranoside (33.12 ± 0.056), kaempferol 3-O-α-L-arabinopyranosyl-(1→2)-O-α-L-rhamnopyranoside (3.98 ± 0.049), and quercetin 3-O-α-L-rhamnopyranoside (4.26 ± 0.022) and showed good linearity, specificity, selectivity, precision, robustness, and accuracy. In vivo studies showed that treatment with the extract at 250 and 500 mg/kg stimulated the healing process in the gastric mucosa with significant ulceration index reduction, followed by improvement in the antioxidant defense system [increased glutathione (GSH) levels, decreased superoxide dismutase upregulation, and malondialdehyde (MDA) levels]. Moreover, the extract decreased interleukin-1β and tumor necrosis factor-a levels and myeloperoxidase (MPO) activity, increased interleukin 10 levels, showed a cytoprotective effect in histological analyzes and also downregulated the expression of cyclooxygenase-2 and NF-κB (p65). The pretreatment with Bp1 at a dose of 5 mg/kg reduced gastric lesions in the ethanol and indomethacin models, increased GSH, and decreased MDA levels. In addition, the pretreatment decreased MPO activity, interleukin-1β and tumor necrosis factor-α levels, while also showing a cytoprotective effect in histological analyzes. Our study suggests that treatment with B. pinnatum extract showed a higher inhibition percentage than pretreatment with the Bp1. This might in turn suggest that Bp1 has gastroprotective activity, but other compounds can act synergistically, potentiating its effect. We conclude that B. pinnatum leaf extract could be a new source of raw material rich in phenolic compounds to be applied in food or medicine.
Collapse
Affiliation(s)
| | | | - Anderson Wilbur Lopes Andrade
- Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Júlia Morais Fernandes
- Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | - Valéria Costa Da Silva
- Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Emanuella De Aragão Tavares
- Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Raimundo Fernandes de Araújo Júnior
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
- Postgraduate Program in Functional and Structural Biology, Department of Morphology, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
- Translational Nanobiomaterials and Imaging (TNI) Group, Radiology Department, Leiden University Medical Centrum, Leiden, Netherlands
- Percuros B.V, Leiden, Netherlands
| | - Silvana Maria Zucolotto
- Postgraduate Program in Health Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
- Postgraduate Program in Drug Development and Technological Innovation, Federal University of Rio Grande do Norte, Natal, Brazil
- Postgraduate Program in Pharmaceutical Science, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
- *Correspondence: Silvana Maria Zucolotto,
| |
Collapse
|
17
|
Shen Y, Zhang N, Tian J, Xin G, Liu L, Sun X, Li B. Advanced approaches for improving bioavailability and controlled release of anthocyanins. J Control Release 2021; 341:285-299. [PMID: 34822910 DOI: 10.1016/j.jconrel.2021.11.031] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
Anthocyanins are a group of phytochemicals responsible for the purple or red color of plants. Additionally, they are recognized to have health promoting functions including anti-cardiovascular, anti-thrombotic, anti-diabetic, antimicrobial, neuroprotective, and visual protective effect as well as anti-cancer activities. Thus, consumption of anthocyanin supplement or anthocyanin-rich foods has been recommended to prevent the risk of development of chronic diseases. However, the low stability and bioavailability of anthocyanins limit the efficacy and distribution of anthocyanins in human body. Thus, strategies to achieve target site-local delivery with good bioavailability and controlled release rate are necessary. This review introduced and discussed the latest advanced techniques of designing lipid-based, polysaccharide-based and protein-based complexes, nano-encapsulation and exosome to overcome the limitation of anthocyanins. The improved bioavailability and controlled release of anthocyanins have great significance for gastrointestinal tract absorption, transepithelial transportation and cellular uptake. The techniques of applying different biocompatible materials and modifying the solubility of anthocyanins complex could achieve target site-local delivery with negligible degradation and good bioavailability in human body.
Collapse
Affiliation(s)
- Yixiao Shen
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Ning Zhang
- College of Horticulture Science and Technology, Hebei Normal University of Science & Technology, Hebei Key Laboratory of Horticulture Germplasm Excavation and Innovative Utilization Qinhuangdao, Hebei, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Guang Xin
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Ling Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Xiyun Sun
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110161, China.
| |
Collapse
|
18
|
Poli CHEC, Thornton-Kurth KJ, Legako JF, Bremm C, Hampel VS, Hall J, Ipharraguerre IR, Villalba JJ. The effect of plant bioactive compounds on lamb performance, intake, gastrointestinal parasite burdens, and lipid peroxidation in muscle. J Anim Sci 2021; 99:6102878. [PMID: 33454733 DOI: 10.1093/jas/skab009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/14/2021] [Indexed: 01/04/2023] Open
Abstract
Plant extracts have been widely used in animal production systems due to the benefits promoted by their bioactive components, mainly through their antioxidant effects and positive effects on animal growth, health, and meat quality. We determined whether bioactive compounds (BC) from grape (Vitis vinifera), olive (Olea europaea), and pomegranate (Punica granatum) have beneficial effects on lamb growth while controlling gastrointestinal parasitism and reducing skeletal lipid peroxidation in muscle. Fourteen lambs (4 to 5 mo of age) were housed in individual pens and assigned to two treatment groups (seven animals/group), where they received: 1) a basal diet of beet pulp:soybean meal (90:10) (CONTROL) and 2) the same diet, but containing 0.3% of BC extracted from grape, olive, and pomegranate (BNP). After 21 d of consuming their respective rations (period 1), all lambs were dosed with 10,000 L3 stage larvae of Haemonchus contortus. Intake, production, and physiological parameters were determined before infection (period 1), after oral dosage with L3 (period 2), and during early (period 3) and later (period 4) developmental stages of infection. Groups of animals did not differ in their ration intake or average daily gain, either before or after the infection, or in their parasitic burdens estimated through fecal egg counts (P > 0.05). Lambs under BNP showed greater feed efficiency before infection (P < 0.001), but the pattern reversed after infection. In addition, the inclusion of plant bioactives to the diet did not have an effect (P > 0.05) on blood or lipid peroxidation in muscle or on hemogram, serum concentration of haptoglobin, and immunoglobulin E. These results could be explained by low dietary doses and constrained absorption or ruminal inactivation of these compounds. Changes in feed efficiencies suggest a negative interaction between BC in the diet and parasitism, which warrants further research.
Collapse
Affiliation(s)
- Cesar H E C Poli
- Programa de Pós-Graduação em Zootecnia, Faculdade de Agronomia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | - Carolina Bremm
- Programa de Pós-Graduação em Zootecnia, Faculdade de Agronomia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Viviane S Hampel
- Programa de Pós-Graduação em Zootecnia, Faculdade de Agronomia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Ignacio R Ipharraguerre
- Institute of Human Nutrition and Food Science, Christian-Albrechts University, Kiel, Germany
| | | |
Collapse
|
19
|
Antihyperglycemic and Lipid Profile Effects of Salvia amarissima Ortega on Streptozocin-Induced Type 2 Diabetic Mice. Molecules 2021; 26:molecules26040947. [PMID: 33670091 PMCID: PMC7916882 DOI: 10.3390/molecules26040947] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022] Open
Abstract
Salvia amarissima Ortega was evaluated to determinate its antihyperglycemic and lipid profile properties. Petroleum ether extract of fresh aerial parts of S. amarissima (PEfAPSa) and a secondary fraction (F6Sa) were evaluated to determine their antihyperglycemic activity in streptozo-cin-induced diabetic (STID) mice, in oral tolerance tests of sucrose, starch, and glucose (OSTT, OStTT, and OGTT, respectively), in terms of glycated hemoglobin (HbA1c), triglycerides (TG), and high-density lipoprotein (HDL). In acute assays at doses of 50 mg/kg body weight (b.w.), PEfAPSa and F6Sa showed a reduction in hyperglycemia in STID mice, at the first and fifth hour after of treatment, respectively, and were comparable with acarbose. In the sub-chronic test, PEfAPSa and F6Sa showed a reduction of glycemia since the first week, and the effect was greater than that of the acarbose control group. In relation to HbA1c, the treatments prevented the increase in HbA1c. In the case of TG and HDL, PEfAPSa and F6Sa showed a reduction in TG and an HDL increase from the second week. OSTT and OStTT showed that PEfAPSa and F6Sa significantly lowered the postprandial peak at 1 h after loading but only in sucrose or starch such as acarbose. The results suggest that S. amarissima activity may be mediated by the inhibition of disaccharide hydrolysis, which may be associated with an α-glucosidase inhibitory effect.
Collapse
|
20
|
Song H, Hou X, Zeng M, Chen X, Chen X, Yang T, Xu F, Peng J, Peng Q, Cai X, Yu R. Traditional Chinese Medicine Li-Zhong-Tang accelerates the healing of indomethacin-induced gastric ulcers in rats by affecting TLR-2/MyD88 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 259:112979. [PMID: 32442585 DOI: 10.1016/j.jep.2020.112979] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/25/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Li-Zhong-Tang (LZT) is a well-known Chinese herbal formulation first described in one of traditional Chinese medicine (TCM) scriptures, Treatise on Febrile Diseases. LZT has been commonly prescribed for the treatment of various gastrointestinal diseases for over 1800 years, and has demonstrated pronounced therapeutic effects on patients with gastric ulcers. AIM OF THE STUDY The present study aimed to scientifically evaluate protective effects of LZT on indomethacin (IND)-induced gastric injury in rats and to elucidate whether LZT exerts its gastro-protective effects via enhancing mucosal immunity by regulating TLR-2/MyD88 signaling pathway. MATERIAL AND METHODS Gastric ulcers were induced in male Sprague-Dawley (SD) rats with a single oral dose of 150 mg/kg IND. Ulcer index (UI) and curative index (CI) were evaluated. Histopathological examinations were performed and microscopic score (MS) was macroscopically calculated. The volume of gastric juice, free acidity, total acidity, and gastric pH was measured. The gastroprotective and inflammatory biomarkers including levels of nitric oxide (NO), tumor necrosis factor-α (TNF-α), prostaglandin E2 (PGE2), and malondialdehyde (MDA) were determined. Expression levels of TLR-2 and MyD88 mRNA were assessed by qRT-PCR. The expression, distribution, and co-localization of TLR-2 and MyD88 protein were determined by Western blot, immunohistochemistry, and immunofluorescence, respectively. RESULTS Induction of gastric ulcers in rats resulted in very significantly increased UI and elevated volume and acidity of gastric juice, which were markedly attenuated by LZT treatment. Microscopic examinations of the IND-induced gastric ulcers revealed severe gastric hemorrhagic necrosis, submucosal edema, and destruction of epithelial cells, which were significantly attenuated in LZT-treated rats. Moreover, treatment with LZT remarkably increased gastric mucosal levels of PGE2 and NO, and lowered highly elevated levels of TNF-α and MDA in gastric ulcerative rats. Mechanistically, LZT inhibited mRNA and protein expression of TLR-2 and MyD88 and enhanced immune function in gastric mucosa. Immunohistochemical analyses and immunofluorescent detection further confirmed a markedly decreased co-localization of TLR-2 and MyD88 protein in the gastric mucosa of LZT-treated rats as compared to that of gastric ulcerative rats. CONCLUSIONS These findings indicate that LZT alleviates serious gastric mucosal ulcerations induced by IND. Protective effects of LZT on gastric ulcers are believed to be associated with the intensification of the anti-oxidative defense system, mitigation of proinflammatory cytokines, stimulation of the production of cytoprotective mediators, and improvement of the mucosal immunity through TLR-2/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Houpan Song
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xueqin Hou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai' an, Shandong, 271016, China.
| | - Meiyan Zeng
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xiaojuan Chen
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xinyi Chen
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Tao Yang
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Fuping Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Jun Peng
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Qinghua Peng
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xiong Cai
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Rong Yu
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
21
|
Zhang L, Wang CX, Wu J, Wang TY, Zhong QQ, Du Y, Ji S, Wang L, Guo MZ, Xu SQ, Tang DQ. Metabolic profiling of mice plasma, bile, urine and feces after oral administration of two licorice flavonones. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112892. [PMID: 32320727 DOI: 10.1016/j.jep.2020.112892] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Licorice is an ancient food and medicinal plant. Liquiritigenin and liquiritin, two kinds of major flavonoes in licorice, are effective substances used as antioxidant, anti-inflammatory and tumor-suppressive food, cosmetics or medicines. However, their in vivo metabolites have not been fully explored. AIM OF STUDY To clarify the metabolism of liquiritigenin and liquiritin in mice. MATERIALS AND METHODS In this study, we developed a liquid chromatography coupled with quadrupole/time-of-flight mass spectrometry approach to determine the metabolites in mice plasma, bile, urine and feces after oral administration of liquiritigenin or liquiritin. The structures of those metabolites were tentatively identified according to their fragment pathways, accurate masses, characteristic product ions, metabolism laws or reference standard matching. RESULTS A total of 26 and 24 metabolites of liquiritigenin or liquiritin were respectively identified. The products related with apigenin, luteolin or quercetin were the major metabolites of liquiritigenin or liquiritin in mice. Seven main metabolic pathways including (de)hydrogenation, (de)hydroxylation, (de)glycosylation, (de)methoxylation, acetylation, glucuronidation and sulfation were summarized to tentatively explain their biotransformation. CONCLUSION This study not only can provide the evidence for in vivo metabolites and pharmacokinetic mechanism of liquiritigenin and liquiritin, but also may lay the foundation for further development and utilization of liquiritigenin, liquiritin and then licorice.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pharmacy, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Chen-Xiang Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Jing Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmaceutical Analysis, Jiangsu College of Nursing, Huai'an, China
| | - Tian-Yun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qiao-Qiao Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yan Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Shuai Ji
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou, China
| | - Liang Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Bioinformatics, School of Medical Informatics, Xuzhou Medical University, Xuzhou, China
| | - Meng-Zhe Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou, China
| | - Sheng-Qiu Xu
- Department of Pharmacy, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Dao-Quan Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China; Department of Pharmaceutical Analysis, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
22
|
Domínguez Moré GP, Feltrin C, Brambila PF, Cardona MI, Echeverry SM, Simões CMO, Aragón DM. Matrix effects of the hydroethanolic extract and the butanol fraction of calyces from Physalis peruviana L. on the biopharmaceutics classification of rutin. ACTA ACUST UNITED AC 2020; 72:738-747. [PMID: 32162346 DOI: 10.1111/jphp.13248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/09/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The Biopharmaceutics Classification System (BCS) categorizes active pharmaceutical ingredients according to their solubility and permeability properties, which are susceptible to matrix or formulation effects. The aim of this research was to evaluate the matrix effects of a hydroethanolic extract of calyces from Physalis peruviana L. (HEE) and its butanol fraction (BF), on the biopharmaceutics classification of their major compound, quercetin-3-O-rutinoside (rutin, RU). METHODS Rutin was quantified by HPLC-UV, and Caco-2 cell monolayer transport studies were performed to obtain the apparent permeability values (Papp ). Aqueous solubility was determined at pH 6.8 and 7.4. KEY FINDINGS The Papp values followed this order: BF > HEE > RU (1.77 ± 0.02 > 1.53 ± 0.07 > 0.90 ± 0.03 × 10-5 cm/s). The lowest solubility values followed this order: HEE > RU > BF (2.988 ± 0.07 > 0.205 ± 0.002 > 0.189 ± 0.005 mg/ml). CONCLUSIONS According to these results, rutin could be classified as BCS classes III (high solubility/low permeability) and IV (low solubility/low permeability), depending on the plant matrix. Further work needs to be done in order to establish how apply the BCS for research and development of new botanical drugs or for bioequivalence purposes.
Collapse
Affiliation(s)
- Gina Paola Domínguez Moré
- Programa de Farmacia, Facultad de Química y Farmacia, Centro de Servicios Farmacéuticos y Monitoreo de Fármacos, Universidad del Atlántico, Atlántico, Colombia.,Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Clarissa Feltrin
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Paula Freire Brambila
- Programa de Pós-Graduação em Farmácia, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| | - María Isabel Cardona
- Programa de Pós-Graduação em Farmácia, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| | - Sandra Milena Echeverry
- Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | - Diana Marcela Aragón
- Programa de Pós-Graduação em Farmácia, Universidade Federal de Santa Catarina - UFSC, Florianópolis, Brazil
| |
Collapse
|
23
|
Oh HN, Lee MH, Kim E, Kwak AW, Yoon G, Cho SS, Liu K, Chae JI, Shim JH. Licochalcone D Induces ROS-Dependent Apoptosis in Gefitinib-Sensitive or Resistant Lung Cancer Cells by Targeting EGFR and MET. Biomolecules 2020; 10:biom10020297. [PMID: 32070026 PMCID: PMC7072161 DOI: 10.3390/biom10020297] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 01/26/2023] Open
Abstract
Licochalcone D (LCD), a flavonoid isolated from a Chinese medicinal plant Glycyrrhiza inflata, has a variety of pharmacological activities. However, the anti-cancer effects of LCD on non-small cell lung cancer (NSCLC) have not been investigated yet. The amplification of MET (hepatocyte growth factor receptor) compensates for the inhibition of epidermal growth factor receptor (EGFR) activity due to tyrosine kinase inhibitor (TKI), leading to TKI resistance. Therefore, EGFR and MET can be attractive targets for lung cancer. We investigated the anti-proliferative and apoptotic effects of LCD in lung cancer cells HCC827 (gefitinib-sensitive) and HCC827GR (gefitinib-resistant) through 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, pull-down/kinase assay, cell cycle analysis, Annexin-V/7-ADD staining, reactive oxygen species (ROS) assay, mitochondrial membrane potential (MMP) assay, multi-caspase assay, and Western blot analysis. The results showed that LCD inhibited phosphorylation and the kinase activity of EGFR and MET. In addition, the predicted pose of LCD was competitively located at the ATP binding site. LCD suppressed lung cancer cells growth by blocking cell cycle progression at the G2/M transition and inducing apoptosis. LCD also induced caspases activation and poly (ADP-ribose) polymerase (PARP) cleavage, thus displaying features of apoptotic signals. These results provide evidence that LCD has anti-tumor effects by inhibiting EGFR and MET activities and inducing ROS-dependent apoptosis in NSCLC, suggesting that LCD has the potential to treat lung cancer.
Collapse
Affiliation(s)
- Ha-Na Oh
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea; (H.-N.O.); (A.-W.K.); (G.Y.); (S.-S.C.)
| | - Mee-Hyun Lee
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, Henan, China; (M.-H.L.); (K.L.)
- Basic Medical College, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Eunae Kim
- College of Pharmacy, Chosun University, Gwangju 61452, Korea;
| | - Ah-Won Kwak
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea; (H.-N.O.); (A.-W.K.); (G.Y.); (S.-S.C.)
| | - Goo Yoon
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea; (H.-N.O.); (A.-W.K.); (G.Y.); (S.-S.C.)
| | - Seung-Sik Cho
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea; (H.-N.O.); (A.-W.K.); (G.Y.); (S.-S.C.)
| | - Kangdong Liu
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, Henan, China; (M.-H.L.); (K.L.)
- Basic Medical College, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry, BK21 Plus, Jeonbuk National University, Jeonju 54896, Korea
- Correspondence: (J.-I.C.); or (J.-H.S.); Tel.: +82-63-270-4024 (J.-I.C.); +82-61-450-2684 (J.-H.S.); Fax: +82-63-270-4037 (J.-I.C.); +82-61-450-2689 (J.-H.S.)
| | - Jung-Hyun Shim
- Department of Pharmacy, Mokpo National University, Jeonnam 58554, Korea; (H.-N.O.); (A.-W.K.); (G.Y.); (S.-S.C.)
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, Henan, China; (M.-H.L.); (K.L.)
- Correspondence: (J.-I.C.); or (J.-H.S.); Tel.: +82-63-270-4024 (J.-I.C.); +82-61-450-2684 (J.-H.S.); Fax: +82-63-270-4037 (J.-I.C.); +82-61-450-2689 (J.-H.S.)
| |
Collapse
|
24
|
Wang C, Zhou Y, Gong X, Zheng L, Li Y. In vitro and in situ study on characterization and mechanism of the intestinal absorption of 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-D-glucoside. BMC Pharmacol Toxicol 2020; 21:7. [PMID: 31969193 PMCID: PMC6977318 DOI: 10.1186/s40360-020-0384-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 01/13/2020] [Indexed: 01/29/2023] Open
Abstract
Background 2,3,5,4′-tetrahydroxystilbence-2-O-β-D-glucoside (TSG) is a polyhydroxyphenolic compound, which exhibited a broad spectrum of pharmacological activities, such as anti-inflammatory, anti-depression, anti-oxidation and anti-atherosclerosis. However, the compound had poor bioavailability and the underlying absorption mechanisms had not been studied. Therefore, the purpose of this study was to investigate the intestinal absorption mechanism of TSG. Methods This study used Caco-2 cell monolayer model and single-pass intestinal perfusion model to explore the gastrointestinal absorption mechanisms of TSG. The effects of basic parameters such as drug concentration, time and pH on the intestinal absorption of TSG were analyzed by high performance liquid chromatography. The absorption susceptibility of TSG to three inhibitors, P-gp inhibitors verapamil hydrochloride and quinidine, and MRP2 inhibitor probenecid were also assessed. Results TSG was poorly absorbed in the intestines and the absorption of TSG in stomach is much higher than that in intestine. Both in vitro and in situ experiments showed that the absorption of TSG was saturated with increasing concentration and it was better absorbed in a weakly acidic environment pH 6.4. Moreover, TSG interacts with P-gp and MRP2, and TSG was not only the substrate of the P-gp and MRP2, but also affected the expression of P-gp and MRP2. Conclusions It was concluded that the gastrointestinal absorption the most unique active ingredient and considered as the mechanisms of TSG involved processes passive transport and the participation of efflux transporters.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu, 611137, China.,National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Yimeng Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu, 611137, China.,National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Xiaohong Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu, 611137, China.,National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Li Zheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu, 611137, China.,National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China
| | - Yunxia Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China. .,Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, Chengdu, 611137, China. .,National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, 611137, China.
| |
Collapse
|
25
|
Song H, Zeng M, Chen X, Chen X, Peng J, Lin Y, Yu R, Cai X, Peng Q. Antiulcerogenic Activity of Li-Zhong Decoction on Duodenal Ulcers Induced by Indomethacin in Rats: Involvement of TLR-2/MyD88 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:6538156. [PMID: 32063985 PMCID: PMC6996687 DOI: 10.1155/2020/6538156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/04/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Administration of nonsteroidal anti-inflammatory drugs (NSAIDs) often causes small intestinal ulcers in patients, but few effective drugs are currently available to manage such serious adverse events of NSAIDs. Li-Zhong decoction (LZD), a well-known traditional Chinese medicine (TCM) formula, is commonly prescribed for treatment of gastrointestinal diseases. The present study aimed to investigate the anti-ulcerogenic activity of LZD on indomethacin- (IND-) induced duodenal ulcer in rats. Mechanistic studies of action of LZD were focused on involvement of TLR-2/MyD88 signaling pathway. METHODS Fifty male Sprague-Dawley (SD) rats were randomly and evenly divided into five groups: normal control, ulcer control (IND, 25 mg/kg), IND + esomeprazole (ESO, 4.17 mg/kg), and IND + low and high doses of LZD (3.75 and 7.50 g/kg). Macroscopic and histopathological examinations were performed for evaluation of ulcer index (UI), curative index (CI), and microscopic score (MS). Levels of duodenal inflammatory biomarkers and cytoprotective mediators including interleukin-4 (IL-4), IL-10, tumor necrosis factor-α (TNF-α (TNF. RESULTS Gross and microscopic examinations of the IND-treated rats revealed severe duodenal hemorrhagic necrosis, inflammatory infiltration, villus destruction, and crypt abscess, while LZD-treated rats manifested these pathological events to a markedly lesser degree. LZD significantly decreased UI and MS, increased CI, preserved the integrity of the villus and crypt, and normalized the tissue architecture of the duodenum of rats. The elevated TNF-α (TNF. CONCLUSIONS Our data demonstrate that LZD protects the duodenal mucosa from IND-caused lesions, which is at least partially attributable to the interaction of its potential cytoprotective and anti-inflammatory mechanisms together with enhancement of the mucosal immunity through TLR-2/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Houpan Song
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Meiyan Zeng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiaojuan Chen
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xinyi Chen
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jun Peng
- Department of Ophthalmology, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ye Lin
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rong Yu
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiong Cai
- Institute of Innovation and Applied Research, Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qinghua Peng
- Institute of Traditional Chinese Medicine Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
26
|
Song D, Cheng L, Zhang X, Wu Z, Zheng X. The modulatory effect and the mechanism of flavonoids on obesity. J Food Biochem 2019; 43:e12954. [PMID: 31368555 DOI: 10.1111/jfbc.12954] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/15/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022]
Abstract
With the improvement of living standards, obesity has become a serious health problem all over the word. Currently, the methods and drugs for obesity treatment have some limitations and side effects. Flavonoids are active constituents with various biological activities, widely found in plants, and numerous studies have shown that flavonoids can inhibit obesity and related metabolism disorders effectively. This perspective reviews the recent progress in understanding the anti-obesity effects of flavonoids through modulating food intake, enzyme activities, nutrition absorption, adipogenesis and adipocyte lifecycle, thermogenesis, energy consumption, and intestinal microbiota. PRACTICAL APPLICATIONS: Natural bioactive substance flavonoids have anti-obesity property, which may play a role in anti-obesity drugs or functional food without any side effects. Flavonoids can inhibit weight gain directly or through their biologically active metabolites by various potential pathways. A better understanding of the modulatory effect and the mechanism of flavonoids on obesity will allow us to better utilize flavonoids in plants to treat obesity and related metabolic syndrome.
Collapse
Affiliation(s)
- Dan Song
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Lu Cheng
- Department of Food Science, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Xiaojie Zheng
- Department of Agriculture and Biotechnology, Wenzhou Vocational College of Science and Technology, Wenzhou, P.R. China
| |
Collapse
|
27
|
Speer JE, Gunasekara DB, Wang Y, Fallon JK, Attayek PJ, Smith PC, Sims CE, Allbritton NL. Molecular transport through primary human small intestinal monolayers by culture on a collagen scaffold with a gradient of chemical cross-linking. J Biol Eng 2019; 13:36. [PMID: 31061676 PMCID: PMC6487070 DOI: 10.1186/s13036-019-0165-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The luminal surface of the small intestine is composed of a monolayer of cells overlying a lamina propria comprised of extracellular matrix (ECM) proteins. The ECM provides a porous substrate critical for nutrient exchange and cellular adhesion. The enterocytes within the epithelial monolayer possess proteins such as transporters, carriers, pumps and channels that participate in the movement of drugs, metabolites, ions and amino acids and whose function can be regulated or altered by the properties of the ECM. Here, we characterized expression and function of proteins involved in transport across the human small intestinal epithelium grown on two different culture platforms. One strategy employs a conventional scaffolding method comprised of a thin ECM film overlaying a porous membrane while the other utilizes a thick ECM hydrogel placed on a porous membrane. The thick hydrogel possesses a gradient of chemical cross-linking along its length to provide a softer substrate than that of the ECM film-coated membrane while maintaining mechanical stability. RESULTS The monolayers on both platforms possessed goblet cells and abundant enterocytes and were impermeable to Lucifer yellow and fluorescein-dextran (70 kD) indicating high barrier integrity. Multiple transporter proteins were present in both primary-cell culture formats at levels similar to those present in freshly isolated crypts/villi; however, expression of breast cancer resistance protein (BCRP) and multidrug resistance protein 2 (MRP2) in the monolayers on the conventional scaffold was substantially less than that on the gradient cross-linked scaffold and freshly isolated crypts/villi. Monolayers on the conventional scaffold failed to transport the BCRP substrate prazosin while cells on the gradient cross-linked scaffold successfully transported this drug to better mimic the properties of in vivo small intestine. CONCLUSIONS The results of this comparison highlight the need to create in vitro intestinal transport platforms whose characteristics mimic the in vivo lamina propria in order to accurately recapitulate epithelial function. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Jennifer E. Speer
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Dulan B. Gunasekara
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Peter J. Attayek
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599 USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27599 USA
| |
Collapse
|
28
|
Zhang X, Cheng X, Wu Y, Feng D, Qian Y, Chen L, Yang B, Gu M. In Vitro and In Situ Characterization of the Intestinal Absorption of Capilliposide B and Capilliposide C from Lysimachia capillipes Hemsl. Molecules 2019; 24:molecules24071227. [PMID: 30925820 PMCID: PMC6479817 DOI: 10.3390/molecules24071227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
The goal of this investigation was to determine the processes and mechanism of intestinal absorption for capilliposide B (CAPB) and capilliposide C (CAPC) from the Chinese herb, Lysimachia capillipes Hemsl. An analysis of basic parameters, such as drug concentrations, time, and behavior in different intestinal segments was analyzed by liquid chromatography-tandem mass spectrometry (LC-MS). The susceptibility of CAPB and CAPC to various inhibitors such as P-glycoprotein (P-gp) inhibitor (verapamil); multidrug resistance-associated protein 2 (MRP2) inhibitor (indomethacin); cytochrome P450 protein 3A4 (CYP3A4) inhibitor (ketoconazole); and the co-inhibitor of P-gp, MRP2 and CYP3A4 (cyclosporine A) were assessed using both caco-2 cell monolayer and single-pass intestinal perfusion (SPIP) models. As a result, CAPB and CAPC are both poorly absorbed in the intestines and exhibited segment-dependent permeability. The intestinal permeability of CAPB and CAPC were significantly increased by the co-treatment of verapamil, indomethacin. In addition, the intestinal permeability of CAPB was also enhanced by ketoconazole and cyclosporine A. It can be concluded that the intestinal absorption mechanisms of CAPB and CAPC involve processes such as facilitated passive diffusion, efflux transporters, and enzyme-mediated metabolism. Both CAPB and CAPC are suggested to be substrates of P-gp and MRP2. However, CAPB may interact with the CYP3A4 system.
Collapse
Affiliation(s)
- Xu Zhang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou, Zhejiang 313000, China.
| | - Yali Wu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Di Feng
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Yifan Qian
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Liping Chen
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Bo Yang
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| | - Mancang Gu
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| |
Collapse
|
29
|
Xu R, Yuan Y, Qi J, Zhou J, Guo X, Zhang J, Zhan R. Elucidation of the Intestinal Absorption Mechanism of Loganin in the Human Intestinal Caco-2 Cell Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:8340563. [PMID: 30671130 PMCID: PMC6323428 DOI: 10.1155/2018/8340563] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/26/2018] [Accepted: 12/04/2018] [Indexed: 11/17/2022]
Abstract
Loganin, iridoid glycosides, is the main bioactive ingredients in the plant Strychnos nux-vomica L. and demonstrates various pharmacological effects, though poor oral bioavailability in rats. In this study, the intestinal absorption mechanism of loganin was investigated using the human intestinal Caco-2 cell monolayer model in both the apical-to-basolateral (A-B) and the basolateral-to-apical (B-A) direction; additionally, transport characteristics were systematically investigated at different concentrations, pHs, temperatures, and potential transporters. The absorption permeability (PappAB) of loganin, which ranged from 12.17 to 14.78 × 10-6cm/s, was high at four tested concentrations (5, 20, 40, and 80μM), while the major permeation mechanism of loganin was found to be passive diffusion with active efflux mediated by multidrug resistance-associated protein (MRP) and breast cancer resistance protein (BCRP). In addition, it was found that loganin was not the substrate of efflux transporter P-glycoprotein (P-gp) since the selective inhibitor (verapamil) of the efflux transporter exhibited little effects on the transport of loganin in the human intestinal Caco-2 cells. Meanwhile, transport from the apical to the basolateral side increased 2.09-fold after addition of a MRP inhibitor and 2.32-fold after addition of a BCRP inhibitor. In summary, our results clearly demonstrate, for the first time, a good permeability of loganin in the human intestinal Caco-2 cell model and elucidate, in detail, the intestinal absorption mechanism and the effects of transporters on iridoid glycosides compounds.
Collapse
Affiliation(s)
- Renjie Xu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai200092, China
| | - Yichu Yuan
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai200127, China
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai200092, China
| | - Jia Zhou
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai200092, China
| | - Xiaowen Guo
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai200092, China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai200092, China
| | - Ruanjuan Zhan
- Department of Pharmacy, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou325035, China
| |
Collapse
|
30
|
Antioxidant Mechanisms of Echinatin and Licochalcone A. Molecules 2018; 24:molecules24010003. [PMID: 30577443 PMCID: PMC6337356 DOI: 10.3390/molecules24010003] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/15/2023] Open
Abstract
Echinatin and its 1,1-dimethyl-2-propenyl derivative licochalcone A are two chalcones found in the Chinese herbal medicine Gancao. First, their antioxidant mechanisms were investigated using four sets of colorimetric measurements in this study. Three sets were performed in aqueous solution, namely Cu2+-reduction, Fe3+-reduction, and 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide radical (PTIO•)-scavenging measurements, while 1,1-diphenyl-2-picrylhydrazyl radical (DPPH•)-scavenging colorimetric measurements were conducted in methanol solution. The four sets of measurements showed that the radical-scavenging (or metal-reduction) percentages for both echinatin and licochalcone A increased dose-dependently. However, echinatin always gave higher IC50 values than licochalcone A. Further, each product of the reactions of the chalcones with DPPH• was determined using electrospray ionization quadrupole time-of-flight tandem mass spectrometry (UPLC-ESI-Q-TOF-MS/MS). The UPLC-ESI-Q-TOF-MS/MS determination for echinatin yielded several echinatin–DPPH adduct peaks (m/z 662, 226, and 196) and dimeric echinatin peaks (m/z 538, 417, and 297). Similarly, that for licochalcone A yielded licochalcone A-DPPH adduct peaks (m/z 730, 226, and 196) and dimeric licochalcone A peaks (m/z 674 and 553). Finally, the above experimental data were analyzed using mass spectrometry data analysis techniques, resonance theory, and ionization constant calculations. It was concluded that, (i) in aqueous solution, both echinatin and licochalcone A may undergo an electron transfer (ET) and a proton transfer (PT) to cause the antioxidant action. In addition, (ii) in alcoholic solution, hydrogen atom transfer (HAT) antioxidant mechanisms may also occur for both. HAT may preferably occur at the 4-OH, rather than the 4′-OH. Accordingly, the oxygen at the 4-position participates in radical adduct formation (RAF). Lastly, (iii) the 1,1-dimethyl-2-propenyl substituent improves the antioxidant action in both aqueous and alcoholic solutions.
Collapse
|
31
|
Gunasekara DB, Speer J, Wang Y, Nguyen DL, Reed MI, Smiddy NM, Parker JS, Fallon JK, Smith PC, Sims CE, Magness ST, Allbritton NL. A Monolayer of Primary Colonic Epithelium Generated on a Scaffold with a Gradient of Stiffness for Drug Transport Studies. Anal Chem 2018; 90:13331-13340. [PMID: 30350627 PMCID: PMC6339567 DOI: 10.1021/acs.analchem.8b02845] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Animal models are frequently used for in vitro physiologic and drug transport studies of the colon, but there exists significant pressure to improve assay throughput as well as to achieve tighter control of experimental variables than can be achieved with animals. Thus, development of a primary in vitro colonic epithelium cultured as high resistance with transport protein expression and functional behavior similar to that of a native colonic would be of enormous value for pharmaceutical research. A collagen scaffold, in which the degree of collagen cross-linking was present as a gradient, was developed to support the proliferation of primary colonic cells. The gradient of cross-linking created a gradient in stiffness across the scaffold, enabling the scaffold to resist deformation by cells. mRNA expression and quantitative proteomic mass spectrometry of cells growing on these surfaces as a monolayer suggested that the transporters present were similar to those in vivo. Confluent monolayers acted as a barrier to small molecules so that drug transport studies were readily performed. Transport function was evaluated using atenolol (a substrate for passive paracellular transport), propranolol (a substrate for passive transcellular transport), rhodamine 123 (Rh123, a substrate for P-glycoprotein), and riboflavin (a substrate for solute carrier transporters). Atenolol was poorly transported with an apparent permeability ( Papp) of <5 × 10-7 cm s-1, while propranolol demonstrated a Papp of 9.69 × 10-6 cm s-1. Rh123 was transported in a luminal direction ( Papp,efflux/ Papp,influx = 7) and was blocked by verapamil, a known inhibitor of P-glycoprotein. Riboflavin was transported in a basal direction, and saturation of the transporter was observed at high riboflavin concentrations as occurs in vivo. It is anticipated that this platform of primary colonic epithelium will find utility in drug development and physiological studies, since the tissue possesses high integrity and active transporters and metabolism similar to that in vivo.
Collapse
Affiliation(s)
- Dulan B. Gunasekara
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
| | - Jennifer Speer
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Daniel L. Nguyen
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Mark I. Reed
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Nicole M. Smiddy
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Joel S. Parker
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC 27514, USA
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Christopher E. Sims
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
| | - Scott T. Magness
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
| | - Nancy L. Allbritton
- Department of Chemistry, University of North Carolina at Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, NC 27599, USA and North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
32
|
Liu T, Zhang X, Zhang Y, Hou J, Fang D, Sun H, Li Q, Xie S. Sulfation disposition of liquiritigenin in SULT1A3 overexpressing HEK293 cells: The role of breast cancer resistance protein (BCRP) and multidrug resistance-associated protein 4 (MRP4) in sulfate efflux of liquiritigenin. Eur J Pharm Sci 2018; 124:228-239. [PMID: 30176366 DOI: 10.1016/j.ejps.2018.08.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/30/2018] [Accepted: 08/28/2018] [Indexed: 12/27/2022]
Abstract
This study aimed to investigate the cellular disposition of liquiritigenin via the sulfonation pathway and the role of efflux transporters in liquiritigenin sulfate excretion. The sulfonation disposition of liquiritigenin was investigated using SULT1A3 overexpressed HEK293 cells (HEK-SULT1A3 cells). Liquiritigenin generated one mono-sulfate metabolite (7-O-sulfate) in HEK-SULT1A3 cell lysate. And the sulfonation followed the Michaelis-Menten kinetic (Vmax = 0.84 nmol/min/mg and Km = 7.12 μM). Expectedly, recombinant SULT1A3 (hSULT1A3) showed a highly similar kinetic profile with cell lysate. Furthermore, 7-O-sulfate was rapidly generated and excreted in HEK-SULT1A3 cells. Ko143 (a BCRP-selective inhibitor) at 20 μM significantly decreased the excretion rate of liquiritigenin sulfate (>42.5%, p < 0.001). Moreover, the pan-MRPs inhibitor MK-571 at 20 μM essentially abolished the liquiritigenin sulfate effluxion, resulting in the marked reduction of excretion rate (>97.4%, p < 0.001). Furthermore, knockdown of BCRP led to moderate reduction in sulfate excretion (15.9%-16.9%, p < 0.05). Silencing of MRP4 caused significant decreased in sulfate excretion (20.2%-32.5%, p < 0.01). In conclusion, one sulfate metabolite was generated from liquiritigenin in HEK-SULT1A3 cells. BCRP and MRP4 should be the key factors for the cellular excretion of liquiritigenin sulfate.
Collapse
Affiliation(s)
- Tong Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Xiaojing Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Yidan Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Jiuzhou Hou
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Hua Sun
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Qin Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Songqiang Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| |
Collapse
|