1
|
Xu Y, Li J, Cui M, Li X, Zhai H, Wu D, Chu X. Therapeutic effects of medicinal and food-based traditional herbal couples on type 2 diabetes mellitus based on pharmacodynamics and pharmacokinetics. Front Pharmacol 2025; 16:1560271. [PMID: 40351422 PMCID: PMC12062019 DOI: 10.3389/fphar.2025.1560271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/14/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Cinnamomi Ramulus (CR) is the dried bark of Cinnamomum cassia Presl, Lauraceae. Puerariae Lobatae Radix (PLR) is the dried root of the Pueraria lobata (Wild.) Ohwi, Leguminosae. This Chinese herb couple come from the classic formula "Gui Zhi Ge Gen Tang," which is included in the TCM classic "Treatise on Febrile Diseases." Our previous studies have found that CR related herbal compound and PLR related herbal compound are useful in improving type 2 diabetes mellitus (T2DM), which is expected to be an antidiabetic candidate with fewer side effects. However the mechanism of action of CR-PLR on T2DM has not yet been fully elucidated. Methods The decoction of CR-PLR was prepared by aqueous extraction method and the composition of it was analyzed using UPLC-Q-TOF-MS and HPLC. The T2DM model was established by intraperitoneal injection of streptozotocin, and the groups of drug administration were metformin, CR, PLR and CR-PLR groups, with continuous gastric gavage for 6 weeks, and the serological indexes were detected by ELISA. The abundance of rats' gut flora was detected by 16s rDNA sequencing, and changes in the content of short-chain fatty acids (SCFAs) in feces of rats were detected by GC-MS; and the expression of G protein-coupled receptor43 (GPR43) and glucagon-like peptide-1 (GLP-1) proteins in colonic tissues of rats were detected by Western Blot. The pharmacokinetic behavior of CR-PLR was investigated in both normal and T2DM model rats. Caco-2/HT29 co-culture cell model was established in vitro, transepithelial electrical resistance (TEER) and ALP activity of epithelial cells were measured to evaluate cell model integrity and cell polarization, Alcian blue staining was used to verify the presence of mucus production, and CCK-8 was used to screen drug safe concentration. The bidirectional transport of puerarin was studied to investigate the transport mechanism of puerarin and the effect of leuric acid on puerarin transport. Results and discussion The results indicated that CR-PLR can stimulate intestinal flora, increase the content of SCFAs, activate intestinal GPR43 protein, and promote the secretion of GLP-1 in intestinal L cells, which plays a therapeutic role in the treatment of T2DM. Additionally, cytology and pharmacokinetics experiments have proved that cinnamic acid (CA) can enhance the absorption and transport of puerarin (PUR) by inhibiting the efflux effects mediated by P-gp and MRP efflux transporters. The present study exhibites the scientific and reasonable menaning of this novel Chinese herb couple treating T2DM from the perspecives of pharmacodynamics and pharmacokinetics.
Collapse
Affiliation(s)
- Yuhang Xu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengyao Cui
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xiaoliang Li
- Anhui Joyfar Pharmaceutical Research Institute Co., Ltd., Hefei, China
| | - Hongyan Zhai
- Anhui Province Institute for Food and Drug Control, Hefei, China
| | - Deling Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Bozhou University, Bozhou, China
- Anhui Provincial Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui, China
| |
Collapse
|
2
|
Kocsis AE, Kucsápszky N, Santa-Maria AR, Hunyadi A, Deli MA, Walter FR. Much More than Nutrients: The Protective Effects of Nutraceuticals on the Blood-Brain Barrier in Diseases. Nutrients 2025; 17:766. [PMID: 40077636 PMCID: PMC11901837 DOI: 10.3390/nu17050766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The dysfunction of the blood-brain barrier (BBB) is well described in several diseases, and is considered a pathological factor in many neurological disorders. This review summarizes the most important groups of natural compounds, including alkaloids, flavonoids, anthocyanidines, carotenoids, lipids, and vitamins that were investigated for their potential protective effects on brain endothelium. The brain penetration of these compounds and their interaction with BBB efflux transporters and solute carriers are discussed. The cerebrovascular endothelium is considered a therapeutic target for natural compounds in diseases. In preclinical studies modeling systemic and central nervous system diseases, nutraceuticals exerted beneficial effects on the BBB. In vivo, they decreased BBB permeability, brain edema, astrocyte swelling, and morphological changes in the vessel structure and basal lamina. At the level of brain endothelial cells, nutraceuticals increased cell survival and decreased apoptosis. From the general endothelial functions, decreased angiogenesis and increased levels of vasodilating agents were demonstrated. From the BBB functions, elevated barrier integrity by tightened intercellular junctions, and increased expression and activity of BBB transporters, such as efflux pumps, solute carriers, and metabolic enzymes, were shown. Nutraceuticals enhanced the antioxidative defense and exerted anti-inflammatory effects at the BBB. The most important signaling changes mediating the increased cell survival and BBB stability were the activation of the WNT, PI3K-AKT, and NRF2 pathways, and inhibition of the MAPK, JNK, ERK, and NF-κB pathways. Nutraceuticals represent a valuable source of new potentially therapeutic molecules to treat brain diseases by protecting the BBB.
Collapse
Affiliation(s)
- Anna E. Kocsis
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Nóra Kucsápszky
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Ana Raquel Santa-Maria
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary;
- Interdisciplinary Centre of Natural Products, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös u. 6, H-6720 Szeged, Hungary
- Graduate Institute of Natural Products, Kaohsiung Medical University, Shih-Chuan 1st Rd. 100, Kaohsiung 807, Taiwan
| | - Mária A. Deli
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (A.E.K.); (N.K.)
| |
Collapse
|
3
|
Li S, Liu Y. Intestinal absorption mechanism and nutritional synergy promotion strategy of dietary flavonoids: transintestinal epithelial pathway mediated by intestinal transport proteins. Crit Rev Food Sci Nutr 2024:1-14. [PMID: 39086266 DOI: 10.1080/10408398.2024.2387320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Dietary flavonoids exhibit a variety of physiological functions in regulating glucose and lipid metabolism, improving cardiovascular function, and enhancing stress resistance. However, poor intestinal absorption limits their health benefits. Previous studies on improving the absorption efficiency of flavonoids have focused on targeted release, enhanced gastrointestinal stability and prolonged retention time in digestive tract. But less attention has been paid to promoting the uptake and transport of flavonoids by intestinal epithelial cells through modulation of transporter protein-mediated pathways. Interestingly, some dietary nutrients have been found to modulate the expression or function of transporter proteins, thereby synergistically or antagonistically affecting flavonoid absorption. Therefore, this paper proposed an innovative regulatory strategy known as the "intestinal transport protein-mediated pathway" to promote intestinal absorption of dietary flavonoids. The flavonoid absorption mechanism in the intestinal epithelium, mediated by intestinal transport proteins, was summarized. The functional differences between the uptake transporter and efflux transporters during flavonoid trans-intestinal cellular transport were discussed. Finally, from the perspective of nutritional synergy promotion of absorption, the feasibility of promoting flavonoid intestinal absorption by regulating the expression/function of transport proteins through dietary nutrients was emphasized. This review provides a new perspective and developing precise dietary nutrient combinations for efficient dietary flavonoid absorption.
Collapse
Affiliation(s)
- Shuqiong Li
- College of Ocean Food and Biological Engineering, National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian, People's Republic of China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, National & Local Joint Engineering Research Center of Deep Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian, People's Republic of China
| |
Collapse
|
4
|
Jiang L, Zhang K, Fan M, Pan W, Qian H, Wang L, Li Y. Quercetin Enhances the Availability of 5-Heptadecylresorcinol by Inhibiting the Expression of P-gp. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18375-18384. [PMID: 37962857 DOI: 10.1021/acs.jafc.3c05518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
5-Heptadecylresorcinol (AR-C17), as the most important active monomer, is found in large quantities in wheat and triticale and plays a variety of health benefits, such as antidiabetic, anti-inflammatory, and antitumor. However, the low bioavailability of AR-C17 due to its low water solubility restricts its application. Moreover, the transport mechanism of AR-C17 is not fully understood. Here, we showed that the transport of AR-C17 in vitro was time- and concentration-dependent, and relatively higher temperature and lower pH obviously promoted the transport of AR-C17. Besides, transporters, especially P-glycoprotein (P-gp), markedly affected the transport of AR-C17 as well. Quercetin, a natural synergist in triticale bran (TB), was confirmed as an inhibitor of P-gp to promote the transport of AR-C17 in this study, and the bioavailability of AR-C17 reached the highest when the concentration ratio of quercetin to AR-C17 was 1:1.
Collapse
Affiliation(s)
- Lei Jiang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Kuiliang Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Mingcong Fan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Wei Pan
- Center for Information of National Medical Products Administration, 8 Sanli River, Beijing 100820, China
| | - Haifeng Qian
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| |
Collapse
|
5
|
Zhang M, Su Y, Li J, Chang C, Gu L, Yang Y. Fabrication of phosphatidylcholine-EGCG nanoparticles with sustained release in simulated gastrointestinal digestion and their transcellular permeability in a Caco-2 monolayer model. Food Chem 2023; 437:137580. [PMID: 39491254 DOI: 10.1016/j.foodchem.2023.137580] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/24/2023] [Accepted: 09/21/2023] [Indexed: 11/05/2024]
Abstract
In this study, we prepared phosphatidylcholine (PC)-EGCG complex nanoparticles (P-E NPs) by solvent reflux method. The physicochemical properties, in vitro digestion, uptake in Caco-2 cells, and bidirectional permeability of P-E NPs were systematically investigated. The constructed P-E1.5:1 NPs had an average particle size of 118 nm, a ζ-potential of -37.8 mV, and a polymerization dispersion index (PDI) of 0.16. The encapsulation efficiency (EE) of EGCG was 85.0% and the loading capacity (LC) was 24.4%. UV spectra, FTIR, XRD and intermolecular force results indicated that hydrophobic, electrostatic and hydrogen bonding interactions contributed to formate P-E1.5:1 NPs. P-E1.5:1 NPs exhibited first-order kinetics sustained release properties in simulated gastrointestinal digestion. Furthermore, P-E1.5:1 NPs were able to enhance absorptive transport and inhibit efflux transport mediated by MRP2 and P-gp compared to EGCG. These results indicated that P-E1.5:1 NPs may be a potential strategy to ameliorate EGCG bioavailability.
Collapse
Affiliation(s)
- Mengya Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yujie Su
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Junhua Li
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Cuihua Chang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Luping Gu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yanjun Yang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
6
|
Hodges JK, Sasaki GY, Vodovotz Y, Bruno RS. Gallation and B-Ring Dihydroxylation Increase Green Tea Catechin Residence Time in Plasma by Differentially Affecting Tissue-Specific Trafficking: Compartmental Model of Catechin Kinetics in Healthy Adults. Nutrients 2023; 15:4021. [PMID: 37764804 PMCID: PMC10536004 DOI: 10.3390/nu15184021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Catechins in green tea extract (GTE) (epigallocatechin gallate (EGCG), epigallocatechin (EGC), epicatechin (EC), epicatechin gallate (ECG)) vary in bioactivity. We developed a physiologically relevant mathematical model of catechin metabolism to test the hypothesis that fractional catabolic rates of catechins would be differentially affected by their structural attributes. Pharmacokinetic data of plasma and urine catechin concentrations were used from healthy adults (n = 19) who ingested confections containing 0.5 g GTE (290 mg EGCG, 87 mg EGC, 39 mg EC, 28 mg ECG). A 7-compartmental model of catechin metabolism comprised of the gastrointestinal tract (stomach, small and large intestine), liver, plasma, extravascular tissues, and kidneys was developed using a mean fraction dose of EGCG, ECG, EGC, and EC. Fitting was by iterative least squares regression analysis, and goodness of fit was ascertained by the estimated variability of parameters (FSD < 0.5). The interaction of gallation and B-ring dihydroxylation most greatly extended plasma residence time such that EGC > EC = EGCG > EGC. The interaction between gallation and B-ring dihydroxylation accelerated the transfer from the upper gastrointestinal tract to the small intestine but delayed subsequent transfers from the small intestine through the liver to plasma and from kidneys to urine. Gallation and B-ring dihydroxylation independently delayed the transfer from plasma to extravascular tissues, except the uptake to kidneys, which was slowed by gallation only. This multi-compartment model, to be validated in a future study, suggests that gallation and B-ring dihydroxylation affect catechin catabolism in a tissue-specific manner and thus their potential bioactivity.
Collapse
Affiliation(s)
- Joanna K Hodges
- Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Geoffrey Y Sasaki
- Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yael Vodovotz
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Rani A, Saini V, Patra P, Prashar T, Pandey RK, Mishra A, Jha HC. Epigallocatechin Gallate: A Multifaceted Molecule for Neurological Disorders and Neurotropic Viral Infections. ACS Chem Neurosci 2023; 14:2968-2980. [PMID: 37590965 DOI: 10.1021/acschemneuro.3c00368] [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] [Indexed: 08/19/2023] Open
Abstract
Epigallocatechin-3-gallate (EGCG), a polyphenolic moiety found in green tea extracts, exhibits pleiotropic bioactivities to combat many diseases including neurological ailments. These neurological diseases include Alzheimer's disease, multiple sclerosis, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. For instance, in the case of Alzheimer's disease, the formation of a β-sheet in the region of the 10th-21st amino acids was significantly reduced in EGCG-induced oligomeric samples of Aβ40. Its interference induces the formation of Aβ structures with an increase in intercenter-of-mass distances, reduction in interchain/intrachain contacts, reduction in β-sheet propensity, and increase in α-helix. Besides, numerous neurotropic viruses are known to instigate or aggravate neurological ailments. It exerts an effect on the oxidative damage caused in neurodegenerative disorders by acting on GSK3-β, PI3K/Akt, and downstream signaling pathways via caspase-3 and cytochrome-c. EGCG also diminishes these viral-mediated effects, such as EGCG delayed HSV-1 infection by blocking the entry for virions, inhibitory effects on NS3/4A protease or NS5B polymerase of HCV and potent inhibitor of ZIKV NS2B-NS3pro/NS3 serine protease (NS3-SP). It showed a reduction in the neurotoxic properties of HIV-gp120 and Tat in the presence of IFN-γ. EGCG also involves numerous viral-mediated inflammatory cascades, such as JAK/STAT. Nonetheless, it also inhibits the Epstein-Barr virus replication protein (Zta and Rta). Moreover, it also impedes certain viruses (influenza A and B strains) by hijacking the endosomal and lysosomal compartments. Therefore, the current article aims to describe the importance of EGCG in numerous neurological diseases and its inhibitory effect against neurotropic viruses.
Collapse
Affiliation(s)
- Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Vaishali Saini
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Priyanka Patra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| | - Tanish Prashar
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Karwar, 342030, Jodhpur India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, 453552, Indore India
| |
Collapse
|
8
|
Zhang S, Mao B, Cui S, Zhang Q, Zhao J, Tang X, Chen W. Absorption, metabolism, bioactivity, and biotransformation of epigallocatechin gallate. Crit Rev Food Sci Nutr 2023; 64:6546-6566. [PMID: 36728926 DOI: 10.1080/10408398.2023.2170972] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Epigallocatechin gallate (EGCG), a typical flavone-3-ol polyphenol containing eight free hydroxyl groups, is associated with a variety of bioactivities, such as antioxidant, anti-inflammatory, anti-cancer, and antibacterial activities. However, the poor bioavailability of EGCG restricts its use. In this review, we discuss the processes involved in the absorption and metabolism of EGCG, with a focus on its metabolic interactions with the gut microbiota. Next, we summarize the bioactivities of some key metabolites, describe the biotransformation of EGCG by different microorganisms, and discuss its catabolism by specific bacteria. A deeper understanding of the absorption, metabolism, and biotransformation of EGCG may enable its disease-preventive and therapeutic properties to be better utilized. This review provides a theoretical basis for further development and utilization of EGCG and its metabolites for improving the gut microbiota and physiological health.
Collapse
Affiliation(s)
- Shuling Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
9
|
Feng D, Zhong G, Zuo Q, Wan Y, Xu W, He C, Lin C, Huang D, Chen F, Huang L. Knockout of ABC transporters by CRISPR/Cas9 contributes to reliable and accurate transporter substrate identification for drug discovery. Front Pharmacol 2022; 13:1015940. [PMID: 36386127 PMCID: PMC9649518 DOI: 10.3389/fphar.2022.1015940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/13/2022] [Indexed: 04/21/2024] Open
Abstract
It is essential to explore the relationship between drugs and transporters in the process of drug development. Strong background signals in nonhuman MDCK or LLC-PK1 cells and overlapping interference of inhibitors or RNAi in human Caco-2 cells mean that an ideal alternative could be to knock out specific transporter genes in Caco-2 cells. However, the application of gene knockout (KO) to Caco-2 cells is challenging because it is still inefficient to obtain rapidly growing Caco-2 subclones with double-allele KO through long-term monoclonal cultivation. Herein, CRISPR/Cas9, a low cost but more efficient and precise gene editing technology, was utilized to singly or doubly knockout the P-gp, BCRP, and MRP2 genes in Caco-2 cells. By combining this with single cell expansion, rapidly growing transporter-deficient subclones were successfully screened and established. Bidirectional transport assays with probe substrates and three protease inhibitors indicated that more reliable and detailed data could be drawn easily with these KO Caco-2 models. The six robust KO Caco-2 subclones could contribute to efficient in vitro drug transport research.
Collapse
Affiliation(s)
- Dongyan Feng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Guorui Zhong
- Bioinformatics Institute, Agency for Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Qingxia Zuo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Yanbin Wan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Wanqing Xu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Changsheng He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Cailing Lin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Dongchao Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Feng Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lizhen Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
10
|
Yu Z, Ma Y, Guan Y, Zhu Y, Wang K, Wang Y, Liu P, Chen J, Yu Y. Metagenomics of Virus Diversities in Solid-State Brewing Process of Traditional Chinese Vinegar. Foods 2022; 11:3296. [PMID: 37431044 PMCID: PMC9602057 DOI: 10.3390/foods11203296] [Citation(s) in RCA: 8] [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: 09/29/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/24/2022] Open
Abstract
Traditional Chinese vinegar offers an exceptional flavor and rich nutrients due to its unique solid-state fermentation process, which is a multiple microbial fermentation system including various bacteria, fungi and viruses. However, few studies on the virus diversities in traditional Chinese vinegar have been reported. In this paper, using Zhenjiang aromatic vinegar as a model system, we systemically explored the viral communities in the solid-state brewing process of traditional Chinese vinegar using bacterial and viral metagenomes. Results showed that the viral diversity in vinegar Pei was extensive and the virus communities varied along with the fermentation process. In addition, there existed some interactions between viral and bacterial communities. Moreover, abundant antibiotic resistance genes were found in viromes, indicating that viruses might protect fermentation bacteria strains from the stress of antibiotics in the fermentation environment. Remarkably, we identified abundant auxiliary carbohydrate metabolic genes (including alcohol oxidases, the key enzymes for acetic acid synthesis) from viromes, implying that viruses might participate in the acetic acid synthesis progress of the host through auxiliary metabolic genes. Taken together, our results indicated the potential roles of viruses in the vinegar brewing process and provided a new perspective for studying the fermentation mechanisms of traditional Chinese vinegar.
Collapse
Affiliation(s)
- Zhen Yu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Yan Ma
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Yingfen Guan
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Yuanyuan Zhu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Ke Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Yuqin Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Peng Liu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Juan Chen
- College of Food Science and Engineering, Moutai Institute, Renhuai 564501, China
| | - Yongjian Yu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| |
Collapse
|
11
|
Pei S, Dou Y, Zhang W, Qi D, Li Y, Wang M, Li W, Shi H, Gao Z, Yao C, Fang D, Sun H, Xie S. O-Sulfation disposition of curcumin and quercetin in SULT1A3 overexpressing HEK293 cells: the role of arylsulfatase B in cellular O-sulfation regulated by transporters. Food Funct 2022; 13:10558-10573. [PMID: 36156668 DOI: 10.1039/d2fo01436j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Extensive phase II metabolic reactions (i.e., glucuronidation and sulfation) have resulted in low bioavailability and decreased biological effects of curcumin and quercetin. Compared to glucuronidation, information on the sulfation disposition of curcumin and quercetin is limited. In this study, we identified that BCRP and MRP4 played a critical role in the cellular excretion of curcumin-O-sulfate (C-O-S) and quercetin-O-sulfate (Q-O-S) by integrating chemical inhibition with transporter knock-down experiments. Inhibited excretion of sulfate (C-O-S and Q-O-S) caused significant reductions in cellular O-sulfation of curcumin (a maximal 74.4% reduction) and quercetin (a maximal 76.9% reduction), revealing a strong interplay of sulfation with efflux transport. It was further identified that arylsulfatase B (ARSB) played a crucial role in the regulation of cellular O-sulfation by transporters. ARSB overexpression significantly enhanced the reduction effect of MK-571 on the cellular O-sulfation (fmet) of the model compound (38.8% reduction for curcumin and 44.2% reduction for quercetin). On the contrary, ARSB knockdown could reverse the effect of MK-571 on the O-sulfation disposition of the model compound (29.7% increase for curcumin and 47.3% increase for quercetin). Taken together, ARSB has been proven to be involved in cellular O-sulfation, accounting for transporter-dependent O-sulfation of curcumin and quercetin. A better understanding of the interplay beneath metabolism and transport will contribute to the exact prediction of in vivo drug disposition and drug-drug interactions.
Collapse
Affiliation(s)
- Shuhua Pei
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Yuanyuan Dou
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Wenke Zhang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Defei Qi
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Yingying Li
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Mengqing Wang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Wenqi Li
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Hongxiang Shi
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Zixuan Gao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Chaoyan Yao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Dong Fang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Hua Sun
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| | - Songqiang Xie
- Academy for advanced interdisciplinary studies, Henan University, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China. .,Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| |
Collapse
|