1
|
Hu Q, Tang X, Long R, Pan X, Shi S, Liu J, Pan Y, Li L, Gong L, Liao W, Zheng P, Luo X, Wang Q, Luo M, Fu C, Li R, Xiao H. Self-assembled nano delivery system of fenugreek polysaccharides: Effects on curcumin bioavailability and molecular mechanisms. Int J Biol Macromol 2025; 286:138294. [PMID: 39631596 DOI: 10.1016/j.ijbiomac.2024.138294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/23/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Self-assembly of fenugreek polysaccharides FS60 (a natural macromolecular material) with curcuminoid has been proved to improve curcumin (Cur) water dispersion in preliminary studies. This study further explored the effect of FS60 on Cur bioavailability in vivo to assess the significance of this delivery method. In this study, we optimized the formulation parameters of FS60-curcuminoid aggregates (FC) and studied their effects on Cur pharmacokinetics in rats. Results showed that the optimized aggregates had an encapsulation efficiency (EE) of 88.22 % and hydrodynamic diameter (DH) of 231.48 nm. Additionally, administering FC significantly increased curcumin glucuronide (Cur-O-Glu) levels. The Cmax was 51 times higher and AUC0-12h was 19 times higher than curcuminoid alone. Moreover, FS60 intervention for seven days increased the absorption speed of Cur-O-Glu into the bloodstream. Further mechanistic studies indicated that FS60 promoted Cur ingestion, increased UGT expression, and inhibited enterocyte transporters, allowing large amounts of Cur-O-Glu to enter the bloodstream. Moreover, the gut microbiota modulated by FS60 accelerated the mutual conversion of pentose and gluconate to provide sufficient glucuronic acid for the glucuronidation of Cur in enterocytes. Consequently, the nano delivery system composed by FS60 and curcuminoid facilitated gastrointestinal Cur glucuronidation and Cur-O-Glu absorption.
Collapse
Affiliation(s)
- Qiongdan Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Xinxing Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Rui Long
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Xiaoqi Pan
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Jia Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Yuan Pan
- Innovative Institute of Chinese Medicine and Pharmay, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Ling Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Leiqiang Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Wan Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China
| | - Ping Zheng
- Chengdu Institute for Drug Control, NMPA Key Laboratory for Quality Monitoring and Evaluation of Traditional Chinese Medicine, Chengdu 610045, PR China
| | - Xiao Luo
- Chengdu Institute for Drug Control, NMPA Key Laboratory for Quality Monitoring and Evaluation of Traditional Chinese Medicine, Chengdu 610045, PR China
| | - Qi Wang
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
| | - Minna Luo
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China.
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, PR China; Sichuan Jinhong Keyou Biotechnology Co., Ltd, PR China.
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, United States.
| |
Collapse
|
2
|
Smith DJ, Bi H, Hamman J, Ma X, Mitchell C, Nyirenda K, Monera-Penduka T, Oketch-Rabah H, Paine MF, Pettit S, Pheiffer W, Van Breemen RB, Embry M. Potential pharmacokinetic interactions with concurrent use of herbal medicines and a ritonavir-boosted COVID-19 protease inhibitor in low and middle-income countries. Front Pharmacol 2023; 14:1210579. [PMID: 37502215 PMCID: PMC10368978 DOI: 10.3389/fphar.2023.1210579] [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: 04/22/2023] [Accepted: 06/30/2023] [Indexed: 07/29/2023] Open
Abstract
The COVID-19 pandemic sparked the development of novel anti-viral drugs that have shown to be effective in reducing both fatality and hospitalization rates in patients with elevated risk for COVID-19 related morbidity or mortality. Currently, nirmatrelvir/ritonavir (Paxlovid™) fixed-dose combination is recommended by the World Health Organization for treatment of COVID-19. The ritonavir component is an inhibitor of cytochrome P450 (CYP) 3A, which is used in this combination to achieve needed therapeutic concentrations of nirmatrelvir. Because of the critical pharmacokinetic effect of this mechanism of action for Paxlovid™, co-administration with needed medications that inhibit or induce CYP3A is contraindicated, reflecting concern for interactions with the potential to alter the efficacy or safety of co-administered drugs that are also metabolized by CYP3A. Some herbal medicines are known to interact with drug metabolizing enzymes and transporters, including but not limited to inhibition or induction of CYP3A and P-glycoprotein. As access to these COVID-19 medications has increased in low- and middle-income countries (LMICs), understanding the potential for herb-drug interactions within these regions is important. Many studies have evaluated the utility of herbal medicines for COVID-19 treatments, yet information on potential herb-drug interactions involving Paxlovid™, specifically with herbal medicines commonly used in LMICs, is lacking. This review presents data on regionally-relevant herbal medicine use (particularly those promoted as treatments for COVID-19) and mechanism of action data on herbal medicines to highlight the potential for herbal medicine interaction Herb-drug interaction mediated by ritonavir-boosted antiviral protease inhibitors This work highlights potential areas for future experimental studies and data collection, identifies herbal medicines for inclusion in future listings of regionally diverse potential HDIs and underscores areas for LMIC-focused provider-patient communication. This overview is presented to support governments and health protection entities as they prepare for an increase of availability and use of Paxlovid™.
Collapse
Affiliation(s)
- Dallas J. Smith
- Epidemic Intelligence Service, Centers for Disease Control and Prevention, Atlanta, GA, United States
- COVID-19 Response International Task Force, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Huichang Bi
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Josias Hamman
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Constance Mitchell
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Kumbukani Nyirenda
- Department of Pharmacy, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Tsitsi Monera-Penduka
- Research Unit for Safety of Herbs and Drugs, University of Zimbabwe, Harare, Zimbabwe
| | | | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Syril Pettit
- Health and Environmental Sciences Institute, Washington, DC, United States
| | - Wihan Pheiffer
- DSI/NWU Preclinical Drug Development Platform, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Richard B. Van Breemen
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Michelle Embry
- Health and Environmental Sciences Institute, Washington, DC, United States
| |
Collapse
|
3
|
Ji X, Yang M, Shen G, Or KH, Yim WS, Zuo Z. Safety evaluations of the processed lateral root of Aconitum carmichaelii Debx. And its hepatotoxicity mechanisms in rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115801. [PMID: 36216199 DOI: 10.1016/j.jep.2022.115801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The processed lateral root of Aconitum carmichaelii Debx. is known as Fuzi, an extensively used Traditional Chinese Medicine to treat cardiovascular diseases, rheumatism arthritis, bronchitis, pains, and hypothyroidism, etc. Although Chinese Pharmacopeia regulates the safe clinical dosage of Fuzi at 3-15 g/person/day, such recommendation not only lacks bench evidence but also does not differentiate Fuzi with different processing types, such as Heishunpian and Paofupian. AIM OF THE STUDY The current study aimed to 1) determine No-Observed-Adverse-Effect-Levels of Heishunpian and Paofupian in rats and 2) investigate the related toxicity mechanisms for their safe clinical use. MATERIALS AND METHODS After giving clinically relevant dosing regimen of Heishunpian/Paofupian to rats, we conducted toxicity assessments including ECG monitoring, histopathological changes and serum biomarkers to detect organ injury. Metabolomic study in the liver revealed changes in endogenous metabolite levels after two-week treatment of Fuzi preparations or its corresponding six toxic alkaloids mixtures. RESULTS The NOAEL for both bolus and two-week treatments of Heishunpian and Paofupian in rats was designated to be 7.5 g/kg and 15 g/kg, respectively. Corresponding recommended doses in humans were 7.5-25 g/person/day for Heishunpian and 15-50 g/person/day for Paofupian. Metabolic profiles revealed more significant alterations in endogenous substances from rats receiving the two Fuzi preparations than their corresponding toxic alkaloids mixtures. Upregulation of bile acid pathway could be responsible for Fuzi induced liver injury. CONCLUSIONS Compared to the current maximum recommended dose, our suggested upper limit of guided dose for Heishunpian was comparable, whereas that for Paofupian could be further elevated. Both C19-diterpenoid alkaloids and co-occurring components in Fuzi preparations contributed to their hepatotoxicity via upregulation of bile acid pathway.
Collapse
Affiliation(s)
- Xiaoyu Ji
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Mengbi Yang
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong SAR, PR China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China
| | - Guolin Shen
- Institute of Chemicals Safety, Chinese Academy of Inspection and Quarantine, Beijing, PR China
| | - Ka Hang Or
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Wan Sze Yim
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Zhong Zuo
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong SAR, PR China.
| |
Collapse
|
4
|
Wang L, Liu Q, Hu X, Zhou C, Ma Y, Wang X, Tang Y, Chen K, Wang X, Liu Y. Enhanced Oral Absorption and Liver Distribution of Polymeric Nanoparticles through Traveling the Enterohepatic Circulation Pathways of Bile Acid. ACS APPLIED MATERIALS & INTERFACES 2022; 14:41712-41725. [PMID: 36069201 DOI: 10.1021/acsami.2c10322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The intestinal epithelium is known to be a main hindrance to oral delivery of nanoparticles. Even though surface ligand modification can enhance cellular uptake of nanoparticles, the "easy entry and hard across" was frequently observed for many active targeting nanoparticles. Here, we fabricated polymeric nanoparticles relayed by bile acid transporters with monomethoxy poly(ethylene glycol)-poly(D,l-lactide) and deoxycholic acid-conjugated poly(2-ethyl-2-oxazoline)-poly(D,l-lactide) based on structural characteristics of intestine epithelium and the absorption characteristics of endogenous substances. As anticipated, deoxycholic acid-modified polymeric nanoparticles featuring good stability in simulated gastrointestinal fluid could notably promote the internalization of their payload by Caco-2 cells through mediation of apical sodium-dependent bile acid transporter (ASBT) and transmembrane transport of the nanoparticles across Caco-2 cell monolayers via relay-guide of ASBT, ileal bile acid-binding protein, and the heteromeric organic solute transporter (OSTα-OSTβ) along with multidrug resistance-associated protein 3 (MRP3) evidenced by competitive inhibition and fluorescence immunoassay, which was further visually confirmed by the stronger fluorescence from C6-labeled nanoparticles inside enterocytes and the basal side of the intestinal epithelium of mice. The transcellular transport of deoxycholic acid-modified nanoparticles in an intact form was mediated by caveolin/lipid rafts and clathrin with intracellular trafficking trace of endosome-lysosome-ER-Golgi apparatus and bile acid transport route. Furthermore, the increased uptake by HepG2 cells compared with unmodified nanoparticles evidenced the target ability of deoxycholic acid-modified nanoparticles to the liver, which was further supported by ex vivo imaging of excised major organs of mice. Thus, this study provided a feasible and potential strategy to further enhance transepithelial transport efficiency and liver-targeted ability of nanoparticles by means of the specific enterohepatic circulation pathways of bile acid.
Collapse
Affiliation(s)
- Leqi Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qi Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinping Hu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chuhang Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yining Ma
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiaoxiao Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yingwei Tang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Kanghao Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xinyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
5
|
Bolger GT, Pucaj K, Minta YO, Sordillo P. Relationship Between the In Vitro Efficacy, Pharmacokinetics and In Vivo Efficacy of Curcumin. Biochem Pharmacol 2022; 205:115251. [PMID: 36130650 DOI: 10.1016/j.bcp.2022.115251] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/19/2022]
Abstract
Considerable interest continues to be focused on the development of curcumin either as an effective stand-alone therapeutic or as an adjunct therapy to established therapies. Curcumin (1, 7-bis (4-hydroxy-3-methoxyphenyl)-1, 6-heptadiene-3, 5- dione; also called diferuloylmethane) is a polyphenolic phytochemical extracted from the root of curcuma longa, commonly called turmeric. Despite evidence from in vitro (cell culture) and preclinical studies in animals, clinical studies have not provided strong evidence for a therapeutic effect of curcumin. The relevance of curcumin as a drug has been questioned based on its classification as a compound with pan assay interference and invalid metabolic panaceas properties bringing into question the relevance of the therapeutic targets identified for curcumin. To some extent this is due to the lack of a complete understanding of the link between the in vitro (cell culture activity), pharmacokinetics and in vivo activity of curcumin. In this review and using NF-κB as a cellular target for curcumin, we have investigated the relationship between the potency of curcumin as an inhibitor of NF-κB in cell culture, the pharmacokinetics of curcumin and curcumin's anticancer and anti-inflammatory effects in preclinical models of cancer and inflammation. Plausible explanations and rationale are provided to link these activities together and suggest that both curcumin and its more soluble Phase II metabolite curcumin glucuronide may play a key role in the treatment effects of curcumin in vivo mediated at NF-κB.
Collapse
Affiliation(s)
| | | | - Yvonne O Minta
- Nucro-Technics, Department of Toxicology, Toronto, ON, Canada
| | | |
Collapse
|
6
|
Islam T, Koboziev I, Albracht-Schulte K, Mistretta B, Scoggin S, Yosofvand M, Moussa H, Zabet-Moghaddam M, Ramalingam L, Gunaratne PH, Moustaid-Moussa N. Curcumin Reduces Adipose Tissue Inflammation and Alters Gut Microbiota in Diet-Induced Obese Male Mice. Mol Nutr Food Res 2021; 65:e2100274. [PMID: 34510720 DOI: 10.1002/mnfr.202100274] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 08/04/2021] [Indexed: 12/24/2022]
Abstract
SCOPE Obesity prevalence continues to increase and contribute to metabolic diseases, potentially by driving systemic inflammation. Curcumin is an anti-inflammatory spice with claimed health benefits. However, mechanisms by which curcumin may reduce obesity-associated inflammation are poorly understood; thus, it is hypothesized that benefits of curcumin consumption may occur through reduced white adipose tissue (WAT) inflammation and/or beneficial changes in gut bacteria. METHODS AND RESULTS Male B6 mice are fed high-fat diets (HFD, 45% kcal fat) or HFD supplemented with 0.4% (w/w) curcumin (HFC) for 14 weeks. Curcumin supplementation significantly reduces adiposity and total macrophage infiltration in WAT, compared to HFD group, consistent with reduced mRNA levels of M1 (Cd80, Cd38, Cd11c) and M2 (Arginase-1) macrophage markers. Moreover, curcumin supplementation reduces expression of other key pro-inflammatory genes, such as NF-κB p65 subunit (p65), Stat1, Tlr4, and Il6, in WAT (p < 0.05). Using microbial 16S RNA sequencing, it is demonstrated that the relative abundance of the Lactococcus, Parasutterella, and Turicibacter genera are increased in the HFC group versus HFD. CONCLUSIONS Curcumin exerts protective metabolic effects in dietary obesity, in part through downregulation of adipose tissue inflammation, which may be mediated by alterations in composition of gut microbiota, and metabolism of curcumin into curcumin-O-glucuronide.
Collapse
Affiliation(s)
- Tariful Islam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Iurii Koboziev
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Kembra Albracht-Schulte
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Brandon Mistretta
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Mohammad Yosofvand
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.,Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Hanna Moussa
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.,Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Masoud Zabet-Moghaddam
- Obesity Research Institute, Texas Tech University, Lubbock, TX, USA.,Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA.,Obesity Research Institute, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
7
|
Wang JQ, Yang Y, Cai CY, Teng QX, Cui Q, Lin J, Assaraf YG, Chen ZS. Multidrug resistance proteins (MRPs): Structure, function and the overcoming of cancer multidrug resistance. Drug Resist Updat 2021; 54:100743. [PMID: 33513557 DOI: 10.1016/j.drup.2021.100743] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/21/2020] [Accepted: 12/25/2020] [Indexed: 02/08/2023]
Abstract
ATP-binding cassette (ABC) transporters mediate the ATP-driven translocation of structurally and mechanistically distinct substrates against steep concentration gradients. Among the seven human ABC subfamilies namely ABCA-ABCG, ABCC is the largest subfamily with 13 members. In this respect, 9 of the ABCC members are termed "multidrug resistance proteins" (MRPs1-9) due to their ability to mediate cancer multidrug resistance (MDR) by extruding various chemotherapeutic agents or their metabolites from tumor cells. Furthermore, MRPs are also responsible for the ATP-driven efflux of physiologically important organic anions such as leukotriene C4, folic acid, bile acids and cAMP. Thus, MRPs are involved in important regulatory pathways. Blocking the anticancer drug efflux function of MRPs has shown promising results in overcoming cancer MDR. As a result, many novel MRP modulators have been developed in the past decade. In the current review, we summarize the structure, tissue distribution, biological and pharmacological functions as well as clinical insights of MRPs. Furthermore, recent updates in MRP modulators and their therapeutic applications in clinical trials are also discussed.
Collapse
Affiliation(s)
- Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chao-Yun Cai
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA; School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong 511436, China; Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Jun Lin
- Department of Anesthesiology, Stony Brook University Health Sciences Center, Stony Brook, NY, 11794, USA
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
8
|
Ji JZ, Li YF, Jiang LP, Tai T, Ge PX, Mi QY, Zhu T, Xie HG. P-glycoprotein deficiency enhances metabolic activation of and platelet response to clopidogrel through marked up-regulation of Cyp3a11 in mice: Direct evidence for the interplay between P-glycoprotein and Cyp3a. Biochem Pharmacol 2020; 183:114313. [PMID: 33137324 DOI: 10.1016/j.bcp.2020.114313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
Variability in P-glycoprotein (P-gp) efflux transporting activity was supposed to be involved in altered intestinal absorption and bioavailability of clopidogrel in patients; however, reliable evidence is still lacking. In this study, we sought to determine whether P-gp could play an important role in the metabolic activation of and platelet response to clopidogrel in mice. Abcb1a/1b knock-out (KO) and wild-type (WT) mice were used to evaluate differences in the intracellular accumulation of clopidogrel in the intestine, liver, and brain tissues and in systemic exposure of clopidogrel and its main metabolites as well as the mechanisms involved. Results indicated that, compared with WT mice, KO mice exhibited an 84% increase in systemic exposure of clopidogrel active thiol metabolite H4 and a 14.5% rise of suppression of ADP-induced platelet integrin αIIbβ3 activation, paralleled by a 41% decrease in systemic exposure of clopidogrel due to enhanced systemic clearance. Furthermore, KO mice displayed a 45% increase in Cyp3a11 but a 23% decrease in Ces1 at their protein levels compared with WT mice. Concurrently, intracellular clopidogrel concentrations in the tissues examined did not differ significantly between KO and WT mice. We conclude that although P-gp does not transport clopidogrel and its major metabolites in mice, P-gp-deficient mice exhibit elevated formation of the active metabolite H4 and enhanced antiplatelet effect of clopidogrel through up-regulation of Cyp3a11 and down-regulation of Ces1, suggesting that P-gp activity may correlate inversely with the formation of H4 and antiplatelet efficacy of clopidogrel in clinical settings due to P-gp and CYP3A4 interplay.
Collapse
Affiliation(s)
- Jin-Zi Ji
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Yi-Fei Li
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Li-Ping Jiang
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ting Tai
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Peng-Xin Ge
- Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qiong-Yu Mi
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Ting Zhu
- Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hong-Guang Xie
- Division of Clinical Pharmacology, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China; Department of Clinical Pharmacy, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Department of Clinical Pharmacy, Nanjing Medical University School of Pharmacy, Nanjing 211166, China.
| |
Collapse
|
9
|
Paine MF. Natural Products: Experimental Approaches to Elucidate Disposition Mechanisms and Predict Pharmacokinetic Drug Interactions. Drug Metab Dispos 2020; 48:956-962. [PMID: 32816868 PMCID: PMC7543467 DOI: 10.1124/dmd.120.000182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Natural products have been used by humans since antiquity for both egregious and beneficial purposes. Regarding the latter, these products have long been valued as a rich source of phytochemicals and developed into numerous life-saving pharmaceutical agents. Today, the sales and use of natural products with purported medicinal qualities continue to increase worldwide. However, natural products are not subject to the same premarket testing requirements as pharmaceutical agents, creating critical gaps in scientific knowledge about their optimal use. In addition, due to the common misperception that "natural" means "safe," patients may supplement or replace their prescription medications with natural products, placing themselves at undue risk for subefficacious pharmacotherapy or potentially toxic exposure. Collectively, with few exceptions, researchers, health care providers, and educators lack definitive information about how to inform consumers, patients, and students in the health professions on the safe and optimal use of these products. Recognition of this deficiency by key stakeholders, including the three pillars of biomedical research-industry, academia, and government-has facilitated multiple collaborations that are actively addressing this fundamental knowledge gap. This special issue contains a collection of articles highlighting the challenges faced by researchers in the field and the use of various experimental systems and methods to improve the mechanistic understanding of the disposition and drug interaction potential of natural products. Continued refinement of existing, and development of new, approaches will progress toward the common overarching goal of improving public health. SIGNIFICANCE STATEMENT: Natural products with purported medicinal value constitute an increasing share of the contemporary health care market. Natural products are not subject to the same premarket testing requirements as drug products, creating fundamental scientific knowledge gaps about the safe and effective use of these products. Collaborations among industrial, academic, and governmental researchers in multiple disciplines are anticipated to provide the definitive information needed to fill these gaps and improve public health.
Collapse
Affiliation(s)
- Mary F Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|