Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Colloid Interface Sci ; 650(Pt A): 526-540, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37423180

ABSTRACT

Tumor microenvironment (TME) stimuli-responsive nanoassemblies are emerging as promising drug delivery systems (DDSs), which acquire controlled release by structural transformation under exogenous stimulation. However, the design of smart stimuli-responsive nanoplatforms integrated with nanomaterials to achieve complete tumor ablation remains challenging. Therefore, it is of utmost importance to develop TME-based stimuli-responsive DDSs to enhance drug-targeted delivery and release at tumor sites. Herein, we proposed an appealing strategy to construct fluorescence-mediated TME stimulus-responsive nanoplatforms for synergistic cancer therapy by assembling photosensitizers (PSs) carbon dots (CDs), chemotherapeutic agent ursolic acid (UA), and copper ions (Cu2+). First, UA nanoparticles (UA NPs) were prepared by self-assembly of UA, then UA NPs were assembled with CDs via hydrogen bonding force to obtain UC NPs. After combining with Cu2+, the resulting particles (named UCCu2+ NPs) exhibited quenched fluorescence and photosensitization due to the aggregation of UC NPs. Upon entering the tumor tissue, the photodynamic therapy (PDT) and the fluorescence function of UCCu2+ were recovered in response to TME stimulation. The introduction of Cu2+ triggered the charge reversal of UCCu2+ NPs, thereby promoting lysosomal escape. Furthermore, Cu2+ resulted in additional chemodynamic therapy (CDT) capacity by reacting with hydrogen peroxide (H2O2) as well as by consuming glutathione (GSH) in cancer cells through a redox reaction, hence magnifying intracellular oxidative stress and enhancing the therapeutic efficacy due to reactive oxygen species (ROS) therapy. In summary, UCCu2+ NPs provided an unprecedented novel approach for improving the therapeutic efficacy through the three-pronged (chemotherapy, phototherapy, and heat-reinforced CDT) attacks to achieve synergistic therapy.


Subject(s)
Biological Products , Carcinoma, Hepatocellular , Liver Neoplasms , Nanoparticles , Neoplasms , Humans , Copper/chemistry , Carcinoma, Hepatocellular/drug therapy , Hydrogen Peroxide , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Cell Line, Tumor , Neoplasms/drug therapy , Glutathione , Tumor Microenvironment
2.
Biomater Sci ; 10(21): 6267-6281, 2022 Oct 25.
Article in English | MEDLINE | ID: mdl-36128848

ABSTRACT

Conventional treatments for cancer, such as chemotherapy, surgical resection, and radiotherapy, have shown limited therapeutic efficacy, with severe side effects, lack of targeting and drug resistance for monotherapies, which limit their clinical application. Therefore, combinatorial strategies have been widely investigated in the battle against cancer. Herein, we fabricated a dual-targeted nanoscale drug delivery system based on EpCAM aptamer- and lactic acid-modified low-polyamidoamine dendrimers to co-deliver the FDA-approved agent disulfiram and photosensitizer indocyanine green, combining the imaging and therapeutic functions in a single platform. The multifunctional nanoparticles with uniform size had high drug-loading payload, sustained release, as well as excellent photothermal conversion. The integrated nanoplatform showed a superior synergistic effect in vitro and possessed precise spatial delivery to HepG2 cells with the dual-targeting nanocarrier. Intriguingly, a robust anticancer response of chemo-phototherapy was achieved; chemotherapy combined with the efficacy of phototherapy to cause cellular apoptosis of HepG2 cells (>35%) and inhibit the regrowth of damaged cells. Furthermore, the theranostic nanosystem displayed fluorescence imaging in vivo, attributed to its splendid accumulation in the tumor site, and it provided exceptional tumor inhibition rate against liver cancer cells (>76%). Overall, our research presents a promising multifunctional theranostic nanoplatform for the development of synergistic therapeutics for tumors in further applications.


Subject(s)
Dendrimers , Hyperthermia, Induced , Nanoparticles , Neoplasms , Humans , Indocyanine Green/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Epithelial Cell Adhesion Molecule , Doxorubicin/pharmacology , Delayed-Action Preparations , Precision Medicine , Disulfiram , Drug Delivery Systems/methods , Neoplasms/therapy , Lactic Acid , Hyperthermia, Induced/methods , Drug Liberation , Theranostic Nanomedicine/methods , Cell Line, Tumor
3.
Am J Chin Med ; 50(3): 749-772, 2022.
Article in English | MEDLINE | ID: mdl-35450513

ABSTRACT

The endothelium covers the internal lumen of the entire circulatory system and plays an important modulatory role in vascular homeostasis. Endothelium dysfunction, characterized by a vasoconstrictive, pro-inflammatory, and pro-coagulant state, usually manifests as a significant pathological process of vascular diseases, including hypertension, atherosclerosis (AS), stroke, diabetes mellitus, coronary artery disease, and cancer. Therefore, there is an urgent necessity to seek promising therapeutic drugs or remedies to ameliorate endothelial dysfunction-induced vascular ailments and complications. Recently, much attention has been attached to ginsenosides, the most significant active components of ginseng, which have always been referred to as "all-healing" and widely used for its extensively medicinal value. Surprisingly, ginsenosides have diverse biological activity which might be related to inflammation, apoptosis, oxidative stress, and angiogenesis. In this review, a brief introduction about endothelial dysfunction and ginsenosides was demonstrated, and the emphasis was put on summarizing multi-faceted pharmacological effects and underlying molecular mechanisms of ginsenosides on the endothelium, including vasorelaxation, anti-oxidation, anti-inflammation, and angio-modulation. Beyond that, nanotechnology to improve efficacy and the existing clinical trials of ginsenosides were concluded. Hopefully, our work will give suggestions for promoting clinical application of traditional Chinese medicine, e.g., hypertension, AS, diabetes, ischemic stroke, and cancer. This review provides a comprehensive base of knowledge for ginsenosides to prevention and treatment of vascular injury- related diseases with clinical significance.


Subject(s)
Ginsenosides , Hypertension , Neoplasms , Panax , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Humans , Hypertension/drug therapy , Neoplasms/drug therapy , Pharmaceutical Preparations
4.
Colloids Surf B Biointerfaces ; 209(Pt 2): 112177, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34749194

ABSTRACT

Rapamycin (RAPA) functions as effectively clinical immunosuppressive agent, its significant tumor growth suppression effect via various pathways in diverse cancers, especially combined with photothermal therapy, is gaining a burgeoning attention. However, its critical defects, low solubility and poor stability, have severely hampered its further application. Herein, RAPA, indocyanine green (ICG) and epigallocatechin gallate (EGCG) serving as chemotherapeutic drug, photosensitizer and biomimetic coatings, respectively, were co-assembled into carrier-free, high biocompatible ICG-RAPA-EGCG nanoparticles (IRE NPs) for synergistic cancer therapy. Particularly, the bioinspired EGCG coatings not only improved the stability of IRE NPs under physiological conditions to avert NPs disassembly and drug release, but also maintained the photostability of ICG to achieve excellent photothermal response. The results indicated that the as-prepared IRE NPs displayed good monodispersity and enhanced stability at various stored media after introducing of EGCG. Compared with monotherapy of RAPA or ICG, IRE NPs showed higher dose-dependent toxicity in MCF-7 cells, HepG2 cells and HeLa cells, especially plus near-infrared laser irradiation. Furthermore, IRE NPs exhibited quicker uptake in cells, higher accumulation in tumor region (even in 48 h) than free ICG and effectively inhibited tumor growth without side effect in H22 tumor-bearing mice. Collectively, the carrier-free IRE NPs provided a simply alternative approach to fabricate RAPA/photosensitizer co-loaded nanoparticles for combinatorial tumor therapy.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Animals , Biomimetics , Cell Line, Tumor , HeLa Cells , Humans , Indocyanine Green , Mice , Photosensitizing Agents , Phototherapy , Photothermal Therapy , Polyphenols , TOR Serine-Threonine Kinases
5.
ACS Appl Mater Interfaces ; 12(51): 57362-57372, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33301289

ABSTRACT

The rapid development of CRISPR/Cas9 systems has opened up tantalizing prospects to sensitize cancers to chemotherapy using efficient targeted genome editing, but safety concerns and possible off-target effects of viral vectors remain a major obstacle for clinical application. Thus, the construction of novel nonviral tumor-targeting nanodelivery systems has great potential for the safe application of CRISPR/Cas9 systems for gene-chemo-combination therapy. Here, we report a polyamidoamine-aptamer-coated hollow mesoporous silica nanoparticle for the co-delivery of sorafenib and CRISPR/Cas9. The core-shell nanoparticles had good stability, enabled ultrahigh drug loading, targeted delivery, and controlled-release of the gene-drug combination. The nanocomplex showed >60% EGFR-editing efficiency without off-target effects in all nine similar sites, regulating the EGFR-PI3K-Akt pathway to inhibit angiogenesis, and exhibited a synergistic effect on cell proliferation. Importantly, the co-delivery nanosystem achieved efficient EGFR gene therapy and caused 85% tumor inhibition in a mouse model. Furthermore, the nanocomplex showed high accumulation at the tumor site in vivo and exhibited good safety with no damage to major organs. Due to these properties, the nanocomplex provides a versatile delivery approach for efficient co-loading of gene-drug combinations, allowing for precise gene editing and synergistic inhibition of tumor growth without apparent side effects on normal tissues.


Subject(s)
CRISPR-Cas Systems , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Sorafenib/therapeutic use , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , CRISPR-Associated Protein 9/genetics , Cell Line, Tumor , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Liberation , Epithelial Cell Adhesion Molecule/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Editing , Genes, erbB-1 , Humans , Mice , Nanoparticles/toxicity , Polyamines/chemistry , Polyamines/toxicity , Porosity , Signal Transduction/drug effects , Silicon Dioxide/toxicity
6.
Pharmacol Res ; 159: 105031, 2020 09.
Article in English | MEDLINE | ID: mdl-32562816

ABSTRACT

Thrombosis initiated by abnormal platelet aggregation is a pivotal pathological event that precedes most cases of cardiovascular diseases (CVD). Recently, growing evidence indicates that platelet could be a potential target for CVD prevention. However, as the conventional antithrombotic management strategy, applications of current antiplatelet agents are somewhat limited by their various side effects, such as bleeding risk and drug resistance. Hence, efforts have been made to search for agents as complementary therapies. Ginsenoside, the principal active component extracted from Panax ginseng, has gained much attention for its regulations on multiple crucial events of platelet aggregation. From structural characteristics to clinical applications, this review anatomized the intrinsic structure-function relationship of antiplatelet potency of ginsenosides, and the involved signal pathways were specifically summarized. Additionally, the emphasis was placed on clinical studies that investigate the antithrombotic efficacy of ginsenosides in the treatment of CVD. Further, a broad overview of approaches for improving the bioavailability of ginsenosides was concluded. Limitations and prospects of current studies were also discussed. This study may provide some new insights into the systematic understanding of ginsenosides in CVD treatment and lay a foundation for future research.


Subject(s)
Blood Platelets/drug effects , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Ginsenosides/therapeutic use , Muscle, Smooth, Vascular/drug effects , Neointima , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Vascular Remodeling/drug effects , Animals , Biological Availability , Blood Platelets/metabolism , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Ginsenosides/adverse effects , Ginsenosides/pharmacokinetics , Humans , Molecular Structure , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Signal Transduction , Structure-Activity Relationship
7.
Eur J Pharm Sci ; 142: 105100, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31669385

ABSTRACT

Warfarin and ginseng have been widely used in the treatment of cardiovascular diseases. However, the clinical safety and effectiveness of herb-drug combination treatment are still controversial. Therefore, it is very essential to probe the interaction between warfarin and ginseng. In this study, in vitro and in vivo study was carried out to demonstrate that whether there is an interaction between warfarin and ginsenosides (GS), which is the main component of ginseng. In vitro study showed that the adhesion ability between endothelial cells and matrigel/platelets was enhanced due to the up-regulating expression of intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1) proteins by treatment of warfarin+GS combination compared to warfarin/GS treatment alone. Moreover, GS could weaken the anticoagulation effect of warfarin in hyperlipemia rats owning to the increased expression levels of coagulation factors and hepatic cytochrome P450 enzymes in plasma after long-term co-administration of warfarin with GS. The results of both in vitro and in vivo study demonstrated that there is a serious interaction between warfarin and ginseng, which may deteriorate atherosclerosis and thrombosis after combined use of warfarin and GS.


Subject(s)
Anticoagulants/pharmacology , Cardiovascular Diseases/drug therapy , Ginsenosides/pharmacology , Herb-Drug Interactions/physiology , Warfarin/pharmacology , Animals , Blood Coagulation/drug effects , Cardiovascular Diseases/metabolism , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , Liver/drug effects , Liver/metabolism , Panax/chemistry , Plant Extracts/pharmacology , Rats , Thrombosis/drug therapy , Thrombosis/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
8.
Int J Pharm ; 570: 118663, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31493497

ABSTRACT

Nanoformulations with advantages in drug delivery, safety and pharmacodynamics have been booming as a promising strategy for cancer therapy. However, the traditional nanocarrier still suffers from the low drug loading capacity, potential systematic toxicity, unclear metabolism, and other uncertainties. To overcome these issues, carrier-free nanodrugs with desirable bioactivity were developed rapidly and drawn considerable attention. Meanwhile, the multifunctional self-delivery nanoarcheticture fabricated by a simple and "green" method, has significant advantages in synergistic cancer therapy and inhibition of multidrug resistant (MDR). Till now, carrier-free nanoparticles for tumor theranostics, phototherapy, chemotherapy, diagnose and synergistic therapy, have made outstanding progress. In this review, we make an integrated and exhaustive overview of lately reports on carrier-free nanodrug delivery systems formed by several active agents. We summarize the self-assembly and modified strategies, with emphasis on application superiority of carrier-free nanocrystal, and give new insight into the establishment of ideal nanosystems for cancer treatment.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Drug Delivery Systems/methods , Drug Resistance, Multiple/drug effects , Humans
9.
Acta Biomater ; 70: 197-210, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29408311

ABSTRACT

Nanosized drug delivery systems (NDDS) with photothermal therapy (PTT) and photodynamic therapy (PDT) have been extensively exploited to improve the therapeutic performance and bio-safety of chemotherapeutic drugs in cancer. In this work, a carrier-free nanodrug was developed by co-assembly of the anti-cancer agent ursolic acid (UA), an asialoglycoprotein receptor (ASGPR), which can recognize the target molecule lactobionic acid (LA), and the near-infrared (NIR) probe dye indocyanine green (ICG) to form UA-LA-ICG NPs by a simple and green self-assembly approach. The UA-LA-ICG NPs had suitable stability, showed controlled release profile of UA drugs, and exhibited preferable temperature response (∼59.4 °C) under laser irradiation (808 nm, 1 W/cm2). Compared with free ICG, the UA-LA-ICG NPs significantly enhanced the intracellular ICG uptake. Upon irradiation of the NIR laser, co-assembled nanodrugs demonstrated great performance as a reactive oxygen species (ROS) producer and exhibited more anti-proliferative activities on ASGPR-overexpressing HepG2 cells than ASGPR low-expressing HeLa cells. Meanwhile, in vivo NIR fluorescence imaging exhibited that the co-assembled nanodrugs were specifically targeted to the tumor by the active targeting property of LA, and its circulation time was much longer than that of free ICG. In addition, UA-LA-ICG NPs + NIR irradiation treatment displayed enhanced inhibitory effect on tumor growth in H22 tumor-bearing mice. Overall, the co-assembly of chemotherapeutic agent and photosensitizer by the self-assembly approach might open an alternative avenue and give inspiration to fabricate new carrier-free nanodrugs for cancer imaging and chemo-photo combination therapy. STATEMENT OF SIGNIFICANCE: The present study for the first time reported carrier-free nanoparticles (NPs) by co-assembly of a natural product ursolic acid (UA), an asialoglycoprotein receptor (ASGPR)-recognized sugar molecule lactobionic acid (LA), and the near-infrared dye indocyanine green (ICG) through a simple and green approach. The preparation process of nanodrugs is simple, rapid, effective, and labor-saving. The co-assembled nanodrugs were capable of stabilizing the ICG molecules and specifically targeting to the tumor, which could increase the tumor accumulation in cancer imaging and also enhance the efficacy of chemo-phototherapy.


Subject(s)
Hyperthermia, Induced , Neoplasms, Experimental , Optical Imaging , Photochemotherapy , Triterpenes/pharmacology , Animals , Asialoglycoprotein Receptor/metabolism , HeLa Cells , Hep G2 Cells , Humans , Male , Mice , Neoplasm Proteins/metabolism , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Reactive Oxygen Species/metabolism , Triterpenes/pharmacokinetics , Xenograft Model Antitumor Assays , Ursolic Acid
10.
J Ethnopharmacol ; 171: 154-60, 2015 Aug 02.
Article in English | MEDLINE | ID: mdl-26055342

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Isoboldine is one of the major bioactive constituents in the total alkaloids from Radix Linderae (TARL) which could effectively alleviate inflammation and joints destruction in mouse collagen-induced arthritis. To better understand its pharmacological activities, we need to determine its pharmacokinetic and metabolic profiles. MATERIALS AND METHODS: In this study, a sensitive and simple UPLC-MS/MS method was developed and validated for determination of isoboldine in rat plasma. Isoboldine in plasma was recovered by liquid-liquid extraction using 1 mL of methyl tert-butyl ether. Chromatographic separation was performed on a C18 column at 45°C, with a gradient elution consisting of acetonitrile and water containing 0.1% (v/v) formic acid at a flow rate of 0.3 mL/min. The detection was performed on an electrospray triple-quadrupole MS/MS by positive ion multiple-reaction monitoring mode. This newly developed method was successfully applied to a pharmacokinetic study after oral and intravenous dosing in rats. For metabolites identification, isoboldine was orally administered to rats and the metabolite in plasma, bile, urine and feces were characterized by the established UPLC-MS/MS method. RESULTS: Good linearity (r(2)>0.9956) was achieved in a concentration range of 4.8-2400 ng/mL with a lower limit of quantification of 4.8 ng/mL for isoboldine. The intra- and inter-day precisions of the assay were 1.7-5.1% and 2.2-4.4% relative standard deviation with an accuracy of 91.3-102.3%. A total of five phase II metabolites in rat plasma, bile, urine and feces were characterized by comparing retention time in UPLC, and by molecular mass and fragmentation pattern of the metabolites by mass spectrometry with those of isoboldine. CONCLUSION: isoboldine has extremely low oral bioavailability due to the strong first-pass effect by the rats, and glucuronidation and sulfonation were involved in metabolic pathways of isoboldine in rats. These results have paved the way for further clarifying therapeutic ingredients and provided new knowledge regarding pharmacokinetic features of this category of isoquinoline alkaloids.


Subject(s)
Alkaloids/pharmacokinetics , Lindera , Plant Roots , Alkaloids/blood , Alkaloids/urine , Animals , Bile/chemistry , Chromatography, High Pressure Liquid , Feces/chemistry , Male , Rats, Sprague-Dawley , Tandem Mass Spectrometry
11.
Zhongguo Zhong Yao Za Zhi ; 33(9): 1014-7, 1089, 2008 May.
Article in Chinese | MEDLINE | ID: mdl-18652346

ABSTRACT

OBJECTIVE: To screen a group of traditional Chinese medicines with effect on pregnane X receptor (PXR)-mediated transcription regulation of P450 3A4 (CYP3A4); and to study whether they can induce the expression of CYP3A4 with a dose, time-dependent manner. METHOD: Transient cotransfection reporter gene assays were performed with pCI-hPXR-neo, pGL3-CYP3A4-Luc and beta-galactosidase expression plasmid in HepG2 cells. RESULT: Rhizoma Curcumae, Atractylodes lancea, A. macrocaphala and Poria cocos could induce transcriptional expression of CYP3A4. In the dose-effect study, 24 h after induction, 500 mg x L(-1) Rhizoma Curcumae, A. lancea, A. macrocaphala and Poria cocos, respectively, could induce the CYP3A4 gene expression with (6.82 +/- 0.09), (6.76 +/- 0.20), (5.49 +/- 0.13) and (4.97 +/- 0.07) folds, as compared with 0.1% DMSO treated cells. In the time-effect study, 500 mg x L(-1) Rhizoma curcumae, A. lancea, A. macrocaphala and Poria cocos for 48 h could induce the CYP3A4 gene expression with (7.74 +/- 0.54), (7.34 +/- 0.10), (5.54 +/- 0.11) and (5.32 +/- 0.18) folds, compared with 0.1% DMSO treated cells. CONCLUSION: Rhizoma Curcumae, A. lancea, A. macrocaphala and Poria cocos could induce the expression of CYP3A4 gene transcription through activating PXR.


Subject(s)
Cytochrome P-450 CYP3A/genetics , Drugs, Chinese Herbal/pharmacology , Transcription, Genetic/drug effects , Cell Line, Tumor , Humans , Pregnane X Receptor , Receptors, Steroid/metabolism
12.
Zhong Yao Cai ; 31(1): 68-71, 2008 Jan.
Article in Chinese | MEDLINE | ID: mdl-18589752

ABSTRACT

OBJECTIVE: To study the effects of four kinds of extracts from anti-cancer Taditional Chinese Medicines on the activity and mRNA expression of CYP3A in rat's liver. METHODS: Rat's liver microsomal cytochrome P450 and CYP3A isoemzymes--erythromycin N-demethylase(ERD) activities were determined by UV chromatography, the mRNA expression levels of CYP3Al and CYP3A2 were detected by reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: Extracts of Rhizoma Curcumae, Rhizoma Atractylodes and Rhizoma Atractylodes Macrocaphalae markedly increased the P450 content of liver microsomes and induced the enzyme activity of CYP3A, but extract of Poria cocos did not. At the mRNA level, extracts of Rhizoma Curcumae, Rhizoma Atractylodes Lanceae and Rhizoma Atractylodes Macraphalae induced the expression of CYP3A1, but extract of Poria cocos did not. The expression of CYP3A2 were induced by extracts of Rhizoma Curcumae and Rhizoma Atractylodes Lanceae, but extracts of Rhizoma Atractylodes Macrocaphalae and Poria cocos were found of remarkable inhibition of the mRNA expression of CYP3A2. CONCLUSION: Extracts of Rhizoma Curcumae, Rhizoma Atractylodes Lanceae and Rhizoma Atractylodes Macrocaphalae can regulate CYP3A on the levels of enzyme activity and mRNA expression, but Poria cocos extract only regulated CYP3A on the level of mRNA expression, possibly by affecting the metabolism of other drugs in the body.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cytochrome P-450 CYP3A/genetics , Drugs, Chinese Herbal/pharmacology , Liver/drug effects , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cytochrome P-450 Enzyme System/genetics , Drugs, Chinese Herbal/isolation & purification , Enzyme Induction/drug effects , Liver/enzymology , Liver/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Plants, Medicinal/chemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL