1
|
Zhou X, Xia X. Ginsenoside Rg3 improves microcystin-induced cardiotoxicity through the miR-128-3p/MDM4 axis. Drug Chem Toxicol 2024; 47:682-692. [PMID: 37990515 DOI: 10.1080/01480545.2023.2251716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 11/23/2023]
Abstract
Microcystin (MC) is the byproduct of cyanobacteria metabolism that is associated with oxidative stress and heart damage. This study aimed to investigate the effect of ginsenoside Rg3 on MC-induced cardiotoxicity. A mouse model of myocardial infarction was constructed by oral MC administration. H9C2 cells were used for in vitro analysis. Cellular oxidative stress, apoptosis, and the relationship between miR-128-3p and double minute 4 protein (MDM4) were analyzed. MiR-128-3p expression was upregulated in vitro and in vivo after MC treatment, which was downregulated after Rg3 treatment. Left ventricular ejection fraction (LVEF) and left ventricular systolic pressure (LVSP) were increased and left ventricular end-diastolic pressure (LVEDP) was decreased after Rg3 treatment. Moreover, Rg3 alleviated MC-induced pathological changes and apoptosis in myocardial tissues. Meanwhile, Rg3 treatment decreased the lactate dehydrogenase (LDH) and malondialdehyde (MDA) levels and inhabited cell apoptosis and oxidative stress in MC-treated myocardial cells. MiR-128-3p overexpression attenuated the protective effect of Rg3 on MC-induced cardiotoxicity. MiR-128-3p negatively regulated MDM4 expression. This study revealed that Rg3 alleviated MC-induced cardiotoxicity through the miR-128-3p/MDM4 axis, which emphasized the potential of Rg3 as a therapeutic agent for MC-induced cardiotoxicity, and miR-128-3p as a target for the Rg3 therapy.
Collapse
Affiliation(s)
- Xiaoming Zhou
- Department of Cardiovascular Medicine, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xiaoyan Xia
- Dean's Office, Changsha Health Vocational College, Changsha, Hunan, China
| |
Collapse
|
2
|
Han S, Zhang X, Li Z, Cui G, Xue B, Yu Y, Guo J, Zhang H, Yang J, Teng L. A ginsenoside G-Rg3 PEGylated long-circulating liposome for hyperglycemia and insulin resistance therapy in streptozotocin-induced type 2 diabetes mice. Eur J Pharm Biopharm 2024; 201:114350. [PMID: 38848783 DOI: 10.1016/j.ejpb.2024.114350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Ginsenoside (GS), one of the main active components in ginseng, can enhance insulin sensitivity, improve the function of islet β cells, and reduce cell apoptosis in the treatment of diabetes. However, the drawbacks of high lipid solubility, poor water solubility, and low oral availability in Ginsenoside Rg3 (G-Rg3) seriously limit further application of GS. In this work, a G-Rg3 PEGylated long-circulating liposome (PEG-L-Rg3) is designed and developed to improve symptoms in type 2 diabetic mice. The as-prepared PEG-L-Rg3 with a spherical structure shows a particle size of ∼ 140.5 ± 1.4 nm, the zeta potential of -0.10 ± 0.05 mV, and a high encapsulation rate of 99.8 %. Notably, in vivo experimental results demonstrate that PEG-L-Rg3 exhibits efficient ability to improve body weight and food intake in streptozotocin-induced type 2 diabetic mice. Moreover, PEG-L-Rg3 also enhances fasting insulin (FINS) and insulin sensitivity index (ISI). In addition, the glucose tolerance of mice is significantly improved after the treatment of PEG-L-Rg3, indicating that PEG-L-Rg3 can be a potential drug for the treatment of type 2 diabetes, which provides a new way for the treatment of type 2 diabetes using ginsenosides.
Collapse
Affiliation(s)
- Songren Han
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Xueyan Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Ziwei Li
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Guilin Cui
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Beilin Xue
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Yang Yu
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Jiaqing Guo
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China
| | - Huan Zhang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Jie Yang
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Lesheng Teng
- School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| |
Collapse
|
3
|
Zheng W, Huang Y, Wu Q, Cheng P, Song Y, Wang B, Huang Q, Hu S. Poly(lactic acid hydroxyacetic acid)-poly(ethylene glycol)-modified ginsenoside Rg3 nanomedicine enhances anti-tumor effect in hepatocellular carcinoma. Drug Dev Ind Pharm 2024; 50:763-775. [PMID: 39259031 DOI: 10.1080/03639045.2024.2402769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE This research aims to improve the bioavailability and anti-hepatocellular carcinoma (HCC) efficacy of Ginsenoside Rg3 by modification with poly (lactic acid hydroxyacetic acid)-poly(ethylene glycol) (PLGA-PEG). METHODS PLGA-PEG-Rg3 was obtained by emulsification and evaluated it physiochemical characterization by FTIR, SEM, laser particle-size analyzer and HPLC. The effect of the PLGA-PEG-Rg3 and Rg3 on HepG2 cells was compared in vitro studies, including cell proliferation, transwell and a series of apoptosis detection, and in-situ HCC model. RESULTS The PLGA-PEG-Rg3 were 122 nm in size and 0.112 in polydispersity index with sustained release profile in vitro. Compared to Rg3, PLGA-PEG-Rg3 was more effective in suppressing HepG2 growth and inducing apoptosis by the mitochondrial apoptosis pathway in vitro. And PLGA-PEG modification enhanced the liver-targeting ability and drug circulation time of Rg3 in vivo, resulting in PLGA-PEG-Rg3 possessing superior performance in inhibiting tumor growth and prolonging the survival time of tumor-bearing mice than Rg3. CONCLUSIONS Overall, these results showed PLGA-PEG-Rg3 enhanced the anti-tumor effect of Rg3 in HCC.
Collapse
Affiliation(s)
- Wei Zheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuqiao Huang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Qiong Wu
- Center for Modern Physics Technology, School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Pu Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yujun Song
- Center for Modern Physics Technology, School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, China
| | - Ben Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shen Hu
- Department of Obstetrics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, Hangzhou, China
| |
Collapse
|
4
|
Zhang XK, Wu Y, Long XN, You XX, Chen D, Bi Y, He S, Cao GH. Widely Targeted Metabolomic Analysis Reveals the Improvement in Panax notoginseng Triterpenoids Triggered by Arbuscular Mycorrhizal Fungi via UPLC-ESI-MS/MS. Molecules 2024; 29:3235. [PMID: 38999186 PMCID: PMC11243182 DOI: 10.3390/molecules29133235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Panax notoginseng is a highly valued perennial medicinal herb in China and is widely used in clinical treatments. The main purpose of this study was to elucidate the changes in the composition of P. notoginseng saponins (PNSs), which are the main bioactive substances, triggered by arbuscular mycorrhizal fungi (AMF) via ultrahigh-performance liquid chromatography-electrospray ionization-tandem mass spectrometry (UPLC-ESI-MS/MS). A total of 202 putative terpenoid metabolites were detected, of which 150 triterpene glycosides were identified, accounting for 74.26% of the total. Correlation analysis, principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) of the metabolites revealed that the samples treated with AMF (group Ce) could be clearly separated from the CK samples. In total, 49 differential terpene metabolites were identified between the Ce and CK groups, of which 38 and 11 metabolites were upregulated and downregulated, respectively, and most of the upregulated differentially abundant metabolites were mainly triterpene glycosides. The relative abundances of the two major notoginsenosides (MNs), ginsenosides Rd and Re, and 13 rare notoginsenosides (RNs), significantly increased. The differential saponins, especially RNs, were more easily clustered into one branch and had a high positive correlation. It could be concluded that the biosynthesis and accumulation of some RNs share the same pathways as those triggered by AMF. This study provides a new way to obtain more notoginsenoside resources, particularly RNs, and sheds new light on the scientization and rationalization of the use of AMF agents in the ecological planting of medicinal plants.
Collapse
Affiliation(s)
- Xing-Kai Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yue Wu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xian-Nv Long
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Xiao-Xu You
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Di Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yue Bi
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Sen He
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
- Kunming Lancang-Mekong Regional R&D Central for the Development Utilization of Traditional Medicine Resources, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Guan-Hua Cao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, China
- Kunming Lancang-Mekong Regional R&D Central for the Development Utilization of Traditional Medicine Resources, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
5
|
Wang X, Wei P, Hu C, Zeng H, Fan Z. 3D printing of Rg3-loaded hydrogel scaffolds: anti-inflammatory and scar-formation related collagen inhibitory effects for scar-free wound healing. J Mater Chem B 2024; 12:4673-4685. [PMID: 38647236 DOI: 10.1039/d3tb02941g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
During the process of wound healing, the stimulation of inflammatory factors often leads to abnormal proliferation of blood vessels and collagen, ultimately resulting in scar formation. To address this challenge, we fabricate a novel dermal extracellular matrix (DECM) hydrogel scaffold loaded with ginsenoside Rg3 (Rg3) using 3D printing technology. Mesoporous silica nanoparticles (MSNs) are introduced into the system to encase the Rg3 to control its release rate and enhance its bioavailability. We systematically evaluate the biological, physicochemical, and wound healing properties of this scaffold. In vitro studies demonstrate that the hydrogel exhibits excellent biocompatibility and solid-like rheological properties, ensuring its successful printing. In vivo studies reveal that the composite hydrogel scaffolds effectively accelerate wound healing and achieve scar-free wound healing within three weeks. Histological and immunohistochemical (IHC) analyses show that the composite hydrogel scaffolds reduce the inflammatory response and inhibit excessive collagen accumulation. These combined effects underscore the potential of our approach in effectively inhibiting scar formation.
Collapse
Affiliation(s)
- Xusen Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Pengyu Wei
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Cewen Hu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Huajing Zeng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
6
|
Liu X, Kong X, Xu L, Su Y, Xu S, Pang X, Wang R, Ma Y, Tian Q, Han L. Synergistic therapeutic effect of ginsenoside Rg3 modified minoxidil transfersomes (MXD-Rg3@TFs) on androgenic alopecia in C57BL/6 mice. Int J Pharm 2024; 654:123963. [PMID: 38430952 DOI: 10.1016/j.ijpharm.2024.123963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
Inflammation in hair follicles will reduce the effectiveness of minoxidil (MXD) in the treatment of androgen alopecia (AGA) caused by elevated androgen levels. To target multiple physiological and pathological processes in AGA, a novel natural bioactive compound modified transfersomes (MXD-Rg3@TFs) was prepared to replace cholesterol that may disrupt hair growth, with ginsenosides Rg3 (Rg3) that have anti-inflammatory effects on AGA. The effects of MXD, Rg3 and their combination on AGA were evaluated using dihydrotestosterone (DHT) induced human dermal papilla cells (DPCs), and the results showed that the combination of MXD and Rg3 can significantly promote the proliferation, reduce the level of intracellular ROS and inflammatory factors, and inhibit the aging of DHT induced DPCs. Compared with cholesterol membrane transfersomes (MXD-Ch@TFs), MXD-Rg3@TFs has similar deformability, smaller particle size and better stability. MXD-Rg3@TFs has also significant advantages in shortening telogen phase and prolonging the growth period of hair follicles in C57BL/6 mice than MXD-Ch@TFs and commercial MXD tincture. The prominent ability of MXD-Rg3@TFs to inhibit the conversion of testosterone to DHT and reduce the level of inflammatory factors suggested that Rg3 and MXD in MXD-Rg3@TFs have synergistic effect on AGA therapy. MXD-Ch@TFs with no irritation to C57BL/6 mice skin is expected to reduce the dose of MXD and shorten the treatment time, which would undoubtedly provide a promising therapeutic option for treatment of AGA.
Collapse
Affiliation(s)
- Xiaxia Liu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Xia Kong
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Li Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Yonghui Su
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Shanshan Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoya Pang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Ruifen Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Yihan Ma
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China
| | - Qingping Tian
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Liwen Han
- School of Pharmacy & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China.
| |
Collapse
|
7
|
Liu Y, Li G, Ning J, Zhao Y. Unveiling the experimental proof of the anticancer potential of ginsenoside Rg3 (Review). Oncol Lett 2024; 27:182. [PMID: 38476209 PMCID: PMC10928969 DOI: 10.3892/ol.2024.14315] [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: 09/28/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Ginsenoside Rg3 (GS-Rg3), a sterol molecule isolated from ginseng, has demonstrated various immunological properties, including inhibition of cancer cell proliferation and metastasis, reversal of drug resistance and enhancement of chemotherapy sensitivity. The recent surge in attention towards GS-Rg3 can be attributed to its potential as an antitumor angiogenesis agent and as a therapeutic candidate for immunotherapy. The development of GS-Rg3 as an agent for these purposes has accelerated research on its mechanisms of action. The present review summarizes recent studies investigating the antitumor activity of GS-Rg3 and its underlying mechanisms, as well as providing essential information for future studies on GS-Rg3.
Collapse
Affiliation(s)
- Yongmin Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Guanchu Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jinyue Ning
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yi Zhao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
8
|
Li W, Cheng X, Zhu G, Hu Y, Wang Y, Niu Y, Li H, Aierken A, Li J, Feng L, Liu G. A review of chemotherapeutic drugs-induced arrhythmia and potential intervention with traditional Chinese medicines. Front Pharmacol 2024; 15:1340855. [PMID: 38572424 PMCID: PMC10987752 DOI: 10.3389/fphar.2024.1340855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Significant advances in chemotherapy drugs have reduced mortality in patients with malignant tumors. However, chemotherapy-related cardiotoxicity increases the morbidity and mortality of patients, and has become the second leading cause of death after tumor recurrence, which has received more and more attention in recent years. Arrhythmia is one of the common types of chemotherapy-induced cardiotoxicity, and has become a new risk related to chemotherapy treatment, which seriously affects the therapeutic outcome in patients. Traditional Chinese medicine has experienced thousands of years of clinical practice in China, and has accumulated a wealth of medical theories and treatment formulas, which has unique advantages in the prevention and treatment of malignant diseases. Traditional Chinese medicine may reduce the arrhythmic toxicity caused by chemotherapy without affecting the anti-cancer effect. This paper mainly discussed the types and pathogenesis of secondary chemotherapeutic drug-induced arrhythmia (CDIA), and summarized the studies on Chinese medicine compounds, Chinese medicine Combination Formula and Chinese medicine injection that may be beneficial in intervention with secondary CDIA including atrial fibrillation, ventricular arrhythmia and sinus bradycardia, in order to provide reference for clinical prevention and treatment of chemotherapy-induced arrhythmias.
Collapse
Affiliation(s)
- Weina Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaozhen Cheng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guanghui Zhu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine (National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion), Tianjin, China
| | - Yunhan Wang
- Henan Province Hospital of Traditional Chinese Medicine (The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine), Zhengzhou, Henan, China
| | - Yueyue Niu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongping Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aikeremu Aierken
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jie Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ling Feng
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guifang Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Luo X. Nanobiotechnology-based strategies in alleviation of chemotherapy-mediated cardiotoxicity. ENVIRONMENTAL RESEARCH 2023; 238:116989. [PMID: 37633635 DOI: 10.1016/j.envres.2023.116989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
The cardiovascular diseases have been among the most common malignancies and the first leading cause of death, even higher than cancer. The cardiovascular diseases can be developed as a result of cardiac dysfunction and damages to heart tissue. Exposure to toxic agents and chemicals that induce cardiac dysfunction has been of interest in recent years. The chemotherapy drugs are commonly used for cancer therapy and in these patients, cardiovascular diseases have been widely observed that is due to negative impact of chemotherapy drugs on the heart. These drugs increase oxidative damage and inflammation, and mediate apoptosis and cardiac dysfunction. Hence, nanotechnological approaches have been emerged as new strategies in attenuation of chemotherapy-mediated cardiotoxicity. The first advantage of nanoparticles can be explored in targeted and selective delivery of drugs to reduce their accumulation in heart tissue. Nanostructures can deliver bioactive and therapeutic compounds in reducing cardiotoxicity and alleviation toxic impacts of chemotherapy drugs. The functionalization of nanostructures increases their selectivity against tumor cells and reduces accumulation of drugs in heart tissue. The bioplatforms such as chitosan and alginate nanostructures can also deliver chemotherapy drugs and reduce their cardiotoxicity. The function of nanostructures is versatile in reduction of cardiotoxicity by chemotherapy drugs and new kind of platforms is hydrogels that can mediate sustained release of drug to reduce its toxic impacts on heart tissue. The various kinds of nanoplatforms have been developed for alleviation of cardiotoxicity and their future clinical application depends on their biocompatibility. High concentration level of chitosan nanoparticles can stimulate cardiotoxicity. Therefore, if nanotechnology is going to be deployed for drug delivery and reducing cardiotoxicity, the first pre-requirement is to lack toxicity on normal cells and have high biocompatibility.
Collapse
Affiliation(s)
- Xuanming Luo
- Department of General Surgery, Zhongshan Hospital, Fudan University, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China.
| |
Collapse
|
10
|
Huang Y, Lu J, Zhao L, Fu X, Peng S, Zhang W, Wang R, Yuan W, Luo R, Wang X, Li Z, Zhang Z. Retinal cell-targeted liposomal ginsenoside Rg3 attenuates retinal ischemia-reperfusion injury via alleviating oxidative stress and promoting microglia/macrophage M2 polarization. Free Radic Biol Med 2023; 206:162-179. [PMID: 37380044 DOI: 10.1016/j.freeradbiomed.2023.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 06/30/2023]
Abstract
Retinal ischemia-reperfusion (RIR) injury remains a major challenge that is detrimental to retinal cell survival in a variety of ocular diseases. However, current clinical treatments focus on a single pathological mechanism, making them unable to provide comprehensive retinal protection. A variety of natural products including ginsenoside Rg3 (Rg3) exhibit potent antioxidant and anti-inflammatory activities. Unfortunately, the hydrophobicity of Rg3 and the presence of various intraocular barriers limit its effective application in clinical settings. Hyaluronic acid (HA)- specifically binds to cell surface receptors, CD44, which is widely expressed in retinal pigment epithelial cells and M1-type macrophage. Here, we developed HA-decorated liposomes loaded with Rg3, termed Rg3@HA-Lips, to protect against retinal damage caused by RIR injury. Treatment with Rg3@HA-Lips significantly inhibited the oxidative stress induced by RIR injury. In addition, Rg3@HA-Lips promoted the transition of M1-type macrophage to the M2 type, ultimately reversing the pro-inflammatory microenvironment. The mechanism of Rg3@HA-Lips was further investigated and found that they can regulateSIRT/FOXO3a, NF-κB and STAT3 signaling pathways. Together with as well demonstrated good safety profiles, this CD44-targeted platform loaded with a natural product alleviates RIR injury by modulating the retinal microenvironment and present a potential clinical treatment strategy.
Collapse
Affiliation(s)
- Yanmei Huang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Laien Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Xiaoxuan Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Shengjun Peng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Wen Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Rong Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Wenze Yuan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Rongrui Luo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Xiaojie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Zelin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, People's Republic of China.
| |
Collapse
|
11
|
Balusamy SR, Perumalsamy H, Huq MA, Yoon TH, Mijakovic I, Thangavelu L, Yang DC, Rahimi S. A comprehensive and systemic review of ginseng-based nanomaterials: Synthesis, targeted delivery, and biomedical applications. Med Res Rev 2023; 43:1374-1410. [PMID: 36939049 DOI: 10.1002/med.21953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 11/22/2022] [Accepted: 02/26/2023] [Indexed: 03/21/2023]
Abstract
Among 17 Panax species identified across the world, Panax ginseng (Korean ginseng), Panax quinquefolius (American ginseng), and Panax notoginseng (Chinese ginseng) are highly recognized for the presence of bioactive compound, ginsenosides and their pharmacological effects. P. ginseng is widely used for synthesis of different types of nanoparticles compared to P. quinquefolius and P. notoginseng. The use of nano-ginseng could increase the oral bioavailability, membrane permeability, and thus provide effective delivery of ginsenosides to the target sites through transport system. In this review, we explore the synthesis of ginseng nanoparticles using plant extracts from various organs, microbes, and polymers, as well as their biomedical applications. Furthermore, we highlight transporters involved in transport of ginsenoside nanoparticles to the target sites. Size, zeta potential, temperature, and pH are also discussed as the critical parameters affecting the quality of ginseng nanoparticles synthesis.
Collapse
Affiliation(s)
- Sri Renukadevi Balusamy
- Department of Food Science and Biotechnology, Sejong University, Seoul, Gwangjin-gu, Republic of Korea
| | - Haribalan Perumalsamy
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Md Amdadul Huq
- Department of Food and Nutrition, Chung Ang University, Anseong-si, Gyeonggi-do, Republic of Korea
| | - Tae Hyun Yoon
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea
- Institute for Next Generation Material Design, Hanyang University, Seoul, Republic of Korea
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Ivan Mijakovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
| | - Deok Chun Yang
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin, Republic of Korea
| | - Shadi Rahimi
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
12
|
Li L, Cao J, Li S, Cui T, Ni J, Zhang H, Zhu Y, Mao J, Gao X, Midgley AC, Zhu M, Fan G. M2 Macrophage-Derived sEV Regulate Pro-Inflammatory CCR2 + Macrophage Subpopulations to Favor Post-AMI Cardiac Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202964. [PMID: 36950739 PMCID: PMC10190454 DOI: 10.1002/advs.202202964] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 02/21/2023] [Indexed: 05/18/2023]
Abstract
Tissue-resident cardiac macrophage subsets mediate cardiac tissue inflammation and repair after acute myocardial infarction (AMI). CC chemokine receptor 2 (CCR2)-expressing macrophages have phenotypical similarities to M1-polarized macrophages, are pro-inflammatory, and recruit CCR2+ circulating monocytes to infarcted myocardium. Small extracellular vesicles (sEV) from CCR2̶ macrophages, which phenotypically resemble M2-polarized macrophages, promote anti-inflammatory activity and cardiac repair. Here, the authors harvested M2 macrophage-derived sEV (M2EV ) from M2-polarized bone-marrow-derived macrophages for intramyocardial injection and recapitulation of sEV-mediated anti-inflammatory activity in ischemic-reperfusion (I/R) injured hearts. Rats and pigs received sham surgery; I/R without treatment; or I/R with autologous M2EV treatment. M2EV rescued cardiac function and attenuated injury markers, infarct size, and scar size. M2EV inhibited CCR2+ macrophage numbers, reduced monocyte-derived CCR2+ macrophage recruitment to infarct sites, induced M1-to-M2 macrophage switching and promoted neovascularization. Analysis of M2EV microRNA content revealed abundant miR-181b-5p, which regulated macrophage glucose uptake, glycolysis, and mitigated mitochondrial reactive oxygen species generation. Functional blockade of miR-181b-5p is detrimental to beneficial M2EV actions and resulted in failure to inhibit CCR2+ macrophage numbers and infarct size. Taken together, this investigation showed that M2EV rescued myocardial function, improved myocardial repair, and regulated CCR2+ macrophages via miR-181b-5p-dependent mechanisms, indicating an option for cell-free therapy for AMI.
Collapse
Affiliation(s)
- Lan Li
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive RegulationTianjin Central Hospital of Gynecology ObstetricsTianjin300052China
| | - Sheng Li
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Tianyi Cui
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Jingyu Ni
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Han Zhang
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Yan Zhu
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials for the Ministry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese MedicineKey Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of EducationTianjin University of Traditional Chinese MedicineTianjin301617China
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionState Key Laboratory of Component‐based Chinese MedicineFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjin300193China
| |
Collapse
|
13
|
Elfadadny A, Ragab RF, Hamada R, Al Jaouni SK, Fu J, Mousa SA, El-Far AH. Natural bioactive compounds-doxorubicin combinations targeting topoisomerase II-alpha: Anticancer efficacy and safety. Toxicol Appl Pharmacol 2023; 461:116405. [PMID: 36716865 DOI: 10.1016/j.taap.2023.116405] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/07/2023] [Accepted: 01/22/2023] [Indexed: 01/29/2023]
Abstract
Cancer is one of the leading causes of death worldwide, so pursuing effective and safe therapeutics for cancer is a key research objective nowadays. Doxorubicin (DOX) is one of the commonly prescribed chemotherapeutic agents that has been used to treat cancer with its antimitotic properties via inhibition of topoisomerase II (TOP2) activity. However, many problems hinder the broad use of DOX in clinical practice, including cardiotoxicity and drug resistance. Research in drug discovery has confirmed that natural bioactive compounds (NBACs) display a wide range of biological activities correlating to anticancer outcomes. The combination of NBACs has been seen to be an ideal candidate that might increase the effectiveness of DOX therapy and decreases its unfavorable adverse consequences. The current review discusses the chemo-modulatory mechanism and the protective effects of combined DOX with NBACs with a binding affinity (pKi) toward TOP2A more than pKi of DOX. This review will also discuss and emphasize the molecular mechanisms to provide a pathway for further studies to reveal other signaling pathways. Taken together, understanding the fundamental mechanisms and implications of combined therapy may provide a practical approach to battling cancer diseases.
Collapse
Affiliation(s)
- Ahmed Elfadadny
- Department of Animal Internal Medicine, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan.
| | - Rokaia F Ragab
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan; Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Rania Hamada
- Department of Pathology, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Soad K Al Jaouni
- Department of Hematology/Pediatric Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, China.
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| |
Collapse
|
14
|
Moini Jazani A, Arabzadeh A, Haghi-Aminjan H, Nasimi Doost Azgomi R. The role of ginseng derivatives against chemotherapy-induced cardiotoxicity: A systematic review of non-clinical studies. Front Cardiovasc Med 2023; 10:1022360. [PMID: 36844721 PMCID: PMC9946988 DOI: 10.3389/fcvm.2023.1022360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
Aims Although chemotherapy agents are used to treating cancers, they have serious side effects, like their harmful effects on the cardiovascular system, limiting the clinical use of these chemotherapy agents. This study aimed to systematically investigate the potential role of ginseng derivatives in the prevention of chemotherapy-induced cardiac toxicity. Methods This systematic review was performed according to PRISMA guidelines strategy in databases till August 2022. First, identify studies related to using search terms in titles and abstracts. After studying and screening 209 articles, 16 articles were selected in this study according to our inclusion and exclusion criteria. Results According to the findings of this study, ginseng derivatives showed significant changes in biochemical, histological, and heart weight loss, as well as a reduction in mortality, which occurred in the groups treated with chemotherapy agents compared to the control groups. Co-administration of ginseng derivatives with chemotherapy agents inhibited or reversed these changes to near-moderate levels. The protective effects of ginseng derivatives can be due to their anti-inflammatory, anti-oxidant, and anti-apoptotic action. Conclusion This systematic review shows evidence that concomitant administration of ginseng derivatives improves chemotherapy-induced cardiac toxicity. However, for better conclusions about the practical mechanisms of ginseng derivatives in reducing the cardiac toxic effects of chemotherapy agents and evaluating the efficacy and safety of the compound simultaneously, it is necessary to design comprehensive studies.
Collapse
Affiliation(s)
- Arezoo Moini Jazani
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran,*Correspondence: Hamed Haghi-Aminjan,✉
| | - Ramin Nasimi Doost Azgomi
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran,Ramin Nasimi Doost Azgomi,✉
| |
Collapse
|
15
|
Jung WH, Song J, You G, Lee JH, Lee SW, Ahn JH, Mok H. Protection of Skin Fibroblasts from Infrared-A-Induced Photo-Damage Using Ginsenoside Rg3(S)-Incorporated Soybean Lecithin Liposomes. J Microbiol Biotechnol 2023; 33:135-141. [PMID: 36575857 PMCID: PMC9895989 DOI: 10.4014/jmb.2210.10048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
Protection of skin cells from chronic infrared-A (IRA) irradiation is crucial for anti-photoaging of the skin. In this study, we investigated the protective activity of Rg3(S) and Rg3(S)-incorporated anionic soybean lecithin liposomes (Rg3/Lipo) with a size of approximately 150 nm against IRA-induced photodamage in human fibroblasts. The formulated Rg3/Lipo showed increased solubility in aqueous solution up to a concentration of 200 μg/ml, compared to free Rg3(S). In addition, Rg3/Lipo exhibited superior colloidal stability in aqueous solutions and biocompatibility for normal human dermal fibroblasts (NHDFs). After repeated IRA irradiation on NHDFs, elevated levels of cellular and mitochondrial reactive oxygen species (ROS) were greatly reduced by Rg3(S) and Rg3/Lipo. In addition, cells treated with Rg3/Lipo exhibited noticeably reduced apoptotic signals following IRA irradiation compared to untreated cells. Thus, considering aqueous solubility and cellular responses, Rg3/Lipo could serve as a promising infrared protector for healthy aging of skin cells.
Collapse
Affiliation(s)
- Won Ho Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyeon Song
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Gayeon You
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jun Hyuk Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Sin Won Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Joong-Hoon Ahn
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea,Corresponding author Phone: +82-2-450-0448 E-mail:
| |
Collapse
|
16
|
Rahimi S, van Leeuwen D, Roshanzamir F, Pandit S, Shi L, Sasanian N, Nielsen J, Esbjörner EK, Mijakovic I. Ginsenoside Rg3 Reduces the Toxicity of Graphene Oxide Used for pH-Responsive Delivery of Doxorubicin to Liver and Breast Cancer Cells. Pharmaceutics 2023; 15:pharmaceutics15020391. [PMID: 36839713 PMCID: PMC9965446 DOI: 10.3390/pharmaceutics15020391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
Doxorubicin (DOX) is extensively used in chemotherapy, but it has serious side effects and is inefficient against some cancers, e.g., hepatocarcinoma. To ameliorate the delivery of DOX and reduce its side effects, we designed a pH-responsive delivery system based on graphene oxide (GO) that is capable of a targeted drug release in the acidic tumor microenvironment. GO itself disrupted glutathione biosynthesis and induced reactive oxygen species (ROS) accumulation in human cells. It induced IL17-directed JAK-STAT signaling and VEGF gene expression, leading to increased cell proliferation as an unwanted effect. To counter this, GO was conjugated with the antioxidant, ginsenoside Rg3, prior to loading with DOX. The conjugation of Rg3 to GO significantly reduced the toxicity of the GO carrier by abolishing ROS production. Furthermore, treatment of cells with GO-Rg3 did not induce IL17-directed JAK-STAT signaling and VEGF gene expression-nor cell proliferation-suggesting GO-Rg3 as a promising drug carrier. The anticancer activity of GO-Rg3-DOX conjugates was investigated against Huh7 hepatocarcinoma and MDA-MB-231 breast cancer cells. GO-Rg3-DOX conjugates significantly reduced cancer cell viability, primarily via downregulation of transcription regulatory genes and upregulation of apoptosis genes. GO-Rg3 is an effective, biocompatible, and pH responsive DOX carrier with potential to improve chemotherapy-at least against liver and breast cancers.
Collapse
Affiliation(s)
- Shadi Rahimi
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Daniel van Leeuwen
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Fariba Roshanzamir
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Santosh Pandit
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Lei Shi
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Nima Sasanian
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Jens Nielsen
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
- BioInnovation Institute, DK-2200 Copenhagen, Denmark
| | - Elin K. Esbjörner
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
| | - Ivan Mijakovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, 412 96 Göteborg, Sweden
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK-2800 Lyngby, Denmark
- Correspondence:
| |
Collapse
|
17
|
Wang J, Liu YM, Hu J, Chen C. Trained immunity in monocyte/macrophage: Novel mechanism of phytochemicals in the treatment of atherosclerotic cardiovascular disease. Front Pharmacol 2023; 14:1109576. [PMID: 36895942 PMCID: PMC9989041 DOI: 10.3389/fphar.2023.1109576] [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: 11/29/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
Atherosclerosis (AS) is the pathology of atherosclerotic cardiovascular diseases (ASCVD), characterized by persistent chronic inflammation in the vessel wall, in which monocytes/macrophages play a key role. It has been reported that innate immune system cells can assume a persistent proinflammatory state after short stimulation with endogenous atherogenic stimuli. The pathogenesis of AS can be influenced by this persistent hyperactivation of the innate immune system, which is termed trained immunity. Trained immunity has also been implicated as a key pathological mechanism, leading to persistent chronic inflammation in AS. Trained immunity is mediated via epigenetic and metabolic reprogramming and occurs in mature innate immune cells and their bone marrow progenitors. Natural products are promising candidates for novel pharmacological agents that can be used to prevent or treat cardiovascular diseases (CVD). A variety of natural products and agents exhibiting antiatherosclerotic abilities have been reported to potentially interfere with the pharmacological targets of trained immunity. This review describes in as much detail as possible the mechanisms involved in trained immunity and how phytochemicals of this process inhibit AS by affecting trained monocytes/macrophages.
Collapse
Affiliation(s)
- Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Yong-Mei Liu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Jun Hu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| | - Cong Chen
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, China
| |
Collapse
|
18
|
Zuo S, Wang J, An X, Wang Z, Zheng X, Zhang Y. Fabrication of Ginsenoside-Based Nanodrugs for Enhanced Antitumor Efficacy on Triple-Negative Breast Cancer. Front Bioeng Biotechnol 2022; 10:945472. [PMID: 36032706 PMCID: PMC9412961 DOI: 10.3389/fbioe.2022.945472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/10/2022] [Indexed: 12/20/2022] Open
Abstract
There is an urgent need to identify chemotherapeutic agents with improved efficacy and safety against triple-negative breast cancer (TNBC). Ginsenosides can reportedly induce tumor cell death, invasion, and metastasis; however, poor water solubility, low oral absorption rate, and rapid blood clearance limit their clinical application. Utilizing the amphiphilic property of ginsenosides as building blocks of biomaterials, we fabricated a carrier-free nanodrug composed of ginsenosides Rg3 and Rb1 using a nano-reprecipitation method without any additional carriers. After characterizing and demonstrating their uniform morphology and pH-sensitive drug release properties, we observed that Rg3-Rb1 nanoparticles (NPs) exhibited stronger antitumor and anti-invasive effects on TNBCs in vitro than those mediated by free ginsenosides. Consequently, Rg3-Rb1 NPs afforded superior inhibition of tumor growth and reduction of pulmonary metastasis than the Rg3 and Rb1 mixture, with no obvious systematic toxicity in vivo. Collectively, our results provide a proof-of-concept that self-assembled engineered ginsenoside nanodrugs may be efficient and safe for TNBC therapy.
Collapse
Affiliation(s)
- Shuting Zuo
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jing Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xianquan An
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Zhenyu Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xiao Zheng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
- *Correspondence: Xiao Zheng, ; Yan Zhang,
| | - Yan Zhang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Xiao Zheng, ; Yan Zhang,
| |
Collapse
|
19
|
Ke Y, Huang L, Song Y, Liu Z, Liang L, Wang L, Wang T. Preparation and pharmacological effects of minor ginsenoside nanoparticles: a review. Front Pharmacol 2022; 13:974274. [PMID: 36003522 PMCID: PMC9393412 DOI: 10.3389/fphar.2022.974274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022] Open
Abstract
Ginseng (Panax ginseng) is a perennial herbaceous plant belonging to Panax genus of Araliaceae. Ginsenosides are a kind of important compounds in ginseng and minor ginsenosides are secondary metabolic derivatives of ginsenosides. Studies have shown that minor ginsenosides have many pharmacological effects, such as antioxidant, anti-tumor, anti-platelet aggregation, and neuroprotective effects. However, the therapeutic effects of minor ginsenosides are limited due to poor solubility in water, short half-life, and poor targeting accuracy. In recent years, to improve the application efficiency, the research on the nanocrystallization of minor ginsenosides have attracted extensive attention from researchers. This review focuses on the classification, preparation methods, pharmacological effects, and action mechanisms of minor ginsenoside nanoparticles, as well as existing problems and future direction of relevant research, which provides a reference for the in-depth research of minor ginsenoside nanoparticles.
Collapse
Affiliation(s)
- Yue Ke
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Lei Huang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Yu Song
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Zhenxin Liu
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Linshuang Liang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
| | - Linmao Wang
- Department of Thoracic Surgery, The First People’s Hospital of Yancheng, Affiliated Hospital 4 of Nantong University, Yancheng, China
- *Correspondence: Taoyun Wang, ; Linmao Wang,
| | - Taoyun Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou, China
- *Correspondence: Taoyun Wang, ; Linmao Wang,
| |
Collapse
|
20
|
Qu PR, Jiang ZL, Song PP, Liu LC, Xiang M, Wang J. Saponins and their derivatives: Potential candidates to alleviate anthracycline-induced cardiotoxicity and multidrug resistance. Pharmacol Res 2022; 182:106352. [PMID: 35835369 DOI: 10.1016/j.phrs.2022.106352] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022]
Abstract
Anthracyclines (ANTs) continue to play an irreplaceable role in oncology treatment. However, the clinical application of ANTs has been limited. In the first place, ANTs can cause dose-dependent cardiotoxicity such as arrhythmia, cardiomyopathy, and congestive heart failure. In the second place, the development of multidrug resistance (MDR) leads to their chemotherapeutic failure. Oncology cardiologists are urgently searching for agents that can both protect the heart and reverse MDR without compromising the antitumor effects of ANTs. Based on in vivo and in vitro data, we found that natural compounds, including saponins, may be active agents for other both natural and chemical compounds in the inhibition of anthracycline-induced cardiotoxicity (AIC) and the reversal of MDR. In this review, we summarize the work of previous researchers, describe the mechanisms of AIC and MDR, and focus on revealing the pharmacological effects and potential molecular targets of saponins and their derivatives in the inhibition of AIC and the reversal of MDR, aiming to encourage future research and clinical trials.
Collapse
Affiliation(s)
- Pei-Rong Qu
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhi-Lin Jiang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Ping-Ping Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medicine Sciences, Beijing 100013, China
| | - Lan-Chun Liu
- Beijing University of traditional Chinese Medicine, Beijing 100029, China
| | - Mi Xiang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| |
Collapse
|
21
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
22
|
Li F, Wan SY, Hu JG, Zhang Y, Yu BY, Kou JP. Recent advances of traditional chinese medicine in the regulation of myocardial mitochondrial function. WORLD JOURNAL OF TRADITIONAL CHINESE MEDICINE 2022. [DOI: 10.4103/wjtcm.wjtcm_78_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Yang F, Yang MY, Le JQ, Luo BY, Yin MD, Chao-Li, Jiang JL, Fang YF, Shao JW. Protective Effects and Therapeutics of Ginsenosides for Improving Endothelial Dysfunction: From Therapeutic Potentials, Pharmaceutical Developments to Clinical Trials. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:749-772. [PMID: 35450513 DOI: 10.1142/s0192415x22500318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
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.
Collapse
Affiliation(s)
- Fang Yang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Ming-Yue Yang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Qing Le
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bang-Yue Luo
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Meng-Die Yin
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Chao-Li
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jia-Li Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yi-Fan Fang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jing-Wei Shao
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
24
|
Liu J, Xin Y, Qiu Z, Zhang Q, He T, Qiu Y, Wang W. Cordyceps sinensis-mediated biotransformation of notoginsenoside R1 into 25-OH-20( S/ R)-R2 with elevated cardioprotective effect against DOX-induced cell injury. RSC Adv 2022; 12:12938-12946. [PMID: 35497008 PMCID: PMC9049007 DOI: 10.1039/d2ra01470j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023] Open
Abstract
Notoginsenoside R1 is a dammarane saponin in Panax notoginseng with promising cardioprotective effects. The bioactivity–structure relationship of such saponins suggested that the presence of a hydroxyl group at C25 could elevate its performance. To fulfill that goal, bioconversion of notoginsenoside R1 was mediated by a biocatalytic system of Cordyceps sinensis that had successfully produced multiple 25-OH derivatives from ginsenoside Re and Rg1. The major metabolic products of notoginsenoside R1 were identified as 25-OH-20(S/R)-R2 via the techniques of HRMS, 13C-NMR, 1H-NMR, HSQC and HMBC. Time-course experiments were designed to monitor the reaction process, establishing a biocatalytic pathway of “R1→20(S/R)-R2→25-OH-20(S/R)-R2”. The bioconversion rate of these 25-OH derivatives added up to 69.87% which greatly precedes the previous report. Afterwards, the effect of these biocatalytic products against doxorubicin-induced cardiotoxicity was evaluated, indicating a significant increase in efficacy after the hydration of the C24–C25 double bond on the dammarane skeleton. In conclusion, the biocatalytic system employed in this paper is able to harvest 25-OH-20(S/R)-R2 in high yield from notoginsenoside R1, which will provide lead compounds or drug candidates to alleviate myocardial injury caused by doxorubicin. The biocatalytic system in this paper preferably yielded 25-OH notoginsenoside R2 from R1 in a regioselective manner. Such a process significantly elevated the effects of these 25-OH derivatives against DOX-induced cardiomyocyte injury.![]()
Collapse
Affiliation(s)
- Jishuang Liu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Yu Xin
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Zhidong Qiu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Qi Zhang
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Tianzhu He
- School of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Ye Qiu
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Weinan Wang
- School of pharmaceutical sciences, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| |
Collapse
|
25
|
Wan Y, Wang J, Xu JF, Tang F, Chen L, Tan YZ, Rao CL, Ao H, Peng C. Panax ginseng and its ginsenosides: potential candidates for the prevention and treatment of chemotherapy-induced side effects. J Ginseng Res 2021; 45:617-630. [PMID: 34764717 PMCID: PMC8569258 DOI: 10.1016/j.jgr.2021.03.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/22/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy-induced side effects affect the quality of life and efficacy of treatment of cancer patients. Current approaches for treating the side effects of chemotherapy are poorly effective and may cause numerous harmful side effects. Therefore, developing new and effective drugs derived from natural non-toxic compounds for the treatment of chemotherapy-induced side effects is necessary. Experiments in vivo and in vitro indicate that Panax ginseng (PG) and its ginsenosides are undoubtedly non-toxic and effective options for the treatment of chemotherapy-induced side effects, such as nephrotoxicity, hepatotoxicity, cardiotoxicity, immunotoxicity, and hematopoietic inhibition. The mechanism focus on anti-oxidation, anti-inflammation, and anti-apoptosis, as well as the modulation of signaling pathways, such as nuclear factor erythroid-2 related factor 2 (Nrf2)/heme oxygenase-1 (HO-1), P62/keap1/Nrf2, c-jun N-terminal kinase (JNK)/P53/caspase 3, mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinases (ERK), AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR), mitogen-activated protein kinase kinase 4 (MKK4)/JNK, and phosphatidylinositol 3-kinase (PI3K)/AKT. Since a systemic review of the effect and mechanism of PG and its ginsenosides on chemotherapy-induced side effects has not yet been published, we provide a comprehensive summarization with this aim and shed light on the future research of PG.
Collapse
Key Words
- 5-FU, 5-fluorouracil
- ADM, Adriamycin
- ALT, alanine aminotransferase
- AMO, Atractylodes macrocephala volatile oil
- AMPK, AMP-activated protein kinase
- ARE, antioxidant response element
- AST, aspartate aminotransferase
- BMNC, bone marrow nucleated cells
- CIA, chemotherapy-induced hair loss
- CK, compound K
- CP, cisplatin
- CY, cyclophosphamide
- CYP2E1, Cytochrome P450 E1
- Chemotherapy
- DAC, doses of docetaxel, doxorubicin as well as cyclophosphamide
- ERG, enzyme-treated eRG
- ERK, extracellular signal-regulated kinases
- FBG, fermented black ginseng
- FRG, probiotic-fermented eRG
- FRGE, fermented red ginseng extract
- GM-CSF, granulocyte macrophage colony-stimulating factor
- Ginsenosides
- HEI-OC1, House Ear Institute-Organ of Corti 1
- HO-1, heme oxygenase-1
- HSPCS, haematopoietic stem and progenitor cells
- IL, interleukin
- JNK, c-jun N-terminal kinase
- KG-KH, the mixture of ginsenosides Rk3 and Rh4
- LLC-PK1, porcine renal proximal epithelial tubular
- LSK, Lin−Sca-1+c-kit+
- MAPK, mitogen-activated protein kinase
- MDA, malonaldehyde
- MEK, mitogen activated protein kinase
- MKK4, mitogen activated protein kinase kinase 4
- Mechanism
- NF-κB, nuclear factor-kappa B p65
- NQO, NAD (P) H quinone oxidoreductase
- Nrf2, nuclear factor erythroid related factor 2
- PG
- PG, Panax ginseng
- PGFR, PG flower
- PGLF, PG leaf
- PGRT, PG root
- PGS, PG total saponins
- PGSD, PG seeds
- PGSM, PG stem
- PI3K, phosphatidylinositol 3-kinase
- PPD, protopanaxadiol
- PPT, protopanaxatriol
- Pharmacological effects
- RG, red ginseng
- RGE, red ginseng extract
- ROS, reactive oxygen species
- SREBP-1, sterol regulatory element binding protein 1
- Side effects
- TNF-α, tumor necrosis factor-α
- eRG, 50% ethanol-extracted RG
- mTOR, mammalian target of rapamycin
- wRG, water-extracted RG
Collapse
Affiliation(s)
- Yan Wan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Wang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin-feng Xu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-zhu Tan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao-long Rao
- College of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
26
|
Wang H, Zheng Y, Sun Q, Zhang Z, Zhao M, Peng C, Shi S. Ginsenosides emerging as both bifunctional drugs and nanocarriers for enhanced antitumor therapies. J Nanobiotechnology 2021; 19:322. [PMID: 34654430 PMCID: PMC8518152 DOI: 10.1186/s12951-021-01062-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ginsenosides, the main components isolated from Panax ginseng, can play a therapeutic role by inducing tumor cell apoptosis and reducing proliferation, invasion, metastasis; by enhancing immune regulation; and by reversing tumor cell multidrug resistance. However, clinical applications have been limited because of ginsenosides' physical and chemical properties such as low solubility and poor stability, as well as their short half-life, easy elimination, degradation, and other pharmacokinetic properties in vivo. In recent years, developing a ginsenoside delivery system for bifunctional drugs or carriers has attracted much attention from researchers. To create a precise treatment strategy for cancer, a variety of nano delivery systems and preparation technologies based on ginsenosides have been conducted (e.g., polymer nanoparticles [NPs], liposomes, micelles, microemulsions, protein NPs, metals and inorganic NPs, biomimetic NPs). It is desirable to design a targeted delivery system to achieve antitumor efficacy that can not only cross various barriers but also can enhance immune regulation, eventually converting to a clinical application. Therefore, this review focused on the latest research about delivery systems encapsulated or modified with ginsenosides, and unification of medicines and excipients based on ginsenosides for improving drug bioavailability and targeting ability. In addition, challenges and new treatment methods were discussed to support the development of these new tumor therapeutic agents for use in clinical treatment.
Collapse
Affiliation(s)
- Hong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yu Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
27
|
Zhang Q, Li X, Gao X, Cao C, Hu Y, Guo H. Total saponins from stems and leaves of Panax quinquefolius L. ameliorate podophyllotoxin-induced myelosuppression and gastrointestinal toxicity. Biomed Chromatogr 2021; 36:e5266. [PMID: 34648200 DOI: 10.1002/bmc.5266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 11/06/2022]
Abstract
Podophyllotoxin (POD), a natural lignan distributed in podophyllum species, possesses significant antitumor and antiviral activities. But POD often causes serious side effects, such as myelosuppression, gastrointestinal toxicity, neurotoxicity, hepatic and renal dysfunction, and even death, which not only hinder its clinical application but also threaten the patient's health. Therefore, an effective treatment against POD-induced toxicity is important. Our preliminary study found that the total saponins from the stems and leaves of Panax quinquefolius L. (PQS) could significantly reduce the death of mice caused by POD. To reveal how PQS can alleviate POD-induced toxicity, further study was needed. Peripheral blood cell analysis, diarrhea score, and histological examination demonstrated that PQS could relieve myelosuppression and gastrointestinal side effects induced by POD. Then, metabolomics was performed to investigate the possible protective mechanism of PQS on POD-induced myelosuppression and gastrointestinal toxicity. Metabolomics analysis showed that metabolic changes caused by POD could be reversed by PQS to some extent; 23 metabolites altered significantly after POD exposure, and 11 metabolites significantly reversed by PQS pretreatment. Metabolic pathway analysis suggested that PQS might exhibit its protective effects by rebalancing disordered arginine, glutamine, and unsaturated fatty acid metabolism.
Collapse
Affiliation(s)
- Qianqian Zhang
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China.,School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xuemei Li
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| | - Xiaoxin Gao
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| | - Chunran Cao
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| | - Yuchi Hu
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| | - Hongzhu Guo
- Beijing Institute for Drug Control, NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Liu Z, Bian X, Gao W, Su J, Ma C, Xiao X, Yu T, Zhang H, Liu X, Fan G. Rg3 promotes the SUMOylation of SERCA2a and corrects cardiac dysfunction in heart failure. Pharmacol Res 2021; 172:105843. [PMID: 34428586 DOI: 10.1016/j.phrs.2021.105843] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 01/14/2023]
Abstract
SUMOylation of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) has been shown to play a critical role in the abnormal Ca2+ cycle of heart failure. Ginsenoside Rg3 (Rg3), the main active constituent of Panax ginseng, exerts a wide range of pharmacological effects in cardiovascular diseases. However, the effect of Rg3 on abnormal Ca2+ homeostasis in heart failure has not been reported. In this study, we showed a novel role of Rg3 in the abnormal Ca2+ cycle in cardiomyocytes of mice with heart failure. Among mice undergoing transverse aortic constriction, animals that received Rg3 showed improvements in cardiac function and Ca2+ homeostasis, accompanied by increases in the SUMOylation level and SERCA2a activity. In an isoproterenol (ISO)-induced cell hypertrophy model, Rg3 reduced the ISO-induced Ca2+ overload in HL-1 cells. Gene knockout of SUMO1 in mice inhibited the cardioprotective effect of Rg3, and SUMO1 knockout mice that received Rg3 did not exhibit improved Ca2+ homeostasis in cardiomyocytes. Additionally, mutation of the SUMOylation sites of SERCA2a blocked the positive effect of Rg3 on the ISO-induced abnormal Ca2+ cycle in HL-1 cells, and was accompanied by an abnormal endoplasmic reticulum stress response and generation of ROS. Our data demonstrated that Rg3 has a positive effect on the abnormal Ca2+ cycle in the cardiomyocytes of mice with heart failure. SUMO1 is an important factor that mediates the protective effect of Rg3. Our findings suggest that drug intervention by regulating the SUMOylation of SERCA2a can provide a novel therapeutic strategy for the treatment of heart failure.
Collapse
Affiliation(s)
- Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiyun Bian
- Central Laboratory, the Fifth Central Hospital of Tianjin, Tianjin 300450, China; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, the Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Wenbo Gao
- Central Laboratory, the Fifth Central Hospital of Tianjin, Tianjin 300450, China; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, the Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Jing Su
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China
| | - Xiaolin Xiao
- Central Laboratory, the Fifth Central Hospital of Tianjin, Tianjin 300450, China; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, the Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Tian Yu
- Central Laboratory, the Fifth Central Hospital of Tianjin, Tianjin 300450, China; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, the Fifth Central Hospital of Tianjin, Tianjin 300450, China
| | - Han Zhang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaozhi Liu
- Central Laboratory, the Fifth Central Hospital of Tianjin, Tianjin 300450, China; Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, the Fifth Central Hospital of Tianjin, Tianjin 300450, China.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300193, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
29
|
Lu Y, Pan J, Zhu X, Zhang S, Liu C, Sun J, Li Y, Chen S, Huang J, Cao C, Wang Y, Li Y, Liu T. Pharmacokinetic herb-drug interactions between Aidi injection and doxorubicin in rats with diethylnitrosamine-induced hepatocellular carcinoma. BMC Pharmacol Toxicol 2021; 22:48. [PMID: 34488896 PMCID: PMC8419969 DOI: 10.1186/s40360-021-00515-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Aidi Injection (ADI), a Chinese herbal preparation with anti-cancer activity, is used for the treatment of hepatocellular carcinoma (HCC). Several clinical studies have shown that co-administration of ADI with doxorubicin (DOX) is associated with reduced toxicity of chemotherapy, enhanced clinical efficacy and improved quality of life for patients. However, limited information is available about the herb-drug interactions between ADI and DOX. The study aimed to investigate the pharmacokinetic mechanism of herb-drug interactions between ADI and DOX in a rat model of HCC. METHODS Experimental HCC was induced in rats by oral administration of diethylnitrosamine. The HCC rats were pretreated with ADI (10 mL/kg, intraperitoneal injection) for 14 consecutive days prior to administration of DOX (7 mg/kg, intravenous injection) to investigate pharmacokinetic interactions. Plasma concentrations of DOX and its major metabolite, doxorubicinol (DOXol), were determined using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS Preadministration of ADI significantly altered the pharmacokinetics of DOX in HCC rats, leading to increased plasma concentrations of both DOX and DOXol. The area under the plasma drug concentration-time curve (AUCs) of DOX and DOXol in rats pretreated with ADI were 3.79-fold and 2.92-fold higher, respectively, than those in control rats that did not receive ADI. CONCLUSIONS Increased levels of DOX and DOXol were found in the plasma of HCC rats pretreated with ADI.
Collapse
Affiliation(s)
- Yuan Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Jie Pan
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Xiaoqing Zhu
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Shuai Zhang
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550000, China
| | - Chunhua Liu
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yueting Li
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China
| | - Siying Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Jing Huang
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Chuang Cao
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yonglin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China
| | - Yongjun Li
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China.
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, 550004, China.
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, 550004, China.
- School of Pharmacy, Guizhou Medical University, No.9, Beijing Road, Yunyan District, Guiyang, 550004, China.
| |
Collapse
|
30
|
Su X, Zhang X, Liu W, Yang X, An N, Yang F, Sun J, Xing Y, Shang H. Advances in the application of nanotechnology in reducing cardiotoxicity induced by cancer chemotherapy. Semin Cancer Biol 2021; 86:929-942. [PMID: 34375726 DOI: 10.1016/j.semcancer.2021.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/08/2023]
Abstract
Advances in the development of anti-tumour drugs and related technologies have resulted in a significant increase in the number of cancer survivors. However, the incidence of chemotherapy-induced cardiotoxicity (CIC) has been rising continuously, threatening their long-term survival. The integration of nanotechnology and biomedicine has brought about an unprecedented technological revolution and has promoted the progress of anti-tumour therapy. In this review, we summarised the possible mechanisms of CIC, evaluated the role of nanoparticles (including liposomes, polymeric micelles, dendrimers, and hydrogels) as drug carriers in preventing cardiotoxicity and proposed five advantages of nanotechnology in reducing cardiotoxicity: Liposomes cannot easily penetrate the heart's endothelial barrier; optimized delivery strategies reduce distribution in important organs, such as the heart; targeting the tumour microenvironment and niche; stimulus-responsive polymer nano-drug carriers rapidly iterate; better economic benefits were obtained. Nanoparticles can effectively deliver chemotherapeutic drugs to tumour tissues, while reducing the toxicity to heart tissues, and break through the dilemma of existing chemotherapy to a certain extent. It is important to explore the interactions between the physicochemical properties of nanoparticles and optimize the highly specific tumour targeting strategy in the future.
Collapse
Affiliation(s)
- Xin Su
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyu Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjing Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Yang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fan Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiahao Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
31
|
Sun X, Song Y, Xie Y, Han J, Chen F, Sun Y, Sui B, Jiang D. Shenlijia Attenuates Doxorubicin-Induced Chronic Heart Failure by Inhibiting Cardiac Fibrosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6659676. [PMID: 34326887 PMCID: PMC8310442 DOI: 10.1155/2021/6659676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/29/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022]
Abstract
Application of the anticancer drug doxorubicin (DOX) is restricted due to its adverse, cardiotoxic side effects, which ultimately result in heart failure. Moreover, there are a limited number of chemical agents for the clinical prevention of DOX-induced cardiotoxicity. Based on the theories of traditional Chinese medicine (TCM) on chronic heart failure (CHF), Shenlijia (SLJ), a new TCM compound, has been developed to fulfill multiple functions, including improving cardiac function and inhibiting cardiac fibrosis. In the present study, the protective effects and molecular mechanisms of SLJ on DOX-induced CHF rats were investigated. The CHF rat model was induced by intraperitoneal injection of DOX for six weeks with the cumulative dose of 15 mg/kg. All rats were then randomly divided into the control, CHF, CHF + SLJ (3.0 g/kg per day), and CHF + captopril (3.8 mg/kg per day) groups and treated for further four weeks. Echocardiography and the assessment of hemodynamic parameters were performed to evaluate heart function. A protein chip was applied to identify proteins with diagnostic values that were differentially expressed following SLJ treatment. The data from these investigations showed that SLJ treatment significantly improved cardiac function by increasing the left ventricular ejection fraction, improving the hemodynamic index, and inhibiting interstitial fibrosis. Protein chip analysis revealed that SLJ upregulated MCP-1, MDC, neuropilin-2, TGF-β3, thrombospondin, TIE-2, EG-VEGF/PK1, and TIMP-1/2/3 expressions and downregulated that of MMP-13. In addition, immunohistochemistry and western blot results further confirmed that SLJ promoted TIMP-1/2/3 and inhibited MMP-13 expression. The results of the present study suggest that SLJ was effective against DOX-induced CHF rats and is related to the improvement of heart function and ultrastructure and the inhibition of myocardial fibrosis.
Collapse
Affiliation(s)
- Xutao Sun
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunjia Song
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Xie
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jieru Han
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fei Chen
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yang Sun
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bowen Sui
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Deyou Jiang
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
32
|
Ni J, Liu Z, Jiang M, Li L, Deng J, Wang X, Su J, Zhu Y, He F, Mao J, Gao X, Fan G. Ginsenoside Rg3 ameliorates myocardial glucose metabolism and insulin resistance via activating the AMPK signaling pathway. J Ginseng Res 2021; 46:235-247. [PMID: 35509823 PMCID: PMC9058838 DOI: 10.1016/j.jgr.2021.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background Ginsenoside Rg3 is one of the main active ingredients in ginseng. Here, we aimed to confirm its protective effect on the heart function in transverse aortic coarctation (TAC)-induced heart failure mice and explore the potential molecular mechanisms involved. Methods The effects of ginsenoside Rg3 on heart and mitochondrial function were investigated by treating TAC-induced heart failure in mice. The mechanism of ginsenoside Rg3 for improving heart and mitochondrial function in mice with heart failure was predicted through integrative analysis of the proteome and plasma metabolome. Glucose uptake and myocardial insulin sensitivity were evaluated using micro-positron emission tomography. The effect of ginsenoside Rg3 on myocardial insulin sensitivity was clarified by combining in vivo animal experiments and in vitro cell experiments. Results Treatment of TAC-induced mouse models with ginsenoside Rg3 significantly improved heart function and protected mitochondrial structure and function. Fusion of metabolomics, proteomics, and targeted metabolomics data showed that Rg3 regulated the glycolysis process, and Rg3 not only regulated glucose uptake but also improve myocardial insulin resistance. The molecular mechanism of ginsenoside Rg3 regulation of glucose metabolism was determined by exploring the interaction pathways of AMPK, insulin resistance, and glucose metabolism. The effect of ginsenoside Rg3 on the promotion of glucose uptake in IR-H9c2 cells by AMPK activation was dependent on the insulin signaling pathway. Conclusions Ginsenoside Rg3 modulates glucose metabolism and significantly ameliorates insulin resistance through activation of the AMPK pathway.
Collapse
|
33
|
Hesari M, Mohammadi P, Khademi F, Shackebaei D, Momtaz S, Moasefi N, Farzaei MH, Abdollahi M. Current Advances in the Use of Nanophytomedicine Therapies for Human Cardiovascular Diseases. Int J Nanomedicine 2021; 16:3293-3315. [PMID: 34007178 PMCID: PMC8123960 DOI: 10.2147/ijn.s295508] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/16/2021] [Indexed: 12/15/2022] Open
Abstract
Considering the high prevalence of cardiovascular diseases (CVDs), the primary cause of death during the last several decades, it is necessary to develop proper strategies for the prevention and treatment of CVDs. Given the excessive side effects of current therapies, alternative therapeutic approaches like medicinal plants and natural products are preferred. Lower toxicity, chemical diversity, cost-effectiveness, and proven therapeutic potentials make natural products superior compared to other products. Nanoformulation methods improve the solubility, bioavailability, circulation time, surface area-to-volume ratio, systemic adverse side effects, and drug delivery efficiency of these medications. This study intended to review the functionality of the most recent nanoformulated medicinal plants and/or natural products against various cardiovascular conditions such as hypertension, atherosclerosis, thrombosis, and myocardial infarction. Literature review revealed that curcumin, quercetin, and resveratrol were the most applied natural products, respectively. Combination therapy, conjugation, or fabrication of nanoparticles and nanocarriers improved the applications and therapeutic efficacy of herbal- or natural-based nanoformulations. In the context of CVDs prevention and/or treatment, available data suggest that natural-based nanoformulations are considerably efficient, alone or in blend with other herbal/synthetic medicines. However, clinical trials are mandatory to elucidate the safety, cardioprotective effect, and mechanism of actions of nanophytomedicines.
Collapse
Affiliation(s)
- Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Khademi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran.,Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Narges Moasefi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Ginsenoside Rg 3 Attenuates Early Hepatic Injury via Inhibiting PPARγ- and Ang II-Related Inflammation and Fibrosis in Type II Diabetic Mice. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211009691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Ginsenoside Rg3 (Rg3), a natural product abundantly present in Korean Red Ginseng, is widely known for its anti-tumor activity. In our previous studies, we had further demonstrated that Rg3 has protective effects on the hearts, kidneys, and aortas of animals with hypertension or hypercholesterolemia, and its main mechanisms include down-regulation of angiotensin II (Ang II) levels and activation of peroxisome proliferator-activated receptor gamma (PPARγ) pathway in those tissues. In this study, the protective effects of Rg3 on liver were determined in db/db mice, a most recognized type II diabetes (T2DM) animal model with nonalcoholic fatty liver disease (NAFLD). The results showed that Rg3 did not have obvious effects to the body weight, blood glucose, and lipids of db/db mice. According to the results of histology examination, Rg3 could not improve steatosis in the hepatic tissue, too. But Rg3 did attenuate alanine aminotransferase (ALT)/aspartate aminotransferase (AST) elevation in serum and collagen deposition in hepatic tissue. Immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) showed that Rg3 upregulated PPARγ and downregulated Ang II in hepatic tissue in db/db mice, which resulted in reducing activities of transforming growth factor β (TGF-β)/connective tissue growth factor (CTGF) pathway, downregulating the levels of inflammatory cytokines and attenuating collagen accumulation. In conclusion, although it has no obvious effect on steatosis in the hepatic tissue, Rg3 indeed attenuates early hepatic injury from NAFLD via inhibiting PPARγ- and Ang II-related inflammation and fibrosis in T2DM db/db mice. These effects are independent of reducing blood glucose and lipids, and the mechanisms are similar to the protective effects of Rg3 in hypertension and hypercholesterolemia animals in our previous studies.
Collapse
|
35
|
Sarhene M, Ni JY, Duncan ES, Liu Z, Li S, Zhang J, Guo R, Gao S, Gao X, Fan G. Ginsenosides for cardiovascular diseases; update on pre-clinical and clinical evidence, pharmacological effects and the mechanisms of action. Pharmacol Res 2021; 166:105481. [PMID: 33549726 DOI: 10.1016/j.phrs.2021.105481] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/20/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
Cardiovascular disease (CVD) remains the major cause of death worldwide, accounting for almost 31% of the global mortality annually. Several preclinical studies have indicated that ginseng and the major bioactive ingredient (ginsenosides) can modulate several CVDs through diverse mechanisms. However, there is paucity in the translation of such experiments into clinical arena for cardiovascular ailments due to lack of conclusive specific pathways through which these activities are initiated and lack of larger, long-term well-structured clinical trials. Therefore, this review elaborates on current pharmacological effects of ginseng and ginsenosides in the cardiovascular system and provides some insights into the safety, toxicity, and synergistic effects in human trials. The review concludes that before ginseng, ginsenosides and their preparations could be utilized in the clinical treatment of CVDs, there should be more preclinical studies in larger animals (like the guinea pig, rabbit, dog, and monkey) to find the specific dosages, address the toxicity, safety and synergistic effects with other conventional drugs. This could lead to the initiation of large-scale, long-term well-structured randomized, and placebo-controlled clinical trials to test whether treatment is effective for a longer period and test the efficacy against other conventional therapies.
Collapse
Affiliation(s)
- Michael Sarhene
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Jing Yu Ni
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Esi Sophia Duncan
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Zhihao Liu
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Sheng Li
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Jing Zhang
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Rui Guo
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Shan Gao
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- First teaching hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin 300193, China.
| |
Collapse
|
36
|
Abstract
Adriamycin (ADR)-induced chronic heart injury (CHI) is a serious complication of chemotherapy. The present study was designed to assess the ability of fasudil, a Rho kinase inhibitor, to prevent ADR-induced CHI. Forty male 6-week-old C57BL6 mice were randomly divided into the following four groups: (1) control group, (2) CHI induced by adriamycin (ADR group), (3) CHI plus low dose fasudil (ADR + L group), and (4) CHI plus high dose fasudil (ADR + H group). Animals from groups 2-4 received ADR (2.5 mg/kg, i.p.) once a week for 8 weeks, and the control group received saline. Meanwhile, the animals in groups 3-4 received 2 mg/kg/day or 10 mg/kg/day fasudil, respectively. After measurement of cardiac functions, blood samples were collected for biochemical assays. The hearts were excised for histological, immunohistochemistry and western blot study, respectively. Adriamycin produced evident cardiac damage revealed by cardiac functions changes: decreased left ventricular fractional shortening (FS), left ventricular ejection fraction (EF), increased left ventricular volume, cardiac injury marker changes (increased creatine kinase, lactate dehydrogenase), antioxidant enzymes activity changes (decreased superoxide dismutase), and lipid peroxidation (elevated malondialdehyde) to the control group. Fasudil treatment notably ameliorated ADR-induced cardiac damage, restored heart function, suppressed cell apoptosis and senescence, ameliorated redox imbalance, and DNA damage. Fasudil has a protective effect on ADR-induced chronic heart injury, which partially attributed to its antioxidant, anti-apoptotic effects of inhibiting the RhoA/Rho kinase (ROCK) signaling pathway.
Collapse
|
37
|
Wang X, Meng H, Wang Q, Shao M, Lu W, Chen X, Jiang Y, Li C, Wang Y, Tu P. Baoyuan decoction ameliorates apoptosis via AT1-CARP signaling pathway in H9C2 cells and heart failure post-acute myocardial infarction rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112536. [PMID: 31931161 DOI: 10.1016/j.jep.2019.112536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/06/2019] [Accepted: 12/27/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Previous studies have approved that Baoyuan decoction (BYD) exerted remarkable cardioprotective effects on heart failure (HF) due to its anti-apoptotic properties. As a novel biomarker and target of HF, Cardiac ankyrin repeat protein (CARP) can exacerbate apoptosis via activation by angiotensin type 1 receptor (AT1) and subsequently deteriorate heart function. Transcriptome results in our previous study indicated BYD was beneficial to HF post-acute myocardial infarction (AMI) with a promising effect on CARP. However, the mechanism remains to be validated. AIM OF THE STUDY This study aims to elucidate whether BYD ameliorates apoptosis to protect against HF via AT1-CARP signaling pathway. MATERIALS AND METHODS Left anterior descending ligation was applied to induce an HF rat model, Ang Ⅱ-stimulated H9C2 cells apoptotic model and overexpression of Ankrd1/CARP H9C2 cells were established to clarify the effects and potential mechanism of BYD. Ethanol extracts of BYD (0.64; 1.28; 2.57 g/kg) were orally administered for four weeks and Fosinopril (4.67 mg/kg) was selected as a positive group in vivo. In vitro, BYD (400, 600, 800 μg/ml) or RNH6270 (an inhibitor of AT1, 1 μM) was co-cultured with Ang Ⅱ stimulation for 48 h in H9C2 cells. Overexpression of Ankrd1/CARP was conducted by transient transfection with H9C2 cells to further confirm the exact mechanism. Finally, to define the active ingredients of anti-cardiomyocyte apoptosis in BYD, we furtherly used the Ang Ⅱ-induced cardiomyocyte apoptosis model to evaluate the effects. RESULTS Echocardiography and TUNEL results showed that BYD in different doses remarkably improved heart function and inhibited apoptosis in vivo. Further study demonstrated that AT1 and CARP expressions in cardiac tissue were suppressed by BYD, accompanied with upregulation of B cell lymphoma-2 (Bcl-2) and downregulation of several pro-apoptotic molecules, including p53, Bcl-2 Associated X Protein (Bax) and Cleaved caspase 3. In parallel with the vivo experiment, in vitro research indicated BYD dramatically reduced the apoptotic cells and regulated expressions of critical apoptosis-related molecules mediated through downregulation of AT1 and CARP simultaneously which were consistent with the results in vivo experiment. Transiently transfected CARP over-expression further confirmed that BYD could suppress severe cardiomyocytes apoptosis induced by overexpression of CARP. Especially, the active ingredients of BYD including Astragaloside IV, Ginsenoside Rg3, Rb1, Rc and Re showed significantly anti-apoptosis effects. CONCLUSION BYD improves cardiac function and protects against cardiomyocytes injury by inhibiting apoptosis via regulating the AT1-CARP signaling pathway.
Collapse
Affiliation(s)
- Xiaoping Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui Meng
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qixin Wang
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingyan Shao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenji Lu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xu Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
38
|
Mendez-Fernandez A, Cabrera-Fuentes HA, Velmurugan B, Irei J, Boisvert WA, Lu S, Hausenloy DJ. Nanoparticle delivery of cardioprotective therapies. CONDITIONING MEDICINE 2020; 3:18-30. [PMID: 34268485 PMCID: PMC8279025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Acute myocardial infarction (AMI), and the heart failure (HF) that often follows, are leading causes of death and disability worldwide. Crucially, there are currently no effective treatments, other than myocardial reperfusion, for reducing myocardial infarct (MI) size and preventing HF following AMI. Thus, there is an unmet need to discover novel cardioprotective therapies to reduce MI size, and prevent HF in AMI patients. Although a large number of therapies have been shown to reduce MI size in experimental studies, the majority have failed to benefit AMI patients. Failure to deliver cardioprotective therapy to the ischemic heart in sufficient concentrations following AMI is a major factor for the lack of success observed in previous clinical cardioprotection studies. Therefore, new strategies are needed to improve the delivery of cardioprotective therapies to the ischemic heart following AMI. In this regard, nanoparticles have emerged as drug delivery systems for improving the bioavailability, delivery, and release of cardioprotective therapies, and should result in improved efficacy in terms of reducing MI size and preventing HF. In this article, we provide a review of currently available nanoparticles, some of which have been FDA-approved, in terms of their use as drug delivery systems in cardiovascular disease and cardioprotection.
Collapse
Affiliation(s)
- Abraham Mendez-Fernandez
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
- National Heart Research Institute Singapore, National Heart Centre, Singapore
| | - Hector A Cabrera-Fuentes
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Nuevo Leon, Mexico
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Russian Federation
- Institute of Physiology, Medical School, Justus-Liebig-University, Germany
| | - Bhaarathy Velmurugan
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - William A. Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, USA
| | - Shengjie Lu
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
| | - Derek J Hausenloy
- National Heart Research Institute Singapore, National Heart Centre, Singapore
- SingHealth Duke-NUS Cardiovascular Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, UK
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
- Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan
| |
Collapse
|
39
|
Ginsenoside Rg3 Attenuates Angiotensin II-Mediated Renal Injury in Rats and Mice by Upregulating Angiotensin-Converting Enzyme 2 in the Renal Tissue. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:6741057. [PMID: 31885658 PMCID: PMC6915024 DOI: 10.1155/2019/6741057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/05/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
Angiotensin II- (Ang II-) mediated renal injury represents a major pathogenetic mechanism in most chronic kidney diseases. Our previous research demonstrated that ginsenoside Rg3 (Rg3) attenuates Ang II elevation in the myocardium in spontaneously hypertensive rats (SHR). It is possible that Rg3 has similar effects in the renal tissue. In this research, we first demonstrated that Rg3 could attenuate Ang II increase in the kidney of SHR and reduce hypertensive nephropathy progression. Then, we found that Rg3 attenuated Ang II increase by upregulating angiotensin-converting enzyme 2 (ACE2) in the renal tissue. We confirmed this finding in an exogenous Ang II-infused mice model of renal injury, and two models showed consistent results. In conclusion, Rg3 attenuates Ang II-mediated renal injury in rats and mice by upregulating ACE2 in the renal tissue. This research is the first to demonstrate that Rg3 increases tissue ACE2 levels in vivo.
Collapse
|
40
|
Li L, Wang Y, Guo R, Li S, Ni J, Gao S, Gao X, Mao J, Zhu Y, Wu P, Wang H, Kong D, Zhang H, Zhu M, Fan G. Ginsenoside Rg3-loaded, reactive oxygen species-responsive polymeric nanoparticles for alleviating myocardial ischemia-reperfusion injury. J Control Release 2019; 317:259-272. [PMID: 31783047 DOI: 10.1016/j.jconrel.2019.11.032] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 01/07/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a serious threat to the health and lives of patients without any effective therapy. Excessive production of reactive oxygen species (ROS) is considered a principal cause of MIRI. Some natural products, including ginsenoside Rg3 (Rg3), exhibit robust antioxidant activity. However, the lack of an effective delivery strategy for this hydrophobic compound hinders its clinical application. In addition, therapeutic targets and molecular mechanisms of Rg3 require further elucidation to establish its mode of action. This study aimed to generate ROS-responsive nanoparticles (PEG-b-PPS) via the self-assembly of diblock copolymers of poly (ethylene glycol) (PEG) and poly (propylene sulfide) (PPS) and use them for Rg3 encapsulation and delivery. We identified FoxO3a as the therapeutic target of Rg3 using molecular docking and gene silencing. In rat ischemia-reperfusion model, an intramyocardial injection of Rg3-loaded PEG-b-PPS nanoparticles improved the cardiac function and reduced the infarct size. The mechanism of action was established as Rg3 targeting of FoxO3a, which inhibited the promotion of oxidative stress, inflammation, and fibrosis via downstream signaling pathways. In conclusion, this approach, involving ROS-responsive drug release, together with the identification of the target and mechanism of action of Rg3, provided an effective strategy for treating ischemic diseases and oxidative stress and could accelerate the implementation of hydrophobic natural products in clinical applications.
Collapse
Affiliation(s)
- Lan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yili Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Sheng Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiumei Gao
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yan Zhu
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pingli Wu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, Tianjin 300071, China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Deling Kong
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, Tianjin 300071, China
| | - Han Zhang
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Meifeng Zhu
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, Tianjin 300071, China; Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
41
|
Targeting Reactive Oxygen Species in Cancer via Chinese Herbal Medicine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9240426. [PMID: 31583051 PMCID: PMC6754955 DOI: 10.1155/2019/9240426] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Recently, reactive oxygen species (ROS), a class of highly bioactive molecules, have been extensively studied in cancers. Cancer cells typically exhibit higher levels of basal ROS than normal cells, primarily due to their increased metabolism, oncogene activation, and mitochondrial dysfunction. This moderate increase in ROS levels facilitates cancer initiation, development, and progression; however, excessive ROS concentrations can lead to various types of cell death. Therefore, therapeutic strategies that either increase intracellular ROS to toxic levels or, conversely, decrease the levels of ROS may be effective in treating cancers via ROS regulation. Chinese herbal medicine (CHM) is a major type of natural medicine and has greatly contributed to human health. CHMs have been increasingly used for adjuvant clinical treatment of tumors. Although their mechanism of action is unclear, CHMs can execute a variety of anticancer effects by regulating intracellular ROS. In this review, we summarize the dual roles of ROS in cancers, present a comprehensive analysis of and update the role of CHM—especially its active compounds and ingredients—in the prevention and treatment of cancers via ROS regulation and emphasize precautions and strategies for the use of CHM in future research and clinical trials.
Collapse
|
42
|
Koczurkiewicz P, Klaś K, Grabowska K, Piska K, Rogowska K, Wójcik‐Pszczoła K, Podolak I, Galanty A, Michalik M, Pękala E. Saponins as chemosensitizing substances that improve effectiveness and selectivity of anticancer drug—Minireview of in vitro studies. Phytother Res 2019; 33:2141-2151. [DOI: 10.1002/ptr.6371] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Paulina Koczurkiewicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Katarzyna Klaś
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Karolina Grabowska
- Department of Pharmacognosy, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Kamil Piska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Katarzyna Rogowska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Katarzyna Wójcik‐Pszczoła
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Irma Podolak
- Department of Pharmacognosy, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Agnieszka Galanty
- Department of Pharmacognosy, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| | - Marta Michalik
- Department of Cell Biology, Faculty of Biophysics, Biochemistry and BiotechnologyJagiellonian University Kraków Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy Medical CollegeJagiellonian University Kraków Poland
| |
Collapse
|
43
|
Xu C, Wang W, Wang B, Zhang T, Cui X, Pu Y, Li N. Analytical methods and biological activities of Panax notoginseng saponins: Recent trends. JOURNAL OF ETHNOPHARMACOLOGY 2019; 236:443-465. [PMID: 30802611 DOI: 10.1016/j.jep.2019.02.035] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 02/02/2019] [Accepted: 02/19/2019] [Indexed: 05/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Panax notoginseng (Burk.) F. H. Chen, also called Sanqi, is a widely used traditional Chinese medicine, which has long history used as herbal medicines. It is currently an important medicinal material in China, holding the first place in the sale volume of the whole patent medicines market in China, and the market size of the single species has exceeded 10 billion yuan. In addition, P. notoginseng is an important constituent part of many famous Chinese patent medicines, such as Compound Danshen Dripping Pills and Yunnan Baiyao. P. notoginseng saponins (PNSs), which are the major active components of P. notoginseng, are a kind of chemical mixture containing different dammarane-type saponins. Many studies show that PNSs have been extensively used in medical research or applications, such as atherosclerosis, diabetes, acute lung injury, cancer, and cardiovascular diseases. In addition, various PNS preparations, such as injections and capsules, have been made commercially available and are widely applied in clinical practice. AIM OF THE REVIEW Since the safety and efficacy of compounds are related to their qualitative and quantitative analyses, this review briefly summarizes the analytic approaches for PNSs and their biological effects developed in the last decade. METHODOLOGY This review conducted a systematic search in electronic databases, such as Pubmed, Google Scholar, SciFinder, ISI Web of Science, and CNKI, since 2009. The information provided in this review is based on peer-reviewed papers and patents in either English or Chinese. RESULTS At present, the chromatographic technique remains the most extensively used approach for the identification or quantitation of PNSs, coupled with different detectors, among which the difference mainly lies in their sensitivity and specificity for analyzing various compounds. It is well-known that PNSs have traditionally strong activity on cardiovascular diseases, such as atherosclerosis, intracerebral hemorrhage, or brain injury. The recent studies showed that PNSs also responded to osteoporosis, cancers, diabetes, and drug toxicity. However, some other studies also showed that some PNSs injections and special PNS components might lead to some biological toxicity under certain dosages. CONCLUSION This review may be used as a basis for further research in the field of quantitative and qualitative analyses, and is expected to provide updated and valuable insights into the potential medicinal applications of PNSs.
Collapse
Affiliation(s)
- Congcong Xu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Weiwei Wang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Bing Wang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiuming Cui
- Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan Province, Kunming 650500, China
| | - Yiqiong Pu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ning Li
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Institute of KPC Pharmaceuticals, Inc., Kunming 650100, China.
| |
Collapse
|
44
|
Lagoa R, Silva J, Rodrigues JR, Bishayee A. Advances in phytochemical delivery systems for improved anticancer activity. Biotechnol Adv 2019; 38:107382. [PMID: 30978386 DOI: 10.1016/j.biotechadv.2019.04.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/23/2019] [Accepted: 04/06/2019] [Indexed: 12/15/2022]
Abstract
Natural compounds have significant anticancer pharmacological activities, but often suffer from low bioavailability and selectivity that limit therapeutic use. The present work critically analyzes the latest advances on drug delivery systems designed to enhance pharmacokinetics, targeting, cellular uptake and efficacy of anticancer phytoconstituents. Various phytochemicals, including flavonoids, resveratrol, celastrol, curcumin, berberine and camptothecins, carried by liposomes, nanoparticles, nanoemulsions and films showed promising results. Strategies to avoid drug metabolism, overcome physiological barriers and achieve higher concentration at cancer sites through skin, buccal, nasal, vaginal, pulmonary and colon targeted delivery are presented. Current limitations, challenges and future research directions are also discussed.
Collapse
Affiliation(s)
- Ricardo Lagoa
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena, Alto do Vieiro, 2411-901 Leiria, Portugal.
| | - João Silva
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena, Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Joaquim Rui Rodrigues
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena, Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Boulevard, Bradenton, FL 34211, USA.
| |
Collapse
|
45
|
Gu Y, Ju A, Jiang B, Zhang J, Man S, Liu C, Gao W. Yiqi Fumai lyophilized injection attenuates doxorubicin-induced cardiotoxicity, hepatotoxicity and nephrotoxicity in rats by inhibition of oxidative stress, inflammation and apoptosis. RSC Adv 2018; 8:40894-40911. [PMID: 35557896 PMCID: PMC9091596 DOI: 10.1039/c8ra07163b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/22/2018] [Indexed: 01/21/2023] Open
Abstract
Doxorubicin (DOX) is one of the most effective antineoplastic drugs, however, its organ toxicity inhibits the clinical utility. This study was aimed at investigating the protective effects of Yiqi Fumai lyophilized injection (YQFM) against DOX-induced tissue injury and exploring the mechanisms which mediated reactive oxygen species (ROS), inflammation and apoptosis. The experiment was as follows: rats were subjected to an intraperitoneal injection (i.p.) of YQFM (0.481 g kg-1, i.p.) for 12 days; DOX (5 mg kg-1, i.p.) was administered on the 4th, 8th and 12th days to achieve a cumulative dose of 15 mg kg-1. Pretreatment of YQFM significantly ameliorated intracellular damage and dysfunction of the heart, liver and kidneys via decreasing activities of injury indexes. The levels of lipid peroxidation and glutathione depletion were clearly reduced following YQFM pretreatment, meanwhile the activities of glutathione peroxidase, superoxide dismutase, and catalase were elevated. Additionally administering YQFM could mitigate the cardiotoxicity, hepatotoxicity and nephrotoxicity via reducing levels of inflammatory factors and decreasing apoptosis. Accordingly, this study indicated that YQFM attenuated DOX-induced toxicity by ameliorating organ function, decreasing ROS production, and preventing excessive inflammation and apoptosis.
Collapse
Affiliation(s)
- Yue Gu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China +86-22-87401895 +86-22-87401895
| | - Aichun Ju
- Tasly Pride Pharmaceutical Company Limited Tianjin 300410 China
| | - Bingjie Jiang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China +86-22-87401895 +86-22-87401895
| | - Jingze Zhang
- Department of Pharmacy, Logistics University of Chinese People's Armed Police Forces Tianjin 300309 China +86-22-84876773
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology Tianjin 300457 China +86-22-60601265
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics Tianjin 300193 China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China +86-22-87401895 +86-22-87401895
| |
Collapse
|
46
|
Production of Minor Ginenosides from Panax notoginseng by Microwave Processing Method and Evaluation of Their Blood-Enriching and Hemostatic Activity. Molecules 2018; 23:molecules23061243. [PMID: 29882854 PMCID: PMC6099712 DOI: 10.3390/molecules23061243] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/19/2018] [Accepted: 05/20/2018] [Indexed: 01/21/2023] Open
Abstract
A green solvent extraction technology involving a microwave processing method was used to increase the content of minor ginsenosides from Panax notoginseng. This article aims to investigate the optimization of preparation of the minor ginsenosides by this microwave processing method using single-factor experiments and response surface methodology (RSM), and discuss the blood-enriching activity and hemostatic activity of the extract of microwave processed P. notoginseng (EMPN) The RSM for production of the minor ginsenosides was based on a three-factor and three-level Box-Behnken design. When the optimum conditions of microwave power, temperature and time were 495.03 W, 150.68 °C and 20.32 min, respectively, results predicted that the yield of total minor ginsenosides (Y9) would be 93.13%. The actual value of Y9 was very similar to the predicted value. In addition, the pharmacological results of EMPN in vivo showed that EMPN had the effect of enriching blood in N-acetylphenylhydrazine (APH) and cyclophosphamide (CTX)-induced blood deficient mice because of the increasing content of white blood cells (WBCs) and hemoglobin (HGB) in blood. Hemostatic activity in vitro of EMPN showed that it had significantly shortened the clotting time in PT testing (p < 0.05). The hemostatic effect of EMPN was mainly caused by its components of Rh4, 20(S)-Rg3 and 20(R)-Rg3. This microwave processing method is simple and suitable to mass-produce the minor ginsenosides from P. notoginseng.
Collapse
|
47
|
Ginsenoside Rg3 Suppresses Proliferation and Induces Apoptosis in Human Osteosarcoma. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4306579. [PMID: 29750154 PMCID: PMC5884244 DOI: 10.1155/2018/4306579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/06/2018] [Accepted: 01/15/2018] [Indexed: 02/07/2023]
Abstract
Osteosarcoma is the most common primary malignancy of bone in children and the elderly. Recently, more and more researches have demonstrated that Ginsenoside Rg3 (Rg3) is involved in chemotherapy resistance in many cancer, making it a promising Chinese herbal monomer for oncotherapy. In this study, we investigated the efficacy of Rg3 in human osteosarcoma cell lines (MG-63, U-2OS, and SaOS-2). Cell proliferation was measured by CCK8 assay. The migration of cells was examined using the scratch assay method. Quantification of apoptosis was assessed further by flow cytometry. In addition, the expression of apoptosis-related genes (caspase9, caspase3, Bcl2, and Bax) were investigated using RT-PCR. We further investigated the protein level expression of Bcl 2, cleaved-caspase3, and PI3K/AKT/mTOR signaling pathway factors by Western blot assay. Our results revealed that Rg3 inhibited the proliferation and migration of human osteosarcoma cells and induced apoptosis in a concentration- and time-dependent manner. Western blot results showed that Rg3 reduced the protein expression of Bcl2 and PI3K/AKT/mTORbut increased the levels of cleaved-caspase3. Therefore, we hypothesized Rg3 inhibits the proliferation of osteosarcoma cell line and induces their apoptosis by affecting apoptosis-related genes (Bcl2, caspase3) as well as the PI3K/AKT/mTOR signaling pathway. To conclude, Rg3 is a new therapeutic agent against osteosarcoma.
Collapse
|