1
|
Zheng Z, Ke L, Ye S, Shi P, Yao H. Pharmacological Mechanisms of Cryptotanshinone: Recent Advances in Cardiovascular, Cancer, and Neurological Disease Applications. Drug Des Devel Ther 2024; 18:6031-6060. [PMID: 39703195 PMCID: PMC11658958 DOI: 10.2147/dddt.s494555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
Cryptotanshinone (CTS) is an important active ingredient of Salvia miltiorrhiza Bge. In recent years, its remarkable pharmacological effects have triggered extensive and in-depth studies. The aim of this study is to retrieve the latest research progress on CTS and provide prospects for future research. The selection of literature for inclusion, data extraction and methodological quality assessment were discussed. Studies included (1) physicochemical and ADME/Tox properties, (2) pharmacological effects and mechanism, (3) conclusion and bioinformatics analysis. A total of 915 titles and abstracts were screened, resulting in 184 papers used in this review; CTS has shown therapeutic effects on a variety of diseases by modulating multiple molecular pathways. For example, CTS primarily targets NF-κB pathway and MAPK pathway to have a therapeutic role in cardiovascular diseases; in cancer, CTS shows superior efficacy through the PI3K/Akt/mTOR pathway and the JAK/STAT pathway; CTS act on the Nrf2/HO-1 pathway to combat neurological diseases. In addition, key targets of CTS were predicted by bioinformatics analysis, referring to disease ontology (DO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analysis, with R Studio; AKT1, MAPK1, STAT3, P53 and EGFR are predicted to be the key targets of CTS against diseases. The key proteins were then docked by Autodock software to preliminarily assess their binding activities. This review provided new insights into research of CTS and its potential applications in the future, and especially the targets and directly binding modes for CTS are waiting to be investigated.
Collapse
Affiliation(s)
- Ziyao Zheng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Liyuan Ke
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Shumin Ye
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Peiying Shi
- Department of Traditional Chinese Medicine Resource and Bee Products, College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 350002, People’s Republic of China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| |
Collapse
|
2
|
Huang DX, Kang X, Jiang LJ, Zhu DL, Yang L, Luo JY, Yang MM, Li W, Wang GP, Wen Y, Huang Z, Tang LJ. Exploring the impact of high-altitude de-acclimatization on renal function: The roles of oxidative and endoplasmic reticulum stress in rat models. Biochem Biophys Res Commun 2024; 708:149770. [PMID: 38518722 DOI: 10.1016/j.bbrc.2024.149770] [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: 01/18/2024] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND High-altitude de-acclimatization (HADA) significantly impacts physiological functions when individuals acclimatize to high altitudes return to lower altitudes. This study investigates HADA's effects on renal function and structure in rats, focusing on oxidative and endoplasmic reticulum stress as potential mechanisms of renal injury. OBJECTIVE To elucidate the pathophysiological mechanisms of renal damage in HADA and evaluate the efficacy of antioxidants Vitamin C (Vit C) and tauroursodeoxycholic acid (TUDCA) in mitigating these effects. METHODS 88 male Sprague-Dawley rats were randomly divided into a control group, a high-altitude (HA) group, a high-altitude de-acclimatization (HADA) group, and a treatment group. The control group was housed in a sea level environment (500 m), while the HA, HADA, and treatment groups were placed in a simulated high-altitude chamber (5000 m) for 90 days. After this period, the HA group completed the modeling phase; the HADA group was further subdivided into four subgroups, each continuing to be housed in a sea level environment for 3, 7, 14, and 30 days, respectively. The treatment group was split into the Vit C group, the TUDCA group, and two placebo groups, receiving medication for 3 consecutive days, once daily upon return to the sea level. The Vit C group received 100 mg/kg Vit C solution via intravenous injection, the TUDCA group received 250 mg/kg TUDCA solution via intraperitoneal injection, and the placebo groups received an equivalent volume of saline similarly. Serum, urine, and kidney tissues were collected immediately after the modeling phase. Renal function and oxidative stress levels were assessed using biochemical and ELISA methods. Renal histopathology was observed with H&E, Masson's trichrome, PAS, and PASM staining. Transmission electron microscopy was used to examine the ultrastructure of glomeruli and filtration barrier. TUNEL staining assessed cortical apoptosis in the kidneys. Metabolomics was employed for differential metabolite screening and pathway enrichment analysis. RESULTS Compared to the control and HA groups, the HADA 3-day group (HADA-3D) exhibited elevated renal function indicators, significant pathological damage, observable ultrastructural alterations including endoplasmic reticulum expansion and apoptosis. TUNEL-positive cells significantly increased, indicating heightened oxidative stress levels. Various differential metabolites were enriched in pathways related to oxidative and endoplasmic reticulum stress. Early intervention with Vit C and TUDCA markedly alleviated renal injury in HADA rats, significantly reducing the number of apoptotic cells, mitigating endoplasmic reticulum stress, and substantially lowering oxidative stress levels. CONCLUSION This study elucidates the pivotal roles of oxidative and endoplasmic reticulum stress in the early-stage renal injury in rats undergoing HADA. Early intervention with the Vit C and TUDCA significantly mitigates renal damage caused by HADA. These findings provide insights into the pathophysiological mechanisms of HADA and suggest potential therapeutic strategies for its future management.
Collapse
Affiliation(s)
- Dong-Xin Huang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Li-Juan Jiang
- Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China
| | - Dan-Ling Zhu
- Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310000, Zhejiang, China
| | - Lin Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Jing-Ya Luo
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Meng-Meng Yang
- Medical Epigenetics Center, Basic Medicine Collage, Chongqing Medical University, Chongqing, 400042, China
| | - Wei Li
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Guo-Ping Wang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yi Wen
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China
| | - Zhu Huang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of Hyperbaric Oxygen, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China.
| | - Li-Jun Tang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China; Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China; Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
3
|
Wang J, Ma R, Wang Y, Zhang S, Wang J, Zheng J, Xue W, Ding X. rhMYDGF Alleviates I/R-induced Kidney Injury by Inhibiting Inflammation and Apoptosis via the Akt Pathway. Transplantation 2023; 107:1729-1739. [PMID: 36698245 PMCID: PMC10358439 DOI: 10.1097/tp.0000000000004497] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) injury is one of the crucial factors affecting the outcome of renal transplantation. In recent years, myeloid-derived growth factor (MYDGF) has received a lot of attention for its extensive beneficial effects on cardiac repair and protection of cardiomyocytes from cell death. Therefore, we hypothesized that the recombinant human MYDGF (rhMYDGF) protein might play an essential role in safeguarding renal I/R injury. METHODS In vivo experiments were conducted using a mouse unilateral I/R model. Mice were pretreated with rhMYDGF by intraperitoneal injection to study the potential mechanism of renal protection. In vitro, we established hypoxia/reoxygenation and H 2 O 2 treatment models to pretreat cells with rhMYDGF. The expression levels of oxidative stress, inflammation, and apoptosis-related factors in tissues and cells were detected. Finally, we explored the role of the protein kinase B (Akt) pathway in the renal protective mechanism of rhMYDGF. RESULTS In this study, we found that intraperitoneal injection of 1.25 μg rhMYDGF could significantly improve renal function of I/R mice, and reduce oxidative stress, inflammation, and apoptosis. For the human proximal tubular epithelial cell line and human kidney cell line, pretreatment with 0.3 μg/mL rhMYDGF for 24 h significantly downregulated oxidative stress, inflammation, and apoptosis via the phosphorylation of Akt, which could be ameliorated by LY294002. CONCLUSIONS rhMYDGF protects kidney from I/R injury by attenuating oxidative stress, inflammation, and apoptosis through the activation of the Akt pathway.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ruiyang Ma
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shucong Zhang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiale Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wujun Xue
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Wang Q, Liu Y. Cryptotanshinone ameliorates MPP +-induced oxidative stress and apoptosis of SH-SY5Y neuroblastoma cells: the role of STAT3 in Parkinson's disease. Metab Brain Dis 2022; 37:1477-1485. [PMID: 35396628 DOI: 10.1007/s11011-022-00905-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 01/06/2022] [Indexed: 11/25/2022]
Abstract
Cryptotanshinone (CTN) has shown its neuroprotective and anti-inflammatory qualities in non-genetic mouse model of Alzheimer's disease. According to bioinformatics analysis, CTN and Signal Transducer and Activator of Transcription 3 (STAT3) may interact to form a drug-target network. This study was conducted to identify the role of CTN-STAT3 interaction in Parkinson's disease (PD). PD model was established with MMP+-stimulated SH-SY5Y cells. After pre-treatment with CTN or co-treatment with CTN and STAT3 agonist, MTT assay was performed to observe cell viability; ELISA kit was used to measure the expression level of pro-inflammatory cytokines; DCFH-DA and corresponding assay kits were employed to determine the production of ROS, SOD, CAT and GSH-px; TUNEL assay and western blot were performed to detect cell apoptosis. STAT3 activity was also detected by western blot. Treatment with CTN alone had no impact on SH-SY5Y cell viability, but CTN pre-treatment effectively improved MPP+-induced loss of viability in SH-SY5Y cells. Moreover, pre-treatment with CTN inhibited MPP+-induced oxidative stress, apoptosis and STAT3 activity in SH-SY5Y cells, whereas this inhibitory effect was diminished after additional treatment with STAT3 agonist. CTN ameliorates MPP+-induced oxidative stress and apoptosis of SH-SY5Y neuroblastoma cells by inhibiting the expression of STAT3. Therefore, CTN could be a promising therapeutic agent, and STAT3 could be a potential target for PD treatment.
Collapse
Affiliation(s)
- Quanzhe Wang
- Department of Pharmacy, The Third Affiliated Hospital of Baotou Medical College, No.16 Tuanjie Street, Qingshan District, Baotou City, 014030, Inner Mongolia, China
| | - Yan Liu
- Department of Pharmacy, The Third Affiliated Hospital of Baotou Medical College, No.16 Tuanjie Street, Qingshan District, Baotou City, 014030, Inner Mongolia, China.
| |
Collapse
|
5
|
Jia Y, Kang X, Tan L, Ren Y, Qu L, Tang J, Liu G, Wang S, Xiong Z, Yang L. Nicotinamide Mononucleotide Attenuates Renal Interstitial Fibrosis After AKI by Suppressing Tubular DNA Damage and Senescence. Front Physiol 2021; 12:649547. [PMID: 33833691 PMCID: PMC8021789 DOI: 10.3389/fphys.2021.649547] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/01/2021] [Indexed: 01/20/2023] Open
Abstract
Acute kidney injury (AKI) is a worldwide health problem currently lacking therapeutics that directly promote renal repair or prevent the occurrence of chronic fibrosis. DNA damage is a feature of many forms of kidney injury, and targeting DNA damage and repair might be effective strategies for kidney protection in AKI. Boosting nicotinamide adenine dinucleotide (NAD+) levels is thought to have beneficial effects on DNA damage repair and fibrosis in other organs. However, no kidney-related studies of such effects have been performed to date. Here, we have shown that NMN (an NAD+ precursor) administration could significantly reduce tubular cell DNA damage and subsequent cellular senescence induced by hydrogen peroxide and hypoxia in human proximal tubular cells (HK-2 cells). The DNA damage inhibition, antiaging and anti-inflammatory effects of NMN were further confirmed in a unilateral ischemia-reperfusion injury (uIRI) mouse model. Most importantly, the antifibrosis activity of NMN was also shown in ischemic AKI mouse models, regardless of whether NMN was administered in advance or during the recovery phase. Collectively, these results suggest that NMN could significantly inhibit tubular cell DNA damage, senescence and inflammation. NMN administration might be an effective strategy for preventing or treating kidney fibrosis after AKI.
Collapse
Affiliation(s)
- Yan Jia
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xin Kang
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| | - Lishan Tan
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yifei Ren
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| | - Lei Qu
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| | - Jiawei Tang
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| | - Gang Liu
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| | - Suxia Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Li Yang
- Key Laboratory of Renal Disease, Renal Division, Department of Medicine, Peking University First Hospital, Peking University Institute of Nephrology, Ministry of Health of China, Beijing, China
| |
Collapse
|
6
|
Wang H, Pang W, Xu X, You B, Zhang C, Li D. Cryptotanshinone Attenuates Ischemia/Reperfusion-induced Apoptosis in Myocardium by Upregulating MAPK3. J Cardiovasc Pharmacol 2021; 77:370-377. [PMID: 33662979 DOI: 10.1097/fjc.0000000000000971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/26/2020] [Indexed: 01/17/2023]
Abstract
ABSTRACT Chinese people have used the root of Salvia miltiorrhiza Bunge (called "Danshen" in Chinese) for centuries as an anticancer agent, anti-inflammatory agent, antioxidant, and cardiovascular disease drug. In addition, Danshen is considered to be a drug that can improve ischemia/reperfusion (I/R)-induced myocardium injury in traditional Chinese medicine. However, Danshen is a mixture that includes various bioactive substances. In this study, we aimed to identify the protective component and mechanism of Danshen on myocardium through network pharmacology and molecular simulation methods. First, cryptotanshinone (CTS) was identified as a potential active compound from Danshen that was associated with apoptosis by a network pharmacology approach. Subsequently, biological experiments validated that CTS inhibited ischemia/reperfusion-induced cardiomyocyte apoptosis in vivo and in vitro. Molecular docking techniques were used to screen key target information. Based on the simulative results, MAPKs were verified as well-connected molecules of CTS. Western blotting assays also demonstrated that CTS could enhance MAPK expression. Furthermore, we demonstrated that inhibition of the MAPK pathway reversed the CTS-mediated effect on cardiomyocyte apoptosis. Altogether, our work screened out CTS from Danshen and demonstrated that it protected cardiomyocytes from apoptosis.
Collapse
Affiliation(s)
- Hefeng Wang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Wenhui Pang
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xingsheng Xu
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Beian You
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Cuijuan Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| | - Dan Li
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| |
Collapse
|
7
|
Li Y, Han Q, Zhao H, Guo Q, Zhang J. Napabucasin Reduces Cancer Stem Cell Characteristics in Hepatocellular Carcinoma. Front Pharmacol 2020; 11:597520. [PMID: 33343368 PMCID: PMC7744694 DOI: 10.3389/fphar.2020.597520] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. Cancer stem cells (CSCs) are a rare population with self-renewal and multipotent differentiation capacity, and reside among the more differentiated cancer cells. CSCs are associated with tumor recurrence, drug resistance and poor prognosis. The aim of this study was to determine the efficacy of napabucasin against HCC and elucidate the underlying molecular mechanisms. Napabucasin significantly decreased the viability of HCC cells in vitro by inducing apoptosis and cell cycle arrest. In addition, it suppressed CSC-related gene expression and spheroid formation in vitro, indicating depletion of CSCs. The anti-neoplastic effects of napabucasin was also evident in homograft tumor-bearing mouse models. Our findings provide the scientific basis of conducting clinical trials on napabucasin as a new therapeutic agent against HCC.
Collapse
Affiliation(s)
- Ya Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Quanjuan Guo
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
8
|
Zhang Y, Luo F, Zhang H, He W, Liu T, Wu Y, Zhang L, Shi G. Cryptotanshinone ameliorates cardiac injury and cardiomyocyte apoptosis in rats with coronary microembolization. Drug Dev Res 2020; 82:581-588. [PMID: 33340142 DOI: 10.1002/ddr.21777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/15/2020] [Accepted: 11/28/2020] [Indexed: 01/08/2023]
Abstract
Coronary microembolization (CME) is a prevalent cardiovascular disease, especially nowadays when percutaneous coronary intervention is widely applied. However, neither cardio-protective agents nor devices for distal protection could effectively prevent the occurrence of CME. Therefore, we aimed to develop a new drug for CME. Rats were orally administrated with different doses of Cryptotanshinone (CTS, 5, 15, 45 mg/kg) daily for 2 weeks, respectively, following CME surgery. Then cardiac function and cardiac injury were evaluated in CME rats as well as measuring oxidative stress and apoptosis in cardiomyocytes. Compared to sham group, CME operation induced cardiac dysfunction, cardiac injury, the activation of platelet and endothelium, cardiomyocyte apoptosis and oxidative stress, all of which could be dose-dependently restored by CTS pretreatment. Moreover, NF-κB signaling pathway participated in the development of CME and also in the preventive process of CTS against CME. CTS might serve as a potential and promising candidate drug to prevent the occurrence of CME.
Collapse
Affiliation(s)
- Yongpeng Zhang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Fei Luo
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Haixia Zhang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Wei He
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Ting Liu
- Department of Gynecology, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| | - Yue Wu
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, Hebei, China
| | - Gang Shi
- Department of Cardiovascular Medicine, Pingxiang People's Hospital, Pingxiang, Jiangxi, China
| |
Collapse
|
9
|
Shi G, Wang Y, Yang J, Liu T, Luo F, Jin G, Ma Y, Zhang Y. Effect of Cryptotanshinone on Measures of Rat Cardiomyocyte Oxidative Stress and Gene Activation Associated with Apoptosis. Cardiorenal Med 2020; 11:18-26. [PMID: 33326961 DOI: 10.1159/000507184] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/11/2020] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Oxidative stress is a key factor that results in cardiomyocyte apoptosis and cardiovascular diseases. Cryptotanshinone (CTS), one of the major bioactive constitutes extracted from the root of the plant Salvia miltiorrhizaBunge, has been widely studied for various disease treatments. However, the roles of CTS on cardiomyocytes remain unclear. METHODS In the present study, neonatal rat cardiomyocytes were pretreated with CTS for 4 h before being exposed to H2O2. Cell viability for the cells with or without pretreatment with CTS before exposure to H2O2 was evaluated by the MTT assay. Production of lactate dehydrogenase (LDH), nitric oxide (NO), prostaglandin E2 (PGE2), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxides (GSH-Px) was quantified by corresponding detection kits. The mRNA levels of Bcl-2 antiapoptotic and Bax-like proapoptotic genes were quantified with RT-PCR. Production of reactive oxygen species (ROS) was qualified and quantified with a dichlorofluorescein diacetate cellular ROS detection assay kit. The extracellular signal-related kinase (ERK) phosphorylation and nuclear factor κB (NF-κB) activation were measured by Western blot. RESULTS Our results revealed that the CTS pretreatment could enhance cell viability and promote Bcl-2 antiapoptotic gene expression. Additionally, CTS could abolish the H2O2-induced production of NO, LDH, and PGE2. Consistent with these findings, CTS could inhibit ROS and MDA production and promote SOD, CAT, and GSH-Px activities. Mechanistically, CTS may achieve these processes by inhibiting ERK and NF-κB signal pathways. CONCLUSION CTS protects cardiomyocytes against the H2O2-induced cellular injuries through ERK and NF-κB inactivation and ROS scavenging. Therefore, CTS is a promising reagent against ROS-induced cardiomyopathy.
Collapse
Affiliation(s)
- Gang Shi
- Department of Cardiovascular Medicine, Pingxiang People's Hospital, Pingxiang, China
| | - Ying Wang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China
| | - Jie Yang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China
| | - Ting Liu
- Department of Gynecology, Pingxiang People's Hospital, Pingxiang, China
| | - Fei Luo
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China
| | - Guoyin Jin
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China
| | - Yuan Ma
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China
| | - Yongpeng Zhang
- College of Traditional Chinese Medicine, Hebei North University, Zhangjiakou, China,
| |
Collapse
|
10
|
Liu B, Deng Q, Zhang L, Zhu W. Nobiletin alleviates ischemia/reperfusion injury in the kidney by activating PI3K/AKT pathway. Mol Med Rep 2020; 22:4655-4662. [PMID: 33173956 PMCID: PMC7646848 DOI: 10.3892/mmr.2020.11554] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022] Open
Abstract
Recent studies have demonstrated that nobiletin (NOB) displays anti-oxidative and anti-apoptotic efficacies against multiple pathological insults. However, the potential effects of NOB on the injury caused by ischemia and reperfusion (I/R) in the kidney remain undetermined. In the present study, I/R injury was elicited by right kidney removal and left renal pedicel clamping for 45 min, followed by reperfusion for 24 h. NOB was added at the start of reperfusion. Histological examination, detection of biomarkers in plasma, and measurement of apoptosis induced by endoplasmic reticulum stress (ERS) were used to evaluate renal injury. Additionally, the PI3K/AKT inhibitor LY294002 was also used in mechanistic experiments. NOB pre-treatment significantly reduced renal damage caused by I/R injury, as indicated by decreased serum levels of creatine, blood urea nitrogen and tubular injury scores. Furthermore, NOB inhibited elevated ERS-associated apoptosis, as evidenced by reduced apoptotic rates and ERS-related signaling molecules (such as, C/EBP homologous protein, caspase-12 and glucose-regulated protein of 78 kDa). NOB increased phosphorylation of proteins in the PI3K/AKT pathway. The inhibition of PI3K/AKT signaling with pharmacological inhibitors could reverse the beneficial effects of NOB during renal I/R insult. In conclusion, NOB pre-treatment may alleviate I/R injury in the kidney by inhibiting reactive oxygen species production and ERS-induced apoptosis, partly through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Bo Liu
- Department of Urology, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Quanhong Deng
- Department of Urology, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Lei Zhang
- Department of Urology, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Wen Zhu
- Department of Urology, Jingmen No. 2 People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
11
|
Wu YH, Wu YR, Li B, Yan ZY. Cryptotanshinone: A review of its pharmacology activities and molecular mechanisms. Fitoterapia 2020; 145:104633. [DOI: 10.1016/j.fitote.2020.104633] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
|
12
|
Liu Y, Wang H, Gao J, Wen Z, Peng L. Cryptotanshinone ameliorates the pathogenicity of Streptococcus suis by targeting suilysin and inflammation. J Appl Microbiol 2020; 130:736-744. [PMID: 32750224 DOI: 10.1111/jam.14810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022]
Abstract
AIMS Streptococcus suis is a highly zoonotic pathogen that is a serious threat to human health and the development of the pig industry worldwide. The virulence factors produced during S. suis infection play an important role, and the pore-forming activity of suilysin is considered an important virulence-related factor, especially in meningitis. Treatment of S. suis infection with traditional antibiotics is becoming increasingly challenging due to bacterial resistance. The purpose of this study is to verify the role of cryptotanshinone in the process of S. suis infection and provide a new drug precursor for the treatment of S. suis infection. METHODS AND RESULTS In this study, we used circular dichroism spectroscopy to demonstrate that cryptotanshinone alters the secondary structure of suilysin. The results of the antibacterial activity and haemolysis assays showed cryptotanshinone could inhibit the pore-forming activity of suilysin without affecting bacterial growth or its expression. We also showed that cryptotanshinone reduces bacterial damage and penetration in vitro, reduce the S. suis-induced inflammatory response and provide protection against bacterial infections in vivo and in vitro. CONCLUSIONS Cryptotanshinone is a potential compound precursor for treating S. suis infection. SIGNIFICANCE AND IMPACT OF THE STUDY Cryptotanshinone may be a promising leading compound for S. suis infection and related diseases.
Collapse
Affiliation(s)
- Y Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - H Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - J Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Z Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - L Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Xu Y, Niu Y, Li H, Pan G. Downregulation of lncRNA TUG1 attenuates inflammation and apoptosis of renal tubular epithelial cell induced by ischemia-reperfusion by sponging miR-449b-5p via targeting HMGB1 and MMP2. Inflammation 2020; 43:1362-1374. [PMID: 32206944 DOI: 10.1007/s10753-020-01214-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We aimed to evaluate the functions of long non-coding RNA taurine upregulated gene 1 (lncRNA TUG1) in renal ischemia-reperfusion (I/R) injury and identify the potential mechanisms. Pathological changes of renal tissues were examined using H&E staining after mimic renal I/R injury in vivo. The contents of serum renal functional parameters and inflammatory factors were measured. The expression of TUG1 and miR-449b-5p in renal tissues and HK-2 cells stimulated by I/R were detected. Then, the effects of TUG1 silencing on inflammation and apoptosis of cells were evaluated. Dual luciferase reporter assays were executed for determining the correlation between miR-449b-5p and TUG1, high mobility group box 1 (HMGB1), or matrix metalloproteinase 2 (MMP2). Subsequently, cells were co-transfected with miR-449b-5p mimic and pcDNA3.1 TUG1. The levels of inflammation, apoptosis, and the expression of HMGB1 and MMP2 were detected. The results revealed that renal tissues were obviously damaged after I/R accompanied by changes in renal functional markers and inflammatory factors. TUG1 was highly expressed whereas miR-449b-5p was lowly expressed. TUG1 silencing reduced the inflammation and apoptosis. Dual luciferase reporter assays confirmed that miR-449b-5p was a target of TUG1 as well as HMGB1 and MMP2 were direct targets of miR-449b-5p. Meanwhile, miR-449b-5p mimic presented the same results with TUG1 silencing, which were reversed after TUG1 overexpression. Moreover, MMP2 and HMGB1 expression was decreased after miR-449b-5p overexpression while that of was increased after TUG1 overexpression. These findings demonstrated that TUG1 silencing attenuates I/R-induced inflammation and apoptosis via targeting miR-449b-5p and regulating HMGB1 and MMP2 expression.
Collapse
Affiliation(s)
- Yuan Xu
- Department of organ transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China
| | - Yulin Niu
- Department of organ transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China
| | - Haiyang Li
- Department of hepatobiliary surgery, Affiliated Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China.
| | - Guanghui Pan
- Department of organ transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang City, 550004, Guizhou Province, China.
| |
Collapse
|
14
|
Liu B, Yan L, Jiao X, Sun X, Zhao Z, Yan J, Guo M, Zang Y. Lycopene Alleviates Hepatic Hypoxia/Reoxygenation Injury Through Nrf2/HO-1 Pathway in AML12 Cell. J Interferon Cytokine Res 2020; 40:406-417. [PMID: 32813603 DOI: 10.1089/jir.2020.0038] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Bing Liu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lihong Yan
- The Library of the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xuefei Jiao
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaozhi Sun
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zonggang Zhao
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Junwei Yan
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mingjin Guo
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yunjin Zang
- Institute of Transplantation Science, Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
15
|
miR-10a overexpression aggravates renal ischemia-reperfusion injury associated with decreased PIK3CA expression. BMC Nephrol 2020; 21:248. [PMID: 32611384 PMCID: PMC7329557 DOI: 10.1186/s12882-020-01898-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/18/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND To investigate the effect of miR-10a on renal tissues with ischemia reperfusion (I/R) injury in rats and to explore the underlying mechanisms of the effect of miR-10a on hypoxia-reoxygenation in HK-2 cells. METHODS MiR-10a level was measured in the renal tissues of rats with I/R rats using RT-PCR. In order to research the role of miR-10a in renal tissues, an miR-10 agonist and an miR-10a antagonist were used to treat I/R-injured rats. Levels of serum creatinine and blood urea nitrogen, renal histopathology, and levels of cell apoptosis were analyzed separately in renal tissues from the rats. Phosphatidylinositol 3-kinase (PI3K)/Akt pathway related proteins were measured by Western blotting. In addition, HK-2 cells were cultured in order to study the mechanism of action of miR-10a in the hypoxia-reoxygenation model being studied. Finally, the dual luciferase reporter gene assay was used to confirm that the PI3K p100 catalytic subunit α (PIK3CA) gene was targeted by miR-10a. RESULTS After renal I/R injury in rats, miR-10a expression increased significantly (p < 0.05). Injection of miR-10a agonist significantly aggravated the renal injury and raised the level of cell apoptosis in the renal tissues of I/R-injured rats (p < 0.05). However, administration of miR-10a antagonist led to obvious improvement of the renal injury, decreased renal cell apoptosis, and inhibited PI3K/Akt pathway activity (p < 0.05). In in vitro experiments, the negative relationship between PIK3CA and miR-10a levels was confirmed. Furthermore, overexpression of miR-10a significantly decreased the proliferation of HK-2 cells, and increased cell apoptosis via up-regulation of the PI3K/Akt pathway (p < 0.05). CONCLUSION The aggravation of renal I/R injury by miR-10a was associated with a decrease in the activity of PIK3CA/PI3K/Akt pathway.
Collapse
|
16
|
Wang N, Dong X, Shi D, Li N, Zhang Q. Cryptotanshinone ameliorates placental oxidative stress and inflammation in mice with gestational diabetes mellitus. Arch Pharm Res 2020; 43:755-764. [PMID: 32601882 DOI: 10.1007/s12272-020-01242-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Gestational diabetes mellitus (GDM) is a conditional diabetes which is defined as any degree of glucose intolerance or high blood glucose levels during any phase of pregnancy. It causes chronic severe damage to health of the pregnant women and their offspring. In this study, we aimed to study the protective effects of Cryptotanshinone on GDM-related impairments. We measured blood glucose levels, serum insulin levels, biochemical indexes, oxidative stress, inflammation and the activation of NF-κB signaling pathway in the blood and placenta of GDM mice. It is found that Cryptotanshinone significantly decreased blood glucose levels, oxidative stress, inflammation and NF-κB signaling with an increase of serum insulin levels in the placenta and blood of GDM mice. Taken together, Cryptotanshinone effectively ameliorated GDM, which suggested that Cryptotanshinone could be served as a promising therapeutic drug for GDM patients.
Collapse
Affiliation(s)
- Na Wang
- Department of Obstetrics, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000, Hebei, China.
| | - Xiujuan Dong
- Department of Obstetrics, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000, Hebei, China
| | - Dandan Shi
- Department of Obstetrics, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000, Hebei, China
| | - Na Li
- Department of Obstetrics, Cangzhou Central Hospital, No. 16 Xinhua Western Road, Cangzhou, 061000, Hebei, China
| | - Qun Zhang
- Dermatological Department, Cangzhou People's Hospital, No. 7 Qingchi Avenue, Cangzhou, 061000, Hebei, China
| |
Collapse
|