1
|
Sun J, Wei Y, Wang J, Hou M, Su L. Treatment of colorectal cancer by traditional Chinese medicine: prevention and treatment mechanisms. Front Pharmacol 2024; 15:1377592. [PMID: 38783955 PMCID: PMC11112518 DOI: 10.3389/fphar.2024.1377592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Colorectal cancer (CRC) is a significant global health burden, with high morbidity and mortality rates. It is often diagnosed at middle to advanced stage, affecting approximately 35% of patients at the time of diagnosis. Currently, chemotherapy has been used to improve patient prognosis and increase overall survival. However, chemotherapy can also have cytotoxic effects and lead to adverse reactions, such as inhibiting bone marrow hematopoiesis, causing digestive dysfunction, hand-foot syndrome, and even life-threatening conditions. In response to these adverse effects, researchers have proposed using Traditional Chinese Medicine (TCM) as an option to treat cancer. TCM research focuses on prescriptions, herbs, and components, which form essential components of the current research in Chinese medicine. The study and implementation of TCM prescriptions and herbs demonstrate its distinctive holistic approach to therapy, characterized by applying multi-component and multi-target treatment. TMC components have advantages in developing new drugs as they consist of single ingredients, require smaller medication dosages, have a precise measure of pharmacodynamic effects, and have a clear mechanism of action compared to TCM prescriptions and herbs. However, further research is still needed to determine whether TMC components can fully substitute the therapeutic efficacy of TCM prescriptions. This paper presents a comprehensive analysis of the research advancements made in TCM prescriptions, herbs, and components. The findings of this study can serve as a theoretical basis for researchers who are interested in exploring the potential of TCM for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Jiaxin Sun
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| | - Ying Wei
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| | - Jia Wang
- Department of Gynaecology, Inner Mongolia People’s Hospital, Hohhot, Inner Mongolia, China
| | - Mingxing Hou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Liya Su
- Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| |
Collapse
|
2
|
Investigation of the Effects of Glabridin on the Proliferation, Apoptosis, and Migration of the Human Colon Cancer Cell Lines SW480 and SW620 and Its Mechanism Based on Reverse Virtual Screening and Proteomics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:1117431. [PMID: 36644579 PMCID: PMC9836797 DOI: 10.1155/2023/1117431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 10/08/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023]
Abstract
Colon cancer is a relatively common malignant tumor of the digestive tract. Currently, most colon cancers originate from adenoma carcinogenesis. By screening various licorice flavonoids with anticancer effects, we found that glabridin (GBN) has a prominent anticolon cancer effect. First, we initially explored whether GBN can inhibit proliferation, migration, and invasion and induce apoptosis in SW480 and SW620 cells. Next, we exploited reverse virtual and proteomics technologies to screen out closely related target pathways on the basis of a drug and target database. At the same time, we constructed the structure of the GBN target pathway in colon cancer. We predicted that GBN can regulate the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-mammalian target of the rapamycin pathway (mTOR) pathway to fight colon cancer. Finally, through Western blot analysis and qRT-PCR, we verified that the expression levels of the PI3K, AKT, and mTOR proteins and genes in this pathway were significantly reduced after GBN administration. In short, the promising discovery of the anticolon cancer mechanism of GBN provides a reliable experimental basis for subsequent new drug development.
Collapse
|
3
|
Abstract
Han N, Zhang B, Wei X, Yu L. The inhibitory function of icariin in cell model of benign prostatic hyperplasia by upregulation of miR-7. BioFactors. 2023;49:203. https://doi.org/10.1002/biof.1591 This article, published online on 29 November 2019 in Wiley Online Library, has been retracted by agreement between the International Union of Biochemistry and Molecular Biology, the Editor in Chief (Dr. Angelo Azzi), and Wiley Periodicals LLC. The retraction has been agreed following an investigation based on allegations raised by a third party. Evidence for image manipulation was found in figures 1, 3, 4, 5, 6, and 7. As a result, the conclusions of this article are considered to be invalid.
Collapse
|
4
|
Zhou L, Yan K, Xing S, Cheng J. Tectorigenin alleviates the apoptosis and inflammation in spinal cord injury cell model through inhibiting insulin-like growth factor-binding protein 6. Open Med (Wars) 2023; 18:20230680. [PMID: 37069938 PMCID: PMC10105551 DOI: 10.1515/med-2023-0680] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/05/2023] [Accepted: 02/23/2023] [Indexed: 04/19/2023] Open
Abstract
Since tectorigenin has been reported to possess anti-inflammation, redox balance restoration, and anti-apoptosis properties, we determine to unravel whether tectorigenin has potential in alleviating spinal cord injury (SCI). Herein, PC12 cells were induced by lipopolysaccharide (LPS) to establish in vitro SCI models. The cell viability and apoptosis were detected through cell counting kit-8 and flow cytometry assays. The caspase-3/8/9 content was measured by colorimetric method. Western blot was conducted to quantify the expressions of cleaved caspse-3/8/9, IGFBP6, TLR4, IκBα, p-IκBα, RELA proto-oncogene, p65, and p-p65. Enzyme-linked immunosorbent assay and real-time quantitative polymerase chain reaction were carried out to quantitate expressions of IGFBP6, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). SwissTargetPrediction and GSE21497 database were utilized to predict the potential therapeutic targets of tectorigenin. Comparison of IGFBP6 expression in SCI tissues and normal tissues was analyzed by GEO2R. Our study found that LPS induced the declined cell viability, elevated cell apoptosis, upregulation of caspase-3/8/9, cleaved caspase-3/8/9, IL-1β, IL-6, TNF-α, IGFBP6, and TLR4, and the activation of IκBα and p65 in PC12 cells. Tectorigenin reversed the above effects of LPS. IGFBP6 was predicted to be the potential therapeutic target of tectorigenin and was overexpressed in SCI tissues. Notably, IGFBP6 overexpression offset the effects of tectorigenin on PC12 cells. In conclusion, tectorigenin could alleviate the LPS-induced apoptosis, inflammation, and activation of NF-κB signaling in SCI cell models via inhibiting IGFBP6.
Collapse
Affiliation(s)
- Liqiang Zhou
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, 611130, China
| | - Kui Yan
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, 611130, China
| | - Shuxing Xing
- Department of Orthopedics, Chengdu Fifth People’s Hospital, No. 33 Mashi Street, Wenjiang
District, Chengdu, Sichuan Province, 611130, China
| | - Jun Cheng
- Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, 611130, China
| |
Collapse
|
5
|
LI M, ZHENG J, WANG S, CHEN L, PENG X, CHEN J, AN H, HU B. Tenglong Buzhong granules inhibits the growth of SW620 human colon cancer. J TRADIT CHIN MED 2022; 42:701-706. [PMID: 36083476 PMCID: PMC9924744 DOI: 10.19852/j.cnki.jtcm.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
OBJECTIVE To observe the anticancer effects of the granular preparation of Tenglong Buzhong decoction (,TBD), i.e Tenglong Buzhong granules (, TBG), in human SW620 colon cancer. METHODS BALB/c nude mice were subcutaneously transplanted with SW620 cells, and treated with TBG (2.56 g/kg, once per day) and/or 5-Fu (104 mg/kg, once per week) for 21 d. Apoptosis, Caspase activities and cellular senescence were measured by commercial kits. The protein expression and phosphorylation were detected by Western blot or immunohistochemistry. RESULTS TBG and 5-Fu inhibited tumor growth. The tumor inhibition rate of the TBG, 5-Fu, and TBG+5-Fu groups was 42.25%, 51.58%, and 76.08%, respectively. Combination of TBG and 5-Fu showed synergetic anti-cancer effects. TBG and 5-Fu induced apoptosis, activated caspase-3, -8, and -9, increased SMAC expression, inhibited XIAP expression. TBG induced cellular senescence, upregulated cyclin-dependent kinase inhibitor 1a (CDKN1a) and cyclin-dependent kinase inhibitor 2a (CDKN2a) expression, and inhibited phosphorylation of retinoblastoma-associated protein (RB) and expression of cyclin E1 (CCNE1) and cyclin-dependent kinases (CDK) 2. TBG also inhibited angiogenesis accompanied by downregulation of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α). CONCLUSIONS TBG inhibits SW620 colon cancer growth, induces apoptosis SMAC-XIAP-Caspases signaling, induces cellular senescence through CDKN1a/CDKN2a-RB-E2F signaling, inhibits angiogenesis by down-regulation of HIF-1α and VEGF, and enhances the effects of 5-Fu.
Collapse
Affiliation(s)
- Miao LI
- 1 Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jialu ZHENG
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Shuangshuang WANG
- 3 Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lei CHEN
- 1 Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiao PENG
- 1 Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jinfang CHEN
- 1 Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Hongmei AN
- 4 Department of Science and Technology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Bing HU
- 1 Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- 2 Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Dr. HU Bing, Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China. Telephone: +86-21-64385700
| |
Collapse
|
6
|
Based on the Network Pharmacology to Investigate the Mechanism of Qingjie Fuzheng Granules against Colorectal Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7242640. [PMID: 35280511 PMCID: PMC8916896 DOI: 10.1155/2022/7242640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/20/2022] [Accepted: 02/10/2022] [Indexed: 01/19/2023]
Abstract
Qingjie Fuzheng granules (QFG) exert an anticancer effect against colorectal cancers (CRC). However, the pharmacological molecular mechanisms are still unclear. This study was aimed to establish a simple method to predict targets of QFG against CRC by the network pharmacology strategy. 461 compounds and 1559 targets in QFG were enriched by BATMAN-TCM. 21 of the common targets were obtained by the groups of “Jun,” “Chen,” “Zuo,” and “Shi” medicine in QFG. The enrichment analyses of GO functional terms, KEGG pathway, and OMIM/TTD diseases displayed the targets in the different and complementary effects of four functional medicines in QFG. Then, 613 differential targets for QFG in CRC were identified. GO functional terms and KEGG pathway analyses showed that QFG regulated the inflammatory function and lipid metabolic process. There were also targets that played a role in the binding to the receptors in membranes, in the activation of the transportation signal, and provided pain relief by regulation of the neural related pathways. Next, the protein-protein interaction network was analyzed, and the levels of the predicted targets in CRC primary tumor were explored, and 7 candidate targets of QFG against CRC were obtained. Furthermore, with real-time PCR and enzyme-linked immunosorbent assay (ELISA) analysis, downregulation of dopamine D2 receptor (DRD2) and interleukin-6 (IL-6), and upregulation of interleukin-10 (IL-10) were identified following the treatment of QFG. At last, the survival and prognosis of the potential targets of QFG in CRC patients were analyzed by GenomicScape, and IL-6 was suggested to be an index for the regulation of QFG in CRC. These results might elucidate the possible antitumor mechanism of QFG and highlight the candidate therapeutic targets and the application direction in clinical treatment for QFG.
Collapse
|
7
|
Qingjie Fuzheng Granule Inhibits EMT and Induces Autophagy in Colorectal Cancer via mTOR Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9950499. [PMID: 34887935 PMCID: PMC8651347 DOI: 10.1155/2021/9950499] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/04/2021] [Accepted: 11/03/2021] [Indexed: 01/10/2023]
Abstract
Qingjie Fuzheng granule (QFG) is a traditional Chinese medicinal formula used extensively as an alternative medicine for cancer treatment, including colorectal cancer (CRC). But its pathological mechanism in CRC is unclear. To study antitumor treatment effects and mechanisms of QFG, we established a CRC HCT-116 xenograft mouse model and assessed QFG on EMT and autophagy progression in vivo. The mice were randomly divided into 2 groups (n = 10 each group) and treated with intragastric administration of 1 g/kg of QFG or saline 6 days a week for 28 days (4 weeks). Body weight was measured every other day with electronic balance. At the end of the treatment, the tumor weight was measured. Immunohistochemical (IHC) and western blot (WB) assay were used to detect the expression level of E-cadherin, N-cadherin, vimentin, and TWIST1 to evaluate the effect of QFG on tumor cell EMT progression. IHC and WB assay were also used to detect the expression level of beclin-1, LC3-II, and p62 to evaluate the effect of QFG on tumor cell autophagy progression. Furthermore, the expression level of relative proteins in mTOR pathway was detected by WB assay to investigate the mechanism of QFG effect on CRC. We discovered that QFG inhibited the rise of tumor weight while it had no effect on mice body weight, which proved that QFG could inhibit CRC growth progression without significant side effects in vivo. In addition, QFG treatment inhibited EMT and induced autophagy progression in CRC tumor cells, including that QFG upregulated the expression of E-cadherin, beclin-1, and LC3-II, but downregulated the expression of N-cadherin, vimentin, TWIST1, and p62. And, QFG decreased the ratio of p-PI3K/PI3K, p-AKT/AKT, and p-mTOR/mTOR, but increased the ratio of p-AMPK/AMPK. All findings from this research proved that QFG can induce autophagy and inhibit EMT progression in CRC via regulating the mTOR signaling pathway.
Collapse
|
8
|
Li N, Yang F, Liu DY, Guo JT, Ge N, Sun SY. Scoparone inhibits pancreatic cancer through PI3K/Akt signaling pathway. World J Gastrointest Oncol 2021; 13:1164-1183. [PMID: 34616521 PMCID: PMC8465440 DOI: 10.4251/wjgo.v13.i9.1164] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic cancer is a highly malignant tumor of the gastrointestinal system whose emerging resistance to chemotherapy has necessitated the development of novel antitumor treatments. Scoparone, a traditional Chinese medicine monomer with a wide range of pharmacological properties, has attracted considerable attention for its antitumor activity. AIM To explore the potential antitumor effect of scoparone on pancreatic cancer and the possible molecular mechanism of action. METHODS The target genes of scoparone were determined using both the bioinformatics and multiplatform analyses. The effect of scoparone on pancreatic cancer cell proliferation, migration, invasion, cell cycle, and apoptosis was detected in vitro. The expression of hub genes was tested using quantitative reverse transcription polymerase chain reaction (qRT-PCR), and the molecular mechanism was analyzed using Western blot. The in vivo effect of scoparone on pancreatic cancer cell proliferation was detected using a xenograft tumor model in nude mice as well as immunohistochemistry. RESULTS The hub genes involved in the suppression of pancreatic cancer by scoparone were obtained by network bioinformatics analyses using publicly available databases and platforms, including SwissTargetPrediction, STITCH, GeneCards, CTD, STRING, WebGestalt, Cytoscape, and Gepia; AKT1 was confirmed using qRT-PCR to be the hub gene. Cell Counting Kit-8 assay revealed that the viability of Capan-2 and SW1990 cells was significantly reduced by scoparone treatment exhibiting IC50 values of 225.2 μmol/L and 209.1 μmol/L, respectively. Wound healing and transwell assays showed that scoparone inhibited the migration and invasion of pancreatic cancer cells. Additionally, flow cytometry confirmed that scoparone caused cell cycle arrest and induced apoptosis. Scoparone also increased the expression levels of Bax and cleaved caspase-3, decreased the levels of MMP9 and Bcl-2, and suppressed the phosphorylation of Akt without affecting total PI3K and Akt. Moreover, compared with the control group, xenograft tumors, in the 200 μmol/L scoparone treatment group, were smaller in volume and lighter in weight, and the percentages of Ki65- and PCNA-positive cells were decreased. CONCLUSION Our findings indicate that scoparone inhibits pancreatic cancer cell proliferation in vitro and in vivo, inhibits migration and invasion, and induces cycle arrest and apoptosis in vitro through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Na Li
- Department of Hospice Care, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning Province, China
| | - Fan Yang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Dong-Yan Liu
- Medical Research Center, Shengjing Hospital of China Medical University, Benxi 111700, Liaoning Province, China
| | - Jin-Tao Guo
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Nan Ge
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Si-Yu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
9
|
Sun Q, He M, Zhang M, Zeng S, Chen L, Zhao H, Yang H, Liu M, Ren S, Xu H. Traditional Chinese Medicine and Colorectal Cancer: Implications for Drug Discovery. Front Pharmacol 2021; 12:685002. [PMID: 34276374 PMCID: PMC8281679 DOI: 10.3389/fphar.2021.685002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
As an important part of complementary and alternative medicine, traditional Chinese medicine (TCM) has been applied to treat a host of diseases for centuries. Over the years, with the incidence rate of human colorectal cancer (CRC) increasing continuously and the advantage of TCM gradually becoming more prominent, the importance of TCM in both domestic and international fields is also growing with each passing day. However, the unknowability of active ingredients, effective substances, and the underlying mechanisms of TCM against this malignant tumor greatly restricts the translation degree of clinical products and the pace of precision medicine. In this review, based on the characteristics of TCM and the oral administration of most ingredients, we herein provide beneficial information for the clinical utilization of TCM in the prevention and treatment of CRC and retrospect the current preclinical studies on the related active ingredients, as well as put forward the research mode for the discovery of active ingredients and effective substances in TCM, to provide novel insights into the research and development of innovative agents from this conventional medicine for CRC treatment and assist the realization of precision medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Sulforaphane Ameliorates the Liver Injury of Traumatic Hemorrhagic Shock Rats. J Surg Res 2021; 267:293-301. [PMID: 34174694 DOI: 10.1016/j.jss.2021.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND The protective effects of sulforaphane on liver injury induced by high-fat diet and sodium valproate were previously reported. The present study preliminarily investigated the effect of sulforaphane on liver injury induced by traumatic hemorrhagic shock. MATERIALS AND METHODS After a traumatic hemorrhagic shock model was established in rats, the survival of rats during the first 24 hours was analyzed by Kaplan-Meier analysis. The serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TB), tumor necrosis factor α (TNF-α), and interleukin 1β (IL-1β) were measured using a biochemical analyzer or enzyme-linked immunosorbent assay (ELISA). The cell apoptosis and histopathology of liver tissues were examined by TUNEL and hematoxylin-eosin (HE) staining. The mRNA and protein expressions of B-cell lymphoma-2 (Bcl-2), Bcl2 associated X (Bax), Caspase-3, TNF-α, IL-1β, Cyclooxygenase-2 (COX-2), nitric oxide synthase (iNOS), nuclear factor E2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1) in the liver tissues were determined by immunohistochemical staining, quantitative reverse transcription PCR (qRT-PCR) or western blot. RESULTS Sulforaphane promoted the health of the animal, reduced liver cell apoptosis and ameliorated the histopathological damage in the liver of rats with traumatic hemorrhagic shock. Sulforaphane downregulated the expressions of liver function-related factors (ALT, AST, TB), inflammation-related factors (TNF-α, IL-1β, COX-2, iNOS), and apoptosis-related factors (Bax, Caspase-3) and upregulated the expressions of factors related to apoptosis (Bcl-2) and Nrf2/HO-1 pathway (Nrf2, HO-1). CONCLUSION Sulforaphane protected the liver against traumatic hemorrhagic shock through ameliorating the apoptosis and inflammation of the liver via activating the Nrf2/HO-1 pathway.
Collapse
|
11
|
The Most Competent Plant-Derived Natural Products for Targeting Apoptosis in Cancer Therapy. Biomolecules 2021; 11:biom11040534. [PMID: 33916780 PMCID: PMC8066452 DOI: 10.3390/biom11040534] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a challenging problem for the global health community, and its increasing burden necessitates seeking novel and alternative therapies. Most cancers share six basic characteristics known as "cancer hallmarks", including uncontrolled proliferation, refractoriness to proliferation blockers, escaping apoptosis, unlimited proliferation, enhanced angiogenesis, and metastatic spread. Apoptosis, as one of the best-known programmed cell death processes, is generally promoted through two signaling pathways, including the intrinsic and extrinsic cascades. These pathways comprise several components that their alterations can render an apoptosis-resistance phenotype to the cell. Therefore, targeting more than one molecule in apoptotic pathways can be a novel and efficient approach for both identifying new anticancer therapeutics and preventing resistance to therapy. The main purpose of this review is to summarize data showing that various plant extracts and plant-derived molecules can activate both intrinsic and extrinsic apoptosis pathways in human cancer cells, making them attractive candidates in cancer treatment.
Collapse
|
12
|
Zheng J, Li X, Yang W, Zhang F. Dihydroartemisinin regulates apoptosis, migration, and invasion of ovarian cancer cells via mediating RECK. J Pharmacol Sci 2021; 146:71-81. [PMID: 33941323 DOI: 10.1016/j.jphs.2021.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 01/18/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Dihydroartemisinin (DHA) possesses an inhibitory effect on ovarian cancer and promotes reversion-inducing cysteine-rich protein with Kazal motifs (RECK) expression in glioma cells. This study explored the role of DHA and RECK on ovarian cancer. METHODS The RECK level in ovarian cancer was analyzed under GEPIA 2 database and proved by RT-qPCR. After being treated with DHA or infected with siRECK lentivirus, the viability, apoptosis, migration, and invasion of ovarian cancer cells were evaluated by CCK-8, flow cytometry, wound healing, and transwell assays. Also, the expressions of factors related to apoptosis and epithelial-mesenchymal transition were measured by Western blot or RT-qPCR. RESULTS DHA-treatment weakened the viability, migration, invasion, and enhanced apoptosis of ovarian cancer cells. DHA also down-regulated the levels of Bcl-2, N-cadherin, and Vimentin, and up-regulated the levels of Bax, C-caspase-3 and E-cadherin in ovarian cancer cells. RECK was lowly expressed in both ovarian cancer tissues and cells. siRECK not only had an effect opposite to DHA on the viability, apoptosis, migration, invasion, and related-factors of ovarian cancer cells but also offset the effect of DHA on ovarian cancer cells. CONCLUSION DHA regulated apoptosis, migration, and invasion of ovarian cancer cells via mediating RECK.
Collapse
Affiliation(s)
- Jingfei Zheng
- Department of Obstetrics and Gynecology, The Affiliated People's Hospital of Ningbo University, China.
| | - Xuehe Li
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, China
| | - Weili Yang
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, China
| | - Fang Zhang
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, China
| |
Collapse
|
13
|
Qingjie Fuzheng Granule suppresses lymphangiogenesis in colorectal cancer via the VEGF-C/VEGFR-3 dependent PI3K/AKT pathway. Biomed Pharmacother 2021; 137:111331. [PMID: 33578235 DOI: 10.1016/j.biopha.2021.111331] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 01/05/2023] Open
Abstract
SCOPE To investigate the effect of Qingjie Fuzheng Granule (QFG) on lymphangiogenesis and lymphatic metastasis in colorectal cancer. METHODS The effects of QFG on the expression and secretion of vascular endothelial growth factor-C (VEGF-C) in HCT-116 cells were investigated both in vitro and in vivo. HCT-116 cells were treated with different concentrations (0.2, 0.5, and 1.0 mg/mL) of QFG. The VEGF-C expression level was determined using RT-qPCR and western blotting, and the VEGF-C concentration in supernatant was measured by ELISA. Tumor xenograft models of HCT-116 cells were generated using BALB/c nude mice, and the mice were randomly divided into a control group (gavaged with normal saline) and QFG group (gavaged with 2 g/kg QFG). The effect of QFG on tumor growth was evaluated by comparing the volume and weight of tumors between two groups. Immunohistochemistry (IHC) and RT-qPCR were performed to detect the expression levels of VEGF-C, vascular endothelial growth factor receptor 3 (VEGFR-3), and LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1). ELISA was performed to measure the concentration of serum VEGF-C. TMT proteomics technology and Reactome pathway analysis were used to explore the mechanism of QFG inhibiting lymphangiogenesis in tumor. The VEGF-C (5 ng/mL)-stimulated human lymphatic endothelial cell (HLEC) model was conducted to evaluate the effect of QFG on lymphangiogenesis in vitro. The model cells were treated with different concentrations (0.2, 0.5, and 1.0 mg/mL) of QFG. Cell viability was then determined using an MTT assay. The cell migration, invasion, and tube-formation ability were analyzed using transwell migration, matrigel invasion and tube formation assays, respectively. The underlying mechanism was uncovered, the levels of VEGFR-3, matrix metalloproteinase 2 (MMP-2), matrix metalloproteinase 9 (MMP-9), p-PI3K/PI3K, p-AKT/AKT and p-mTOR/ mTOR were detected using western blotting. RESULTS QFG significantly reduced VEGF-C expression and secretion in HCT-116 cells. QFG evidently suppressed in vivo tumor growth and the expression of VEGF-C, VEGFR-3, and LYVE-1. The serum VEGF-C level was also reduced by QFG. Moreover, TMT proteomics technology and Reactome pathway analysis identified 95 differentially expressed protein and multiple enriched pathway about matrix metalloproteinase and extracellular matrix, which is direct associate with lymphangiogenesis. In vitro experiment, QFG inhibited the viability, migration, invasion and tube formation of HLECs. Additionally, QFG reduced the VEGFR-3, MMP-2, MMP-9 expression levels, and the p-PI3K/PI3K, p-AKT/AKT, p-mTOR/ mTOR ratios. CONCLUSION QFG can exert its effect on both tumor cells and HLECs, exhibiting ani- lymphangiogenesis in colorectal cancer via the VEGF-C/VEGFR-3 dependent PI3K/AKT pathway pathway.
Collapse
|
14
|
Zhu XQ, Yang H, Lin MH, Shang HX, Peng J, Chen WJ, Chen XZ, Lin JM. Qingjie Fuzheng Granules regulates cancer cell proliferation, apoptosis and tumor angiogenesis in colorectal cancer xenograft mice via Sonic Hedgehog pathway. J Gastrointest Oncol 2021; 11:1123-1134. [PMID: 33456987 DOI: 10.21037/jgo-20-213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Sonic Hedgehog (SHh) signaling pathway plays a critical role in cell proliferation, apoptosis, and tumor angiogenesis in various types of malignancies including colorectal cancer (CRC). Qingjie Fuzheng Granules (QFG) is a traditional Chinese medicinal formula, which has been clinically used in various cancer treatments, including CRC. In this study, we explored the potential molecular mechanisms of QFG treatment effects on CRC via the SHh pathway. Methods A CRC HCT-116 xenograft mouse model was utilized for all experiments. Mice were treated with intra-gastric administration of 1 g/kg of QFG or saline 6 days a week for 28 days (4 weeks). Body weight, length and shortest diameter of the tumor were measured every 3 days. At the end of the treatment, the tumor weight was measured. TUNEL staining assays were used to detect tumor apoptosis. Western blot and immunohistochemistry (IHC) assays were used to detect the expression of relative proteins. Results In our results, QFG inhibited the increase of tumor volume and weight, and exhibited no impact on mouse body weight. Furthermore, QFG significantly decreased the expression of SHh, Smo and Gli proteins, indicating the action of SHh signaling. Consequently, the expression of pro-proliferative survivin, Ki-67, Cyclin-D1 and CDK4 were decreased and expression of anti-proliferative p21 was increased. The pro-apoptotic Bax/Bcl-2 ratio, cle-caspase-3 and TUNEL-positive cell percentage in tumor tissues were increased. Meanwhile, the pro-angiogenic VEGF-A and VEGFR-2 expression was down-regulated. Conclusions QFG inhibited CRC cell proliferation and promoted CRC cell apoptosis and tumor angiogenesis in vivo through the suppression of SHh pathway, suggesting that QFG could be a potential therapeutic drug for CRC.
Collapse
Affiliation(s)
- Xiao-Qin Zhu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Hong Yang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Ming-He Lin
- Editorial Department of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hai-Xia Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Wu-Jin Chen
- Department of Oncology, Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xu-Zheng Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Jiu-Mao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| |
Collapse
|
15
|
Zhao P, Cheng J, Li B, Nie D, Wang H, Li C, Gui S, Zhang Y. LncRNA PCAT6 regulates the progression of pituitary adenomas by regulating the miR-139-3p/BRD4 axis. Cancer Cell Int 2021; 21:14. [PMID: 33407504 PMCID: PMC7789787 DOI: 10.1186/s12935-020-01698-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/02/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Dysregulated lncRNA PCAT6 was discovered in many cancers excluding pituitary adenomas (PA). Therefore, we explored the role of PCAT6 in PA in this research. METHODS Abnormally expressed miRNAs were analyzed by bioinformatics and RT-qPCR. The target and regulator of miR-139-3p were determined by bioinformatics, dual-luciferase reporter assay, or RIP. The correlation among PCAT6, miR-139-3p, and BRD4 was further analyzed. The viability, apoptosis, cell cycle distribution of PA cells, as well as their ability to invade, migrate, and proliferate, were tested after transfection through CCK-8, flow cytometry, transwell, wound healing, and colony formation assays. After construction of transplanted-tumor model in nude mice, cell apoptosis in the tumor was detected by TUNEL. The expressions of PCAT6, BRD4, miR-139-3p, and apoptosis-related factors in PA tissues, cells, or tumor tissues were detected by RT-qPCR, Western blot, or IHC. RESULTS PCAT6 and BRD4 were high-expressed but miR-139-3p was low-expressed in PA. Both the 3'-untranslated regions of PCAT6 and BRD4 mRNAs were demonstrated to contain a potential binding site for miR-139-3p. PCAT6 was positively correlated to BRD4, and miR-139-3p was negatively correlated to PCAT6 and BRD4. MiR-139-3p mimic, shPCAT6 and siBRD4 inhibited the viability, migration, invasion, and proliferation of PA cells while inducing apoptosis. MiR-139-3p mimic and shPCAT6 inhibited the cell cycle progression of PA cells, decreased the weight and volume of the xenotransplanted tumor, and reduced the levels of Bcl-2 and BRD4 while enhancing the levels of Bax, miR-139-3p, and Cleaved caspase-3. MiR-139-3p inhibitor caused the opposite effect of miR-139-3p mimic and further reversed the effect of shPCAT6 on on PA cells. CONCLUSION PCAT6 regulated the progression of PA via modulating the miR-139-3p/BRD4 axis, which might provide a novel biomarker for the prevention, diagnosis, and treatment of PA.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China.
| | - Jianhua Cheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Bin Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Ding Nie
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Hongyun Wang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Yazhuo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| |
Collapse
|
16
|
Fan Y, Ma Z, Zhao L, Wang W, Gao M, Jia X, Ouyang H, He J. Anti-tumor activities and mechanisms of Traditional Chinese medicines formulas: A review. Biomed Pharmacother 2020; 132:110820. [DOI: 10.1016/j.biopha.2020.110820] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/19/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023] Open
|
17
|
Zhang L, Tong Z, Sun Z, Zhu G, Shen E, Huang Y. MiR-25-3p targets PTEN to regulate the migration, invasion, and apoptosis of esophageal cancer cells via the PI3K/AKT pathway. Biosci Rep 2020; 40:BSR20201901. [PMID: 32985648 PMCID: PMC7560540 DOI: 10.1042/bsr20201901] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/13/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Esophageal cancer (EC) is one of the most common malignant tumors of the digestive system. MiR-25-3p was proved to be a biomarker for the diagnosis and treatment of many cancers. MiR-25-3p was found to be high expressed in the blood of EC patients. The aim of the present study was to explore the effect of miR-25-3p and its target gene on EC. METHODS miR-25-3p expression in the blood of EC patients and EC cells was detected by RT-qPCR. The target of miR-25-3p was identified by bioinformatics and luciferase reporter assay. After transfection, cell viability, apoptosis, migration, and invasion were detected by MTT, flow cytometry, wound healing, and transwell assays, respectively. The expressions of PTEN, Bax, Bcl-2, cleaved Caspase-3, p-PI3K, PI3K, p-AKT, and AKT were detected by Western blot. RESULTS MiR-25-3p was high expressed in the blood of EC patients and EC cells. MiR-25-3p targeted PTEN and inhibited the expression of PTEN. MiR-25-3p mimic increased the viability, migration, invasion and the expressions of Bcl-2, and inhibited the apoptosis and the expression of Bax and cleaved caspase-3 in EC cells. MiR-25-3p mimic also enhanced the expressions of p-PI3K and p-AKT and the ratios of p-PI3K/PI3K and p-AKT/AKT in EC cells. PTEN overexpression not only had an opposite effect of miR-25-3p mimic, but also reversed the effect of miR-25-3p mimic on EC cells. CONCLUSION MiR-25-3p targeted PTEN to promote the migration and invasion, and inhibit apoptosis of EC cells via the PI3K/AKT pathway, which might provide a new therapeutic target for EC treatment.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province 110042, China
| | - Zhuang Tong
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province 110042, China
| | - Zhe Sun
- Department of Surgical Oncology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, China
| | - Guolian Zhu
- Department of Oncology, Shenyang Fifth People Hospital, Shenyang, Liaoning Province 110001, China
| | - Erdong Shen
- Department of Oncology, Yueyang First People’s Hospital, Yueyang, Hunan 414000, China
| | - Yanfeng Huang
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning Province 110042, China
| |
Collapse
|
18
|
Zhang L, Liu J, Lin S, Tan J, Huang B, Lin J. Qingjie Fuzheng Granule Inhibited the Migration and Invasion of Colorectal Cancer Cells by Regulating the lncRNA ANRIL/let-7a/TGF- β1/Smad Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:5264651. [PMID: 32714407 PMCID: PMC7341385 DOI: 10.1155/2020/5264651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/02/2020] [Indexed: 12/24/2022]
Abstract
Qingjie Fuzheng granule (QFG) promotes cancer cell apoptosis and ameliorates intestinal mucosal damage caused by 5-fluorouracil. However, the antitumor role of QFG in colorectal cancer (CRC) progression remains unclear. In this study, the growth of HCT-8 and HCT116 cells incubated with various concentrations of QFG for 24 and 48 h was evaluated using MTT assays; their abilities of migration and invasion were investigated through wound healing and Transwell assays. The expression of lncRNA ANRIL, let-7a, and the TGF-β1/Smad signaling pathway components was assessed using real-time PCR and western blotting. The results elicited that QFG significantly suppressed the growth of HCT-8 and HCT116 cells; the half-maximal inhibitory concentrations (IC50) of QFG for HCT-8 and HCT116 cells for 48 h were 1.849 and 1.608 mg/mL, respectively. The abilities of wound healing, migration, and invasion of HCT-8 and HCT116 cells were dose-dependently decreased by QFG treatment for 24 h, respectively. QFG decreased the expression of lncRNA ANRIL, TGF-β1, phosphorylated (p)-Smad2/3, Smad4, and N-cadherin and upregulated the expression of let-7a in HCT-8 and HCT116 cells. Collectively, our data demonstrated that QFG inhibited the metastasis of CRC cells by regulating the lncRNA ANRIL/let-7a/TGF-β1/Smad axis, indicating that they might serve as an adjunctive medicine for CRC treatment.
Collapse
Affiliation(s)
- Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jianxin Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jingzhuang Tan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Bin Huang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| |
Collapse
|
19
|
Zhang H, Hui D, Li Y, Xiong G, Fu X. Canmei Formula Reduces Colitis-Associated Colorectal Carcinogenesis in Mice by Modulating the Composition of Gut Microbiota. Front Oncol 2019; 9:1149. [PMID: 31803606 PMCID: PMC6877713 DOI: 10.3389/fonc.2019.01149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota, including pathogenic microorganisms and probiotics, has been involved in tumor initiation and progression by regulating the components of intestinal flora. Canmei formula (CMF), a traditional Chinese medicine, chronicled in the Chuang Yang Jing Yan Quan Shu, has been clinically used as an adjuvant therapy to treat patients with colorectal carcinoma (CRC) in China. In this study, we investigate the treatment effect of CMF in the azoxymethane (AOM) and dextran sodium sulfate (DSS) induced and high-fat diet augmented colitis-associated colorectal cancer in vivo, and explore its mechanism of action. We found that CMF treatment relieved the inflammation and alteration of the gut microbiota and significantly inhibited the development of intestinal adenoma. Linear discriminant analysis showed that the flora diversity in the normal mice, model mice and CMF treatment mice was different. At the family level, the relative abundance of Desulfovibrionaceae decreased in CMF groups. The relative abundance of Desulfovibrionaceae were lower in the CMF groups than in model group, whereas Rikenellaceae and Alistipes were increased. Altogether our results indicate that CMF treatment ameliorate colitis-associated colorectal carcinogenesis by modulating the composition of the gut microbiota in vivo.
Collapse
Affiliation(s)
- Huayue Zhang
- Department of Medical Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dengcheng Hui
- Department of Cirrhosis, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Li
- Department of Medical Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangsu Xiong
- Endoscopic Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoling Fu
- Department of Medical Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Yang PP, Yu XH, Zhou J. Tryptophanyl-tRNA synthetase (WARS) expression in uveal melanoma - possible contributor during uveal melanoma progression. Biosci Biotechnol Biochem 2019; 84:471-480. [PMID: 31694485 DOI: 10.1080/09168451.2019.1686967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study aimed to explore the influence of Tryptophanyl-tRNA synthetase (WARS) expression on the proliferation and migration of uveal melanoma (UM) cells, and the potential mechanisms. Bioinformatics analysis based on Gene Expression Omnibus (GEO) database showed that WARS expression in metastatic cancer was significantly higher than that in no-metastatic group. Kaplan-Meier analysis based on The Cancer Genome Atlas (TCGA) database showed that high WARS expression was associated with lower survival. Biological function experiments showed that overexpression of WARS in OCM-1A cells can promote cell proliferation, migration, and invasion, whereas knockdown of WARS in C918 cells showed the opposite effect. Finally, we observed that the up-regulation of WARS induced the activation of phosphatidylinositol 3-kinase/AKT (PI3K/AKT) signaling, whilst depletion of WARS resulted in opponent outcomes. Taken together, our results illustrated that WARS was overexpressed in UM cells and contributed to the viability and motility of UM cells via modulating PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Pan-Pan Yang
- Department of Oncology, Jining First People's Hospital, Jining, P.R. China
| | - Xiao-Hui Yu
- Department of Ophthalmology, Yantai Yuhuangding Hospital, Yantai, P.R. China
| | - Jiao Zhou
- Department of blood transfusion, Suizhou central hospital, Hubei university of medicine, Hubei, China
| |
Collapse
|