1
|
WANG XIAOXI, JIA YANFEI, LI QIANG, YANG QIANG, LIU YINGFENG, WEI BEIFENG, NIU XIANG, ZHANG YINJIE, LUO XIAODONG, ZHAO ZIYU, WANG PENG. miR-200b-3p accelerates progression of pituitary adenomas by negatively regulating expression of RECK. Oncol Res 2024; 32:933-941. [PMID: 38686051 PMCID: PMC11055999 DOI: 10.32604/or.2023.042581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/12/2023] [Indexed: 05/02/2024] Open
Abstract
MicroRNA (miR)-200b-3p has been associated with many tumors, but its involvement in pituitary adenoma is unclear. This study investigated the molecular mechanism underlying miR-200b-3p regulation in pituitary adenomas to provide a theoretical basis for treatment. Bioinformatics was used to analyze pituitary adenoma-related genes and screen new targets related to RECK and miRNA. As well, the relationship between miR-200b-3p and RECK protein was verified using a double-luciferase reporter gene assay. The expression of miR-200b-3p in clinical samples was analyzed by in situ hybridization. Transfection of the miR-200b-3p inhibitor and small interfering-RECK (si-RECK) was verified by qPCR. GH3 cell viability and proliferation were detected using CCK8 and EdU assays. Apoptosis was detected by flow cytometry and western blotting. Wound healing and Transwell assays were used to detect cell migration and invasion. The effects of miR-200b-3p and RECK on GH3 cells were verified using salvage experiments. miR-200b-3p was highly expressed in pituitary tumor tissue. Inhibitors of miR-200b-3p inhibited cell proliferation promoted cell apoptosis, inhibited invasion and migration, and inhibited the expression of matrix metalloproteinases. Interestingly, miR-200b-3p negatively regulated RECK. The expression of RECK in pituitary adenoma tissues was lower than that in neighboring tissues. Si-RECK rescued the function of miR-200b-3p inhibitors in the above cellular behaviors, and miR-200b-3p accelerated the development of pituitary adenoma by negatively regulating RECK expression. In summary, this study investigated the molecular mechanism by which miR-200b-3p regulates the progression of pituitary adenoma through the negative regulation of RECK. The findings provide a new target for the treatment of pituitary adenoma.
Collapse
Affiliation(s)
- XIAOXI WANG
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - YANFEI JIA
- Department of Neurosurgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - QIANG LI
- Department of Neurosurgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - QIANG YANG
- Department of Neurosurgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - YINGFENG LIU
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - BEIFENG WEI
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - XIANG NIU
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - YINJIE ZHANG
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - XIAODONG LUO
- Department of Neurosurgery, The Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - ZIYU ZHAO
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| | - PENG WANG
- Department of Neurosurgery, Tianshui First People’s Hospital, Tianshui, China
| |
Collapse
|
2
|
Aanniz T, Bouyahya A, Balahbib A, El Kadri K, Khalid A, Makeen HA, Alhazmi HA, El Omari N, Zaid Y, Wong RSY, Yeo CI, Goh BH, Bakrim S. Natural bioactive compounds targeting DNA methyltransferase enzymes in cancer: Mechanisms insights and efficiencies. Chem Biol Interact 2024; 392:110907. [PMID: 38395253 DOI: 10.1016/j.cbi.2024.110907] [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: 11/16/2023] [Revised: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024]
Abstract
The regulation of gene expression is fundamental to health and life and is essentially carried out at the promoter region of the DNA of each gene. Depending on the molecular context, this region may be accessible or non-accessible (possibility of integration of RNA polymerase or not at this region). Among enzymes that control this process, DNA methyltransferase enzymes (DNMTs), are responsible for DNA demethylation at the CpG islands, particularly at the promoter regions, to regulate transcription. The aberrant activity of these enzymes, i.e. their abnormal expression or activity, can result in the repression or overactivation of gene expression. Consequently, this can generate cellular dysregulation leading to instability and tumor development. Several reports highlighted the involvement of DNMTs in human cancers. The inhibition or activation of DNMTs is a promising therapeutic approach in many human cancers. In the present work, we provide a comprehensive and critical summary of natural bioactive molecules as primary inhibitors of DNMTs in human cancers. The active compounds hold the potential to be developed as anti-cancer epidrugs targeting DNMTs.
Collapse
Affiliation(s)
- Tarik Aanniz
- Medical Biotechnology Laboratory, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Rabat, B.P, 6203, Morocco.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, 10106, Morocco.
| | - Abdelaali Balahbib
- High Institute of Nursing Professions and Health Techniques of Errachidia, Errachidia, Morocco.
| | - Kawtar El Kadri
- High Institute of Nursing Professions and Health Techniques of Errachidia, Errachidia, Morocco
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia; Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum, 11111, Sudan.
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia.
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia; Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia.
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco.
| | - Younes Zaid
- Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Rebecca Shin-Yee Wong
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia; Department of Medical Education, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| | - Chien Ing Yeo
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia; Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, 47500, Malaysia; College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| | - Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco.
| |
Collapse
|
3
|
Carbone K, Gervasi F, Kozhamzharova L, Altybaeva N, Sönmez Gürer E, Sharifi-Rad J, Hano C, Calina D. Casticin as potential anticancer agent: recent advancements in multi-mechanistic approaches. Front Mol Biosci 2023; 10:1157558. [PMID: 37304067 PMCID: PMC10250667 DOI: 10.3389/fmolb.2023.1157558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Plants, with their range of pharmacologically active molecules, represent the most promising source for the production of new anticancer drugs and for the formulation of adjuvants in chemotherapy treatments to reduce drug content and/or counteract the side effects of chemotherapy. Casticin is a major bioactive flavonoid isolated from several plants, mainly from the Vitex species. This compound is well known for its anti-inflammatory and antioxidant properties, which are mainly exploited in traditional medicine. Recently, the antineoplastic potential of casticin has attracted the attention of the scientific community for its ability to target multiple cancer pathways. The purpose of this review is, therefore, to present and critically analyze the antineoplastic potential of casticin, highlighting the molecular pathways underlying its antitumor effects. Bibliometric data were extracted from the Scopus database using the search strings "casticin" and "cancer" and analyzed using VOSviewer software to generate network maps to visualize the results. Overall, more than 50% of the articles were published since 2018 and even more recent studies have expanded the knowledge of casticin's antitumor activity by adding interesting new mechanisms of action as a topoisomerase IIα inhibitor, DNA methylase 1 inhibitor, and an upregulator of the onco-suppressive miR-338-3p. Casticin counteracts cancer progression through the induction of apoptosis, cell cycle arrest, and metastasis arrest, acting on several pathways that are generally dysregulated in different types of cancer. In addition, they highlight that casticin can be considered as a promising epigenetic drug candidate to target not only cancer cells but also cancer stem-like cells.
Collapse
Affiliation(s)
- Katya Carbone
- CREA—Research Centre for Olive, Fruit and Citrus Crops, Rome, Italy
| | - Fabio Gervasi
- CREA—Research Centre for Olive, Fruit and Citrus Crops, Rome, Italy
| | - Latipa Kozhamzharova
- Department of Scientific Works and International Relations, International Taraz Innovative Institute Named After Sherkhan Murtaza, Taraz, Kazakhstan
| | - Nazgul Altybaeva
- Department of Molecular Biology and Genetics, Al-Farabi Kazakh National University, Al-frabi, Kazakhstan
| | - Eda Sönmez Gürer
- Department of Pharmacognosy, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | | | - Christophe Hano
- Department of Biological Chemistry, Université ď Orléans, Orléans, France
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
4
|
Wang XL, Cao XZ, Wang DY, Qiu YB, Deng KY, Cao JG, Lin SQ, Xu Y, Ren KQ. Casticin Attenuates Stemness in Cervical Cancer Stem-Like Cells by Regulating Activity and Expression of DNMT1. Chin J Integr Med 2023; 29:224-232. [PMID: 35809177 DOI: 10.1007/s11655-022-3469-z] [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] [Accepted: 12/21/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To explore whether casticin (CAS) suppresses stemness in cancer stem-like cells (CSLCs) obtained from human cervical cancer (CCSLCs) and the underlying mechanism. METHODS Spheres from HeLa and CaSki cells were used as CCSLCs. DNA methyltransferase 1 (DNMT1) activity and mRNA levels, self-renewal capability (Nanog and Sox2), and cancer stem cell markers (CD133 and CD44), were detected by a colorimetric DNMT activity/inhibition assay kit, quantitative real-time reverse transcription-polymerase chain reaction, sphere and colony formation assays, and immunoblot, respectively. Knockdown and overexpression of DNMT1 by transfection with shRNA and cDNA, respectively, were performed to explore the mechanism for action of CAS (0, 10, 30, and 100 nmol/L). RESULTS DNMT1 activity was increased in CCSLCs compared with HeLa and CaSki cells (P<0.05). In addition, HeLa-derived CCSLCs transfected with DNMT1 shRNA showed reduced sphere and colony formation abilities, and lower CD133, CD44, Nanog and Sox2 protein expressions (P<0.05). Conversely, overexpression of DNMT1 in HeLa cells exhibited the oppositive effects. Furthermore, CAS significantly reduced DNMT1 activity and transcription levels as well as stemness in HeLa-derived CCSLCs (P<0.05). Interestingly, DNMT1 knockdown enhanced the inhibitory effect of CAS on stemness. As expected, DNMT1 overexpression reversed the inhibitory effect of CAS on stemness in HeLa cells. CONCLUSION CAS effectively inhibits stemness in CCSLCs through suppression of DNMT1 activation, suggesting that CAS acts as a promising preventive and therapeutic candidate in cervical cancer.
Collapse
Affiliation(s)
- Xue-Li Wang
- Medical College, Hunan University of Medicine, Huaihua, Hunan Province, 418000, China
| | - Xiao-Zheng Cao
- Clinical Department of Guangdong Metabolic Disease Research Centre of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510062, China
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dao-Yuan Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Ye-Bei Qiu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Kai-Yu Deng
- Medical College, Hunan University of Medicine, Huaihua, Hunan Province, 418000, China
| | - Jian-Guo Cao
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
- The Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China
| | - Shao-Qiang Lin
- Clinical Department of Guangdong Metabolic Disease Research Centre of Integrated Chinese and Western Medicine, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510062, China
| | - Yong Xu
- Institute of Chemical Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Kai-Qun Ren
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Medical College, Hunan Normal University, Changsha, 410013, China.
| |
Collapse
|
5
|
The Role of Epigenetic Modifications in Human Cancers and the Use of Natural Compounds as Epidrugs: Mechanistic Pathways and Pharmacodynamic Actions. Biomolecules 2022; 12:biom12030367. [PMID: 35327559 PMCID: PMC8945214 DOI: 10.3390/biom12030367] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer is a complex disease resulting from the genetic and epigenetic disruption of normal cells. The mechanistic understanding of the pathways involved in tumor transformation has implicated a priori predominance of epigenetic perturbations and a posteriori genetic instability. In this work, we aimed to explain the mechanistic involvement of epigenetic pathways in the cancer process, as well as the abilities of natural bioactive compounds isolated from medicinal plants (flavonoids, phenolic acids, stilbenes, and ketones) to specifically target the epigenome of tumor cells. The molecular events leading to transformation, angiogenesis, and dissemination are often complex, stochastic, and take turns. On the other hand, the decisive advances in genomics, epigenomics, transcriptomics, and proteomics have allowed, in recent years, for the mechanistic decryption of the molecular pathways of the cancerization process. This could explain the possibility of specifically targeting this or that mechanism leading to cancerization. With the plasticity and flexibility of epigenetic modifications, some studies have started the pharmacological screening of natural substances against different epigenetic pathways (DNA methylation, histone acetylation, histone methylation, and chromatin remodeling) to restore the cellular memory lost during tumor transformation. These substances can inhibit DNMTs, modify chromatin remodeling, and adjust histone modifications in favor of pre-established cell identity by the differentiation program. Epidrugs are molecules that target the epigenome program and can therefore restore cell memory in cancerous diseases. Natural products isolated from medicinal plants such as flavonoids and phenolic acids have shown their ability to exhibit several actions on epigenetic modifiers, such as the inhibition of DNMT, HMT, and HAT. The mechanisms of these substances are specific and pleiotropic and can sometimes be stochastic, and their use as anticancer epidrugs is currently a remarkable avenue in the fight against human cancers.
Collapse
|
6
|
Shang HS, Chen KW, Chou JS, Peng SF, Chen YL, Chen PY, Huang HC, Lu HF, Chang HY, Shih YL, Huang WW. Casticin Inhibits In Vivo Growth of Xenograft Tumors of Human Oral Cancer SCC-4 Cells. In Vivo 2021; 34:2461-2467. [PMID: 32871773 DOI: 10.21873/invivo.12061] [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: 03/16/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND/AIM Casticin, one of the active components of Vitex rotundifolia L., presents biological and pharmacological activities including inhibition of migration, invasion and induction of apoptosis in numerous human cancer cells in vitro. This study aimed to assess the effects of casticin on tumor growth in a human oral cancer SCC-4 cell xenograft mouse model in vivo. MATERIALS AND METHODS Twenty-four nude mice were injected subcutaneously with SCC-4 cells and when palpable tumors reached a volume of 100-120 mm3 the mice were randomly divided into three groups. The control (0.1% dimethyl sulfoxide), casticin (0.2 mg/kg), and casticin (0.4 mg/kg) groups were intraperitoneally injected every two days for 18 days. Tumor volume and body weights were measured every two days. RESULTS Casticin significantly decreased tumor volume and weight in SCC-4 cell xenograft mice but there was no statistically significant difference between the body weights of control mice and mice treated with 0.2 mg/kg or 0.4 mg/kg casticin. Therefore, the growth of SCC-4 cells in athymic nude mice can be inhibited by casticin in vivo. CONCLUSION These findings support further investigations in the potential use of casticin as an oral anti-cancer drug in the future.
Collapse
Affiliation(s)
- Hung-Sheng Shang
- Graduate Institute of Clinical of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C.,Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Kuo-Wei Chen
- Division of Hematology and Oncology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Jiann-Shang Chou
- Department of Anatomic Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.,Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Yung-Liang Chen
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan, R.O.C
| | - Po-Yuan Chen
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Hsieh-Chou Huang
- Anesthesiology and Pain Medicine, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C.,Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C.,Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei, Taiwan, R.O.C
| | - Hsin-Yu Chang
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Yung-Luen Shih
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan, R.O.C. .,Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C.,School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
| |
Collapse
|
7
|
Li X, Wang L, Cao X, Zhou L, Xu C, Cui Y, Qiu Y, Cao J. Casticin inhibits stemness of hepatocellular carcinoma cells via disrupting the reciprocal negative regulation between DNMT1 and miR-148a-3p. Toxicol Appl Pharmacol 2020; 396:114998. [PMID: 32268151 DOI: 10.1016/j.taap.2020.114998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 03/21/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023]
Abstract
Casticin (CAS) is a polymethyl flavonoid from Fructus viticis and has multiple pharmacological activities, including anticancer. However, whether the molecular mechanism underlying CAS represses stemness characteristics in hepatocellular carcinoma (HCC) cells involves intervention in the reciprocal negative regulation between DNA methyltransferase 1 (DNMT1) and miR-148a-3p has not yet been reported. In this study, the effect of CAS on stemness characteristics of HCC cells and its mechanism were investigated. Results showed that CAS selectively reduced the viabilities of HCC cells but not L02 cells, as determined by CCK-8 assay. Importantly, the sub-cytotoxic concentrations of CAS could inhibit the stemness characteristics in HCC cells, as demonstrated by the expression of stemness biomarkers (CD44, EpCAM, Bmi1, Nanog, and Oct4), sphere forming assay, RT-qPCR, and Western blotting. In addition, CAS repressed DNMT1 activity and expression and increased miR-148a-3p. The effect of CAS on stemness characteristics was abolished by stable DNMT1 overexpression. MiR-148a-3p overexpression enhanced the reduction of CAS on stemness characteristics. DNMT1 overexpression promoted miR-148a-3p promoter hypermethylation as detected by methylation-specific PCR (MSP), which repressed its expression. Conversely, miR-148a-3p repressed DNMT1 expression by specific site binding to 3'-UTR of DNMT1 mRNA, as determined by luciferase assay. Moreover, the combination of CAS and agomir-148a-3p had robust effects on tumor suppression as compared to the sole activity of either molecule in nude mouse xenograft experiments in vivo. The findings suggested that CAS could inhibit stemness characteristics in HCC cells by interruption of the reciprocal negative regulation between DNMT1 and miR-148a-3p.
Collapse
Affiliation(s)
- Xiang Li
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha 410013, China.
| | - Lianghou Wang
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha 410013, China
| | - Xiaocheng Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, China; Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha 410013, China
| | - Lingli Zhou
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha 410013, China
| | - Chang Xu
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, China; Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha 410013, China
| | - Yinghong Cui
- Department of Preclinical Medicine, Medical College, Hunan Normal University, Changsha 410013, China
| | - Yebei Qiu
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, China; Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha 410013, China
| | - Jianguo Cao
- Department of Pharmaceutical Science, Medical College, Hunan Normal University, Changsha 410013, China; Key Laboratory of Study and Discover of Small Targeted Molecules of Hunan Province, Medical College, Hunan Normal University, Changsha 410013, China.
| |
Collapse
|
8
|
Yuan J, Li W, Zhu J, Deng S, Tao X. Low expression of RECK in oral squamous cell carcinoma patients induces a shorter survival rate through an imbalance of RECK/MMPs. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:501-508. [PMID: 32269688 PMCID: PMC7136997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/07/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND RECK, as a negative MMP regulator, is extensively expressed in normal cells but decreased in tumors. In OSCC, the relationship between RECK and MMPs and the potential prognostic impact remains unclear. In this research, for the first time, we investigated the expression of RECK associated with MMPs during OSCC carcinogenesis in a large sample and its association with 5-year survival rate. MATERIAL AND METHODS Immunohistochemical SP technique was applied to study the expression of RECK and MMP-2 and MMP-9 in 108 cases of OSCC and 30 normal oral mucosae. Univariate and multivariate Cox regression analysis was utilized for disease-free survival and overall survival, and analyzed by Kaplan-Meier method regarding RECK expression in patients of OSCC. RESULTS We found lower expression of RECK in OSCC was 51.85% (56/108) compared with 93.33% (28/30) in the control group. However, the higher expression of MMP-2 and MMP-9 was 74.07% (80/108) and 70.37% (76/108) in OSCC, respectively, compared with 20% (6/30) and 13.3% (4/30) in the control group. Furthermore, the decrease of RECK expression and the increase of MMP-2, and -9 expression were significantly correlated with the loss of histologic differentiation, the occurrence of lymphatic metastasis, and the increase of OSCC clinical stage (P<0.05). OSCC patients with a low level of RECK expression had a lower rate of 5-year survival. CONCLUSION RECK may prevent metastasis and improve OSCC patients' prognosis through a RECK/MMP-2, and -9 imbalance. Furthermore, RECK is a prospective prognostic indicator and therapeutic target for cancer molecular targeting therapy. Low expression of RECK may be a significant negative prognostic predictor.
Collapse
Affiliation(s)
- Jian Yuan
- Department of Conservative Dentistry and Endodontics, The Affiliated Stomatology Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of StomatologyHangzhou 310006, Zhejiang, China
| | - Wen Li
- Department of Conservative Dentistry and Endodontics, The Affiliated Stomatology Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of StomatologyHangzhou 310006, Zhejiang, China
| | - Jinxiao Zhu
- Department of Stomatology, Wuxi Children’s HospitalWuxi, Jiangsu, China
| | - Shuli Deng
- Department of Conservative Dentistry and Endodontics, The Affiliated Stomatology Hospital, Zhejiang University School of MedicineHangzhou, Zhejiang, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of StomatologyHangzhou 310006, Zhejiang, China
| | - Xuejin Tao
- Center of Stomatology, Tongji Hospital Affiliated to Tongji Medical College Hua Zhong University of Science and TechnologyWuhan, Hubei, China
| |
Collapse
|
9
|
Li LT, Wang X, Zhu WT, Qian GW, Pei DS, Zheng JN. Reciprocal Role Of DNA Methylation And Sp1 Binding In Ki-67 Gene Transcription. Cancer Manag Res 2019; 11:9749-9759. [PMID: 31819613 PMCID: PMC6874502 DOI: 10.2147/cmar.s213769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 10/16/2019] [Indexed: 01/26/2023] Open
Abstract
Purpose DNA methylation plays major regulatory roles in gene transcription. Our previous studies confirmed that Ki-67 promoter is hypomethylated and Sp1 is a transcriptional activator of Ki-67 gene in cancer cells. However, whether Sp1-mediated transcriptional activation of Ki-67 is related to its methylation has not been studied yet. Materials and methods In this study, we confirmed that methylated CpG binding protein 2 (MBD2) binding to methylated DNA hindered the binding of Sp1 to Ki-67 promoter and then repressed Ki-67 transcription through chromatin immunoprecipitation (ChIP) and quantitative real-time PCR (qRT-PCR). Co-immunoprecipitation (Co-IP), ChIP, methylation-specific PCR (MS-PCR) and Western blot were utilized to analyze the effects of Sp1 binding to Ki-67 promoter on its methylation status. Results Less DNA methyltransferase 1 (DNMT1) bound to the Ki-67 promoter in MKN45 cells than in HK-2 cells. Histone acetyltransferase p300 that was recruited by Sp1 to Ki-67 promoter could attenuate the methylation level of Ki-67 promoter. Furthermore, higher expression of Sp1 and Ki-67 was related to the overall survival (OS), first progression (FP) and post-progression survival (PPS) in gastric cancer by scrutinizing bioinformatics datasets. Conclusion Taken together, our findings suggested that hypomethylation of Ki-67 promoter enhanced the binding of Sp1, which in turn maintained hypomethylation of promoter, leading to increase Ki-67 expression in cancer cells. Sp1 and Ki-67 could act promising prognostic biomarkers for clinical diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Lian-Tao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Xun Wang
- Department of Interventional Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Wen-Tao Zhu
- Department of Pathology, Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Guo-Wei Qian
- Department of Medical Oncology, Shanghai Sixth People's Hospital, Shanghai 200000, People's Republic of China
| | - Dong-Sheng Pei
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Department of Pathology, Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| | - Jun-Nian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou 221000, People's Republic of China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, People's Republic of China
| |
Collapse
|
10
|
The DNMT1/miR-34a Axis Is Involved in the Stemness of Human Osteosarcoma Cells and Derived Stem-Like Cells. Stem Cells Int 2019; 2019:7028901. [PMID: 31781245 PMCID: PMC6875320 DOI: 10.1155/2019/7028901] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/25/2019] [Indexed: 12/23/2022] Open
Abstract
The DNA methyltransferase 1 (DNMT1)/miR-34a axis promoted carcinogenesis of various types of cancers. However, no literature reported its contribution to the stemness of osteosarcoma cancer stem-like cells (OSLCs). We sought to determine whether the DNMT1/miR-34a axis facilitates the stemness of OSLCs. We here revealed the higher DNMT1 activity and expression, lower miR-34a expression with high methylation of its promoter, and stronger stemness of OSLCs, as manifested by elevated sphere and colony formation capacities; CD133, CD44, ABCG2, Bmi1, Sox2, and Oct4 protein amounts in vitro; and carcinogenicity in a nude mouse xenograft model, when compared to the parental U2OS cells. 5-Azacytidine (Aza-dC) repressed DNMT1 activation and upregulated miR-34a expression by promoter demethylation and suppressed the stemness of OSLCs in a dose-dependent manner. DNMT1 knockdown increased miR-34a and reduced the stemness of OSLCs. Transfection with a miR-34a mimic repressed the stemness of OSLCs but did not alter DNMT1 activity and expression. Conversely, DNMT1 overexpression declined miR-34a levels, promoting the stemness of U2OS cells. Transfection with a miR-34a inhibitor enhanced the stemness of U2OS cells, without affecting the DNMT1 activity and expression. Importantly, reexpression of miR-34a could rescue the effects of DNMT1 overexpression on miR-34a inhibition as well as the stemness promotion without affecting the activity and expression of DNMT1. Our results revealed that aberrant activation of DNMT1 caused promoter methylation of miR-34a, leading to miR-34a underexpression, and the role of the DNMT1/miR-34a axis in promoting and sustaining the stemness of OSLCs.
Collapse
|
11
|
Casticin inhibits PDGF-induced proliferation and migration of airway smooth muscle cells. Eur J Pharmacol 2018; 830:39-46. [PMID: 29665364 DOI: 10.1016/j.ejphar.2018.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 01/04/2023]
Abstract
Casticin (3', 5-dihydroxy-3, 4', 6, 7-tetramethoxyflavone), one of the main components from Vitex rotundifolia L., was reported to possess several pharmacological properties, including anti-inflammatory, hepatoprotective, anticancer, anti-asthma activities. However, the effects of casticin on airway smooth muscle cells (ASMCs) proliferation and migration have not been explored. This study aimed to evaluate the effects of casticin on the proliferation and migration of ASMCs, and study the possible molecular mechanism. Our results demonstrated that casticin significantly suppressed the proliferation and migration of ASMCs exposed to platelet-derived growth factor (PDGF), as well as reversed the PDGF-induced inhibition of the expression of contractile phenotype markers in ASMCs. In addition, casticin also inhibited PDGF-induced the expression of type I collagen and fibronectin in ASMCs induced by PDGF. Furthermore, casticin significantly prevented the activation of ERK1/2 and NF-κB pathways in PDGF-stimulated ASMCs. Taken together, these data demonstrated that casticin inhibits PDGF-induced human ASMC proliferation and migration through suppressing the activation of ERK1/2 and NF-κB signaling pathways.
Collapse
|
12
|
Chan EWC, Wong SK, Chan HT. Casticin from Vitex species: a short review on its anticancer and anti-inflammatory properties. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2018; 16:147-152. [PMID: 29559215 DOI: 10.1016/j.joim.2018.03.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/18/2018] [Indexed: 01/04/2023]
Abstract
This short review provides an update of the anticancer and anti-inflammatory properties of casticin from Vitex species. Casticin is a polymethylflavone with three rings, an orthocatechol moiety, a double bond, two hydroxyl groups and four methoxyl groups. Casticin has been isolated from various tissues of plants in the Vitex genus: fruits and leaves of V. trifolia, aerial parts and seeds of V. agnus-castus and leaves of V. negundo. Studies have reported the antiproliferative and apoptotic activities of casticin from Vitex species. The compound is effective against many cancer cell lines via different molecular mechanisms. Studies have also affirmed the anti-inflammatory properties of casticin, with several molecular mechanisms identified. Other pharmacological properties include anti-asthmatic, tracheospasmolytic, analgesic, antihyperprolactinemia, immunomodulatory, opioidergic, oestrogenic, anti-angiogenic, antiglioma, lung injury protection, rheumatoid arthritis amelioration and liver fibrosis attenuation activities. Clinical trials and commercial use of the casticin-rich fruit extract of V. agnus-castus among women with premenstrual syndrome were briefly discussed.
Collapse
Affiliation(s)
- Eric Wei Chiang Chan
- Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia.
| | - Siu Kuin Wong
- School of Science, Monash University, Petaling Jaya, Selangor 46150, Malaysia
| | - Hung Tuck Chan
- Secretariat of International Society for Mangrove Ecosystems, Faculty of Agriculture, University of the Ryukyus, Okinawa 903-0129, Japan
| |
Collapse
|