1
|
Kharazi DM, Karam L, El Boustany C, Ibrahim JN. Sodium butyrate and sodium propionate inhibit breast cancer cell migration and invasion through regulation of epithelial-to-mesenchymal transition and suppression of MEK/ERK signaling pathway. Front Cell Dev Biol 2025; 13:1535563. [PMID: 40143970 PMCID: PMC11937081 DOI: 10.3389/fcell.2025.1535563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Objective This study aims to investigate the roles played by NaB and NaP in breast carcinogenesis by elucidating their potential anti-metastatic effects in the context of tumor migration, invasion, and EMT regulation in two distinct breast cancer cell lines, MCF-7 and MDA-MB-231. Methods The cytotoxic effect of both compounds on 3D spheroid formation was evaluated using a hanging drop assay. The anti-migratory and anti-invasive potentials of NaB and NaP were investigated through transwell migration and invasion assays. Moreover, their role in regulating epithelial-to-mesenchymal transition (EMT) was examined by assessing E-cadherin, vimentin, and β-catenin mRNA and protein expression levels through RT-qPCR and Western blot or flow cytometry. β-Catenin localization upon treatment was further visualized via immunofluorescence. Protein expression of MEK, p-MEK, ERK, and p-ERK was analyzed by Western blot. Results Our results revealed a dose- and time-dependent impairment of spheroid formation in both cell lines, with NaB exerting a more potent effect than NaP. Both SCFAs were able to significantly inhibit migration and invasion of MDA-MB-231 cells following 24 h of treatment. Moreover, treatment with NaB or NaP altered the mRNA and protein profile of EMT-associated markers and abrogated the nuclear translocation of β-catenin. Finally, ERK and MEK phosphorylation was reduced in MDA-MB-231 and MCF-7 cells upon treatment with NaB, and less prominently with NaP. Conclusion Our study highlights the promising therapeutic potential of NaB and NaP, providing insight into their inhibitory effects on 3D formation, migration, and invasion through EMT regulation and deactivation of MEK/ERK signaling in breast cancer.
Collapse
Affiliation(s)
- Dania Mahmoud Kharazi
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Louna Karam
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Charbel El Boustany
- Department of Laboratory Science, Faculty of Public Health - Branch 2, Lebanese University, Fanar, Lebanon
| | - José-Noel Ibrahim
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| |
Collapse
|
2
|
Li F, Zhuo L, Xie F, Luo H, Li Y, Lin H, Li X. Exploration of small molecule compounds targeting abdominal aortic aneurysm based on CMap database and molecular dynamics simulation. Vascular 2024:17085381241273289. [PMID: 39155144 DOI: 10.1177/17085381241273289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
OBJECTIVE The mitigation of abdominal aortic aneurysm (AAA) growth through pharmaceutical intervention offers the potential to avert the perils associated with AAA rupture and the subsequent need for surgical intervention. Nevertheless, the existing effective drugs for AAA treatment are limited, necessitating a pressing exploration for novel therapeutic medications. METHODS AAA-related transcriptome data were downloaded from GEO, and differentially expressed genes (DEGs) in AAA tissue were screened for GO and KEGG enrichment analyses. Small molecule compounds and their target proteins with negative connectivity to the AAA expression profile were predicted in the Connectivity Map (CMap) database. Molecular docking and molecular dynamics simulation were performed to predict the binding of the target protein to the small molecule compound, and the MM/GBSA method was used to calculate the binding free energy. Cluster analysis was performed using the cluster tool in the GROMACS package. An AAA cell-free model was built, and CETSA experiments were used to demonstrate the binding ability of small molecules to the target protein in cells. RESULTS A total of 2244 DEGs in AAA were obtained through differential analysis, and the DEGs were mainly enriched in the tubulin binding biological function and cell cycle pathway. The CMap results showed that Apicidin had a potential therapeutic effect on AAA with a connectivity score of -97.74, and HDAC4 was the target protein of Apicidin. Based on literature, HDAC4-Apicidin was selected as the subsequent research object. The lowest affinity of Apicidin-HDAC4 molecular docking was -8.218 kcal/mol. Molecular dynamics simulation results indicated that Apicidin-HDAC4 could form a stable complex. MM/GBSA analysis showed a total binding free energy of -55.40 ± 0.79 kcal/mol, and cluster analysis showed that there were two main conformational clusters during the binding process, accounting for 22.4% and 57.8%, respectively. Apicidin could form hydrogen bonds with surrounding residues for stable binding. CETSA experiment proved the stable binding ability of Apicidin and HDAC4. CONCLUSION Apicidin inhibited HDAC4 in AAA and exhibited favorable protein-ligand interactions and stability, making it a potential candidate drug for treating AAA.
Collapse
Affiliation(s)
- Fushan Li
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Liqing Zhuo
- Department of Electrocardiography, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Fangtao Xie
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Haiping Luo
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Ying Li
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Huyu Lin
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| | - Xiaoguang Li
- Department of Vascular, Sanming First Hospital Affiliated to Fujian Medical University, Sanming, China
| |
Collapse
|
3
|
Xu L, Yan X, Wang J, Zhao Y, Liu Q, Fu J, Shi X, Su J. The Roles of Histone Deacetylases in the Regulation of Ovarian Cancer Metastasis. Int J Mol Sci 2023; 24:15066. [PMID: 37894746 PMCID: PMC10606123 DOI: 10.3390/ijms242015066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, and metastasis is the major cause of death in patients with ovarian cancer, which is regulated by the coordinated interplay of genetic and epigenetic mechanisms. Histone deacetylases (HDACs) are enzymes that can catalyze the deacetylation of histone and some non-histone proteins and that are involved in the regulation of a variety of biological processes via the regulation of gene transcription and the functions of non-histone proteins such as transcription factors and enzymes. Aberrant expressions of HDACs are common in ovarian cancer. Many studies have found that HDACs are involved in regulating a variety of events associated with ovarian cancer metastasis, including cell migration, invasion, and the epithelial-mesenchymal transformation. Herein, we provide a brief overview of ovarian cancer metastasis and the dysregulated expression of HDACs in ovarian cancer. In addition, we discuss the roles of HDACs in the regulation of ovarian cancer metastasis. Finally, we discuss the development of compounds that target HDACs and highlight their importance in the future of ovarian cancer therapy.
Collapse
Affiliation(s)
- Long Xu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
- School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Xiaoyu Yan
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Jian Wang
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Qingqing Liu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Jiaying Fu
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Xinyi Shi
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China; (L.X.); (X.Y.); (J.W.); (Y.Z.); (Q.L.); (J.F.); (X.S.)
| |
Collapse
|
4
|
Synergistic Interaction of the Class IIa HDAC Inhibitor CHDI0039 with Bortezomib in Head and Neck Cancer Cells. Int J Mol Sci 2023; 24:ijms24065553. [PMID: 36982651 PMCID: PMC10056166 DOI: 10.3390/ijms24065553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
In contrast to class I/IIb/pan histone deacetylase inhibitors (HDACi), the role of class IIa HDACi as anti-cancer chemosensitizing agents is less well understood. Here, we studied the effects of HDAC4 in particular and the class IIa HDACi CHDI0039 on proliferation and chemosensitivity in Cal27 and cisplatin-resistant Cal27CisR head and neck squamous cell cancer (HNSCC). HDAC4 and HDAC5 overexpression clones were generated. HDAC4 overexpression (Cal27_HDAC4) increased proliferation significantly compared to vector control cells (Cal27_VC). Chicken chorioallantoic membrane (CAM) studies confirmed the in vitro results: Cal27_HDAC4 tumors were slightly larger than tumors from Cal27_VC, and treatment with CHDI0039 resulted in a significant decrease in tumor size and weight of Cal27_HDAC4 but not Cal27_VC. Unlike class I/pan-HDACi, treatment with CHDI0039 had only a marginal impact on cisplatin cytotoxicity irrespective of HDAC4 and HDAC5 expression. In contrast, the combination of CHDI0039 with bortezomib was synergistic (Chou–Talalay) in MTT and caspase 3/7 activation experiments. RNAseq indicated that treatment with CHDI0039 alters the expression of genes whose up- or downregulation is associated with increased survival in HNSCC patients according to Kaplan–Meier data. We conclude that the combination of class IIa HDACi with proteasome inhibitors constitutes an effective treatment option for HNSCC, particularly for platinum-resistant cancers.
Collapse
|
5
|
Si L, Lai T, Zhao J, Jin Y, Qi M, Li M, Fu H, Shi X, Ma L, Guo R. Identification of a novel pyridine derivative with inhibitory activity against ovarian cancer progression in vivo and in vitro. Front Pharmacol 2022; 13:1064485. [PMID: 36467091 PMCID: PMC9715740 DOI: 10.3389/fphar.2022.1064485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 10/31/2022] [Indexed: 11/21/2022] Open
Abstract
Ovarian cancer is the second leading cause of death of female gynecological malignant tumor patients worldwide. Although surgery and chemotherapy have achieved dramatic achievement, the mortality remains high, resulting in the demand for new specific drug discovery. Disrupting ovarian cancer growth via histone deacetylase (HDAC) inhibition is a strategy for cancer therapy or prevention. In this work, we synthesized a novel pyridine derivative named compound H42 and investigated its anti-cancer activity in vivo and in vitro. We found that compound H42 inhibited ovarian cancer cell proliferation with IC50 values of 0.87 μM (SKOV3) and 5.4 μM (A2780). Further studies confirmed that compound H42 induced apoptosis, intracellular ROS production, and DNA damage. Moreover, compound H42 downregulated the expression of histone deacetylase 6 (HDAC6) with a distinct increase in the acetylation of α-tubulin and heat shock protein 90 (HSP90), followed by the degradation of cyclin D1, resulting in cell cycle arrest at the G0/G1 phase. Importantly, ectopic expression of HDAC6 induced deacetylation of HSP90 and α-tubulin, while HDAC6 knockdown upregulated the acetylation of HSP90 and α-tubulin. However, in the nude xenograft mouse study, compound H42 treatment can inhibit ovarian cancer growth without obvious toxicity. These findings indicated that compound H42 inhibited ovarian cancer cell proliferation through inducing cell cycle arrest at the G0/G1 phase via regulating HDAC6-mediated acetylation, suggesting compound H42 could serve as a lead compound for further development of ovarian cancer therapeutic agents.
Collapse
Affiliation(s)
- Lulu Si
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Tianjiao Lai
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Junru Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuxi Jin
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Meng Qi
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Mingyue Li
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Hanlin Fu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| | - Xiaojing Shi
- Laboratory Animal Center, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- China Meheco Topfond Pharmaceutical Co., Zhumadian, China
- Key Laboratory of Cardio-cerebrovascular Drug, Zhumadian, Henan, China
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Medical Key Laboratory for Prevention and Treatment of Malignant Gynecological Tumor, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Alseksek RK, Ramadan WS, Saleh E, El-Awady R. The Role of HDACs in the Response of Cancer Cells to Cellular Stress and the Potential for Therapeutic Intervention. Int J Mol Sci 2022; 23:8141. [PMID: 35897717 PMCID: PMC9331760 DOI: 10.3390/ijms23158141] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 02/01/2023] Open
Abstract
Throughout the process of carcinogenesis, cancer cells develop intricate networks to adapt to a variety of stressful conditions including DNA damage, nutrient deprivation, and hypoxia. These molecular networks encounter genomic instability and mutations coupled with changes in the gene expression programs due to genetic and epigenetic alterations. Histone deacetylases (HDACs) are important modulators of the epigenetic constitution of cancer cells. It has become increasingly known that HDACs have the capacity to regulate various cellular systems through the deacetylation of histone and bounteous nonhistone proteins that are rooted in complex pathways in cancer cells to evade death pathways and immune surveillance. Elucidation of the signaling pathways involved in the adaptive responses to cellular stress and the role of HDACs may lead to the development of novel therapeutic agents. In this article, we overview the dominant stress types including metabolic, oxidative, genotoxic, and proteotoxic stress imposed on cancer cells in the context of HDACs, which guide stress adaptation responses. Next, we expose a closer view on the therapeutic interventions and clinical trials that involve HDACs inhibitors, in addition to highlighting the impact of using HDAC inhibitors in combination with stress-inducing agents for the management of cancer and to overcome the resistance to current cancer therapy.
Collapse
Affiliation(s)
- Rahma K. Alseksek
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ekram Saleh
- Clinical Biochemistry and Molecular Biology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 12613, Egypt;
| | - Raafat El-Awady
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
7
|
Qiu X, Zhu L, Wang H, Tan Y, Yang Z, Yang L, Wan L. From natural products to HDAC inhibitors: An overview of drug discovery and design strategy. Bioorg Med Chem 2021; 52:116510. [PMID: 34826681 DOI: 10.1016/j.bmc.2021.116510] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 02/08/2023]
Abstract
Histone deacetylases (HDACs) play a key role in the homeostasis of protein acetylation in histones and have recently emerged as a therapeutic target for numerous diseases. The inhibition of HDACs may block angiogenesis, arrest cell growth, and lead to differentiation and apoptosis in tumour cells. Thus, HDAC inhibitors (HDACi) have received increasing attention and many of which are developed from natural sources. In the past few decades, naturally occurring HDACi have been identified to have potent anticancer activities, some of which have demonstrated promising therapeutic effects on haematological malignancies. In this review, we summarized the discovery and modification of HDAC inhibitors from natural sources, novel drug design that uses natural products as parent nuclei, and dual target design strategies that combine HDAC with non-HDAC targets.
Collapse
Affiliation(s)
- Xiang Qiu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lv Zhu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuang Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Linyu Yang
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Li Wan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
8
|
Fan Q, Li L, Wang TL, Emerson RE, Xu Y. A Novel ZIP4-HDAC4-VEGFA Axis in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13153821. [PMID: 34359722 PMCID: PMC8345154 DOI: 10.3390/cancers13153821] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/25/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Despite tremendous research efforts, epithelial ovarian cancer (EOC) remains one of the most difficult cancers to detect early and treat successfully for >5-year survival. We have recently shown that ZIP4, a zinc transporter, is a novel cancer stem cell (CSC) marker and a therapeutic target for EOC. The current work focuses on developing new strategies to target ZIP4 and inhibit its CSC activities in EOC. We found that cells expressing high levels of ZIP4 were supersensitive to a group of inhibitors called HDACis. One of the major targets of these inhibitors is a protein called HDAC4. We revealed the new molecular bases for the ZIP4-HDAC4 axis and tested the efficacies of targeting this axis in the lab and in mouse models. Our study provides a new mechanistic-based targeting strategy for EOC. Abstract We have recently identified ZIP4 as a novel cancer stem cell (CSC) marker in high-grade serous ovarian cancer (HGSOC). While it converts drug-resistance to cisplatin (CDDP), we unexpectedly found that ZIP4 induced sensitization of HGSOC cells to histone deacetylase inhibitors (HDACis). Mechanistically, ZIP4 selectively upregulated HDAC IIa HDACs, with little or no effect on HDACs in other classes. HDAC4 knockdown (KD) and LMK-235 inhibited spheroid formation in vitro and tumorigenesis in vivo, with hypoxia inducible factor-1 alpha (HIF1α) and endothelial growth factor A (VEGFA) as functional downstream mediators of HDAC4. Moreover, we found that ZIP4, HDAC4, and HIF1α were involved in regulating secreted VEGFA in HGSOC cells. Furthermore, we tested our hypothesis that co-targeting CSC via the ZIP4-HDAC4 axis and non-CSC using CDDP is necessary and highly effective by comparing the effects of ZIP4-knockout/KD, HDAC4-KD, and HDACis, in the presence or absence of CDDP on tumorigenesis in mouse models. Our results showed that the co-targeting strategy was highly effective. Finally, data from human HGSOC tissues showed that ZIP4 and HDAC4 were upregulated in a subset of recurrent tumors, justifying the clinical relevance of the study. In summary, our study provides a new mechanistic-based targeting strategy for HGSOC.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
| | - Lihong Li
- Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, 600 North Wolfe St., Baltimore, MD 21287, USA;
| | - Tian-Li Wang
- Department of Gynecology, Oncology, and Pathology, Johns Hopkins Medical Institutions, 1550 Orleans Street, Baltimore, MD 21231, USA;
| | - Robert E. Emerson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana University Health Pathology Laboratory, 350 W. 11th Street, Room 4010, Indianapolis, IN 46202, USA;
| | - Yan Xu
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, 950 W. Walnut St. R2-E380, Indianapolis, IN 46202, USA;
- Correspondence: ; Tel.: +1-(317)-274-3972
| |
Collapse
|
9
|
Herlan CN, Sonnefeld A, Gloge T, Brückel J, Schlee LC, Muhle-Goll C, Nieger M, Bräse S. Macrocyclic Tetramers-Structural Investigation of Peptide-Peptoid Hybrids. Molecules 2021; 26:molecules26154548. [PMID: 34361700 PMCID: PMC8348019 DOI: 10.3390/molecules26154548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 11/16/2022] Open
Abstract
Outstanding affinity and specificity are the main characteristics of peptides, rendering them interesting compounds for basic and medicinal research. However, their biological applicability is limited due to fast proteolytic degradation. The use of mimetic peptoids overcomes this disadvantage, though they lack stereochemical information at the α-carbon. Hybrids composed of amino acids and peptoid monomers combine the unique properties of both parent classes. Rigidification of the backbone increases the affinity towards various targets. However, only little is known about the spatial structure of such constrained hybrids. The determination of the three-dimensional structure is a key step for the identification of new targets as well as the rational design of bioactive compounds. Herein, we report the synthesis and the structural elucidation of novel tetrameric macrocycles. Measurements were taken in solid and solution states with the help of X-ray scattering and NMR spectroscopy. The investigations made will help to find diverse applications for this new, promising compound class.
Collapse
Affiliation(s)
- Claudine Nicole Herlan
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Anna Sonnefeld
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Thomas Gloge
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Julian Brückel
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Luisa Chiara Schlee
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
| | - Claudia Muhle-Goll
- Institute for Biological Interfaces 4, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany; (A.S.); (T.G.); (C.M.-G.)
| | - Martin Nieger
- Department of Chemistry, University of Helsinki, P.O. Box 55 (A.I. Virtasen aukio 1), FIN-00014 Helsinki, Finland;
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany; (C.N.H.); (J.B.); (L.C.S.)
- Institute of Biological and Chemical Systems—Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Correspondence:
| |
Collapse
|
10
|
Russell JJ, Grisanti LA, Brown SM, Bailey CA, Bender SB, Chandrasekar B. Reversion inducing cysteine rich protein with Kazal motifs and cardiovascular diseases: The RECKlessness of adverse remodeling. Cell Signal 2021; 83:109993. [PMID: 33781845 DOI: 10.1016/j.cellsig.2021.109993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022]
Abstract
The Reversion Inducing Cysteine Rich Protein With Kazal Motifs (RECK) is a glycosylphosphatidylinositol (GPI) anchored membrane-bound regulator of matrix metalloproteinases (MMPs). It is expressed throughout the body and plays a role in extracellular matrix (ECM) homeostasis and inflammation. In initial studies, RECK expression was found to be downregulated in various invasive cancers and associated with poor prognostic outcome. Restoring RECK, however, has been shown to reverse the metastatic phenotype. Downregulation of RECK expression is also reported in non-malignant diseases, such as periodontal disease, renal fibrosis, and myocardial fibrosis. As such, RECK induction has therapeutic potential in several chronic diseases. Mechanistically, RECK negatively regulates various matrixins involved in cell migration, proliferation, and adverse remodeling by targeting the expression and/or activation of multiple MMPs, A Disintegrin And Metalloproteinase Domain-Containing Proteins (ADAMs), and A Disintegrin And Metalloproteinase With Thrombospondin Motifs (ADAMTS). Outside of its role in remodeling, RECK has also been reported to exert anti-inflammatory effects. In cardiac diseases, for example, it has been shown to counteract several downstream effectors of Angiotensin II (Ang-II) that play a role in adverse cardiac and vascular remodeling, such as Interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/glycoprotein 130 (IL-6 signal transducer) signaling and Epidermal Growth Factor Receptor (EGFR) transactivation. This review article focuses on the current understanding of the multifunctional effects of RECK and how its downregulation may contribute to adverse cardiovascular remodeling.
Collapse
Affiliation(s)
- Jacob J Russell
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America.
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| | - B Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America; Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| |
Collapse
|
11
|
Montalvo-Casimiro M, González-Barrios R, Meraz-Rodriguez MA, Juárez-González VT, Arriaga-Canon C, Herrera LA. Epidrug Repurposing: Discovering New Faces of Old Acquaintances in Cancer Therapy. Front Oncol 2020; 10:605386. [PMID: 33312959 PMCID: PMC7708379 DOI: 10.3389/fonc.2020.605386] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Gene mutations are strongly associated with tumor progression and are well known in cancer development. However, recently discovered epigenetic alterations have shown the potential to greatly influence tumoral response to therapy regimens. Such epigenetic alterations have proven to be dynamic, and thus could be restored. Due to their reversible nature, the promising opportunity to improve chemotherapy response using epigenetic therapy has arisen. Beyond helping to understand the biology of the disease, the use of modern clinical epigenetics is being incorporated into the management of the cancer patient. Potential epidrug candidates can be found through a process known as drug repositioning or repurposing, a promising strategy for the discovery of novel potential targets in already approved drugs. At present, novel epidrug candidates have been identified in preclinical studies and some others are currently being tested in clinical trials, ready to be repositioned. This epidrug repurposing could circumvent the classic paradigm where the main focus is the development of agents with one indication only, while giving patients lower cost therapies and a novel precision medical approach to optimize treatment efficacy and reduce toxicity. This review focuses on the main approved epidrugs, and their druggable targets, that are currently being used in cancer therapy. Also, we highlight the importance of epidrug repurposing by the rediscovery of known chemical entities that may enhance epigenetic therapy in cancer, contributing to the development of precision medicine in oncology.
Collapse
Affiliation(s)
- Michel Montalvo-Casimiro
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Rodrigo González-Barrios
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Marco Antonio Meraz-Rodriguez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | | | - Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
12
|
Shi J, Xu X, Zhang D, Zhang J, Yang H, Li C, Li R, Wei X, Luan W, Liu P. Long non-coding RNA PTPRG-AS1 promotes cell tumorigenicity in epithelial ovarian cancer by decoying microRNA-545-3p and consequently enhancing HDAC4 expression. J Ovarian Res 2020; 13:127. [PMID: 33099316 PMCID: PMC7585679 DOI: 10.1186/s13048-020-00723-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Long non-coding RNA PTPRG antisense RNA 1 (PTPRG-AS1) deregulation has been reported in various human malignancies and identified as an important modulator of cancer development. Few reports have focused on the detailed role of PTPRG-AS1 in epithelial ovarian cancer (EOC) and its underlying mechanism. This study aimed to determine the physiological function of PTPRG-AS1 in EOC. A series of experiments were also performed to identify the mechanisms through which PTPRG-AS1 exerts its function in EOC. METHODS Reverse transcription-quantitative polymerase chain reaction was used to determine PTPRG-AS1 expression in EOC tissues and cell lines. PTPRG-AS1 was silenced in EOC cells and studied with respect to cell proliferation, apoptosis, migration, and invasion in vitro and tumor growth in vivo. The putative miRNAs that target PTPRG-AS1 were predicted using bioinformatics analysis and further confirmed in luciferase reporter and RNA immunoprecipitation assays. RESULTS Our data verified the upregulation of PTPRG-AS1 in EOC tissues and cell lines. High PTPRG-AS1 expression was associated with shorter overall survival in patients with EOC. Functionally, EOC cell proliferation, migration, invasion in vitro, and tumor growth in vivo were suppressed by PTPRG-AS1 silencing. In contrast, cell apoptosis was promoted by loss of PTPRG-AS1. Regarding the mechanism, PTPRG-AS1 could serve as a competing endogenous RNA in EOC cells by decoying microRNA-545-3p (miR-545-3p), thereby elevating histone deacetylase 4 (HDAC4) expression. Furthermore, rescue experiments revealed that PTPRG-AS1 knockdown-mediated effects on EOC cells were, in part, counteracted by the inhibition of miR-545-3p or restoration of HDAC4. CONCLUSIONS PTPRG-AS1 functioned as an oncogenic lncRNA that aggravated the malignancy of EOC through the miR-545-3p/HDAC4 ceRNA network. Thus, targeting the PTPRG-AS1/miR-545-3p/HDAC4 pathway may be a novel strategy for EOC anticancer therapy.
Collapse
Affiliation(s)
- Juanjuan Shi
- Department of Gynaecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Jinan, 277599 Shandong China ,Department of Gynaecology, Tengzhou Center People’s Hospital, Zaozhuang, 277500 Shandong China
| | - Xijian Xu
- Department of Gynaecology, Rizhao Central Hospital, Rizhao, 276800 Shandong China
| | - Dan Zhang
- Department of TCM Pharmacy, Tengzhou Center People’s Hospital, Zaozhuang, 277500 Shandong China
| | - Jiuyan Zhang
- Department of Clinical Pharmacy, Tengzhou Center People’s Hospital, Zaozhuang, 277500 Shandong China
| | - Hui Yang
- Department of Gynaecology, Tengzhou Center People’s Hospital, Zaozhuang, 277500 Shandong China
| | - Chang Li
- Department of Pathology, Tengzhou Center People’s Hospital, Zaozhuang, 277500 Shandong China
| | - Rui Li
- Department of Gynaecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Jinan, 277599 Shandong China
| | - Xuan Wei
- Department of Gynaecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Jinan, 277599 Shandong China
| | - Wenqing Luan
- Department of Gynaecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Jinan, 277599 Shandong China
| | - Peishu Liu
- Department of Gynaecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Jinan, 277599, Shandong, China.
| |
Collapse
|
13
|
Akone SH, Ntie-Kang F, Stuhldreier F, Ewonkem MB, Noah AM, Mouelle SEM, Müller R. Natural Products Impacting DNA Methyltransferases and Histone Deacetylases. Front Pharmacol 2020; 11:992. [PMID: 32903500 PMCID: PMC7438611 DOI: 10.3389/fphar.2020.00992] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Abstract
Epigenetics refers to heritable changes in gene expression and chromatin structure without change in a DNA sequence. Several epigenetic modifications and respective regulators have been reported. These include DNA methylation, chromatin remodeling, histone post-translational modifications, and non-coding RNAs. Emerging evidence has revealed that epigenetic dysregulations are involved in a wide range of diseases including cancers. Therefore, the reversible nature of epigenetic modifications concerning activation or inhibition of enzymes involved could be promising targets and useful tools for the elucidation of cellular and biological phenomena. In this review, emphasis is laid on natural products that inhibit DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) making them promising candidates for the development of lead structures for anticancer-drugs targeting epigenetic modifications. However, most of the natural products targeting HDAC and/or DNMT lack isoform selectivity, which is important for determining their potential use as therapeutic agents. Nevertheless, the structures presented in this review offer the well-founded basis that screening and chemical modifications of natural products will in future provide not only leads to the identification of more specific inhibitors with fewer side effects, but also important features for the elucidation of HDAC and DNMT function with respect to cancer treatment.
Collapse
Affiliation(s)
- Sergi Herve Akone
- Department of Chemistry, Faculty of Science, University of Douala, Douala, Cameroon
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
- Institute for Pharmacy, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
- Institut für Botanik, Technische Universität Dresden, Dresden, Germany
| | - Fabian Stuhldreier
- Medical Faculty, Institute of Molecular Medicine I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Alexandre Mboene Noah
- Department of Biochemistry, Faculty of Science, University of Douala, Douala, Cameroon
| | | | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research and Department of Pharmacy, Saarland University, Saarbrücken, Germany
| |
Collapse
|
14
|
Gerlitz G. The Emerging Roles of Heterochromatin in Cell Migration. Front Cell Dev Biol 2020; 8:394. [PMID: 32528959 PMCID: PMC7266953 DOI: 10.3389/fcell.2020.00394] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022] Open
Abstract
Cell migration is a key process in health and disease. In the last decade an increasing attention is given to chromatin organization in migrating cells. In various types of cells induction of migration leads to a global increase in heterochromatin levels. Heterochromatin is required for optimal cell migration capabilities, since various interventions with heterochromatin formation impeded the migration rate of numerous cell types. Heterochromatin supports the migration process by affecting both the mechanical properties of the nucleus as well as the genetic processes taking place within it. Increased heterochromatin levels elevate nuclear rigidity in a manner that allows faster cell migration in 3D environments. Condensed chromatin and a more rigid nucleus may increase nuclear durability to shear stress and prevent DNA damage during the migration process. In addition, heterochromatin reorganization in migrating cells is important for induction of migration-specific transcriptional plan together with inhibition of many other unnecessary transcriptional changes. Thus, chromatin organization appears to have a key role in the cellular migration process.
Collapse
Affiliation(s)
- Gabi Gerlitz
- Department of Molecular Biology and Ariel Center for Applied Cancer Research, Faculty of Life Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
15
|
Histone Deacetylases (HDACs): Evolution, Specificity, Role in Transcriptional Complexes, and Pharmacological Actionability. Genes (Basel) 2020; 11:genes11050556. [PMID: 32429325 PMCID: PMC7288346 DOI: 10.3390/genes11050556] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Histone deacetylases (HDACs) are evolutionary conserved enzymes which operate by removing acetyl groups from histones and other protein regulatory factors, with functional consequences on chromatin remodeling and gene expression profiles. We provide here a review on the recent knowledge accrued on the zinc-dependent HDAC protein family across different species, tissues, and human pathologies, specifically focusing on the role of HDAC inhibitors as anti-cancer agents. We will investigate the chemical specificity of different HDACs and discuss their role in the human interactome as members of chromatin-binding and regulatory complexes.
Collapse
|
16
|
Moreira-Silva F, Camilo V, Gaspar V, Mano JF, Henrique R, Jerónimo C. Repurposing Old Drugs into New Epigenetic Inhibitors: Promising Candidates for Cancer Treatment? Pharmaceutics 2020; 12:E410. [PMID: 32365701 PMCID: PMC7284583 DOI: 10.3390/pharmaceutics12050410] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/24/2022] Open
Abstract
Epigenetic alterations, as a cancer hallmark, are associated with cancer initiation, progression and aggressiveness. Considering, however, that these alterations are reversible, drugs that target epigenetic machinery may have an inhibitory effect upon cancer treatment. The traditional drug discovery pathway is time-consuming and expensive, and thus, new and more effective strategies are required. Drug Repurposing (DR) comprises the discovery of a new medical indication for a drug that is approved for another indication, which has been recalled, that was not accepted or failed to prove efficacy. DR presents several advantages, mainly reduced resources, absence of the initial target discovery process and the reduced time necessary for the drug to be commercially available. There are numerous old drugs that are under study as repurposed epigenetic inhibitors which have demonstrated promising results in in vitro tumor models. Herein, we summarize the DR process and explore several repurposed drugs with different epigenetic targets that constitute promising candidates for cancer treatment, highlighting their mechanisms of action.
Collapse
Affiliation(s)
- Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (F.M.-S.); (V.C.)
| | - Vânia Camilo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (F.M.-S.); (V.C.)
| | - Vítor Gaspar
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (V.G.); (J.F.M.)
| | - João F. Mano
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; (V.G.); (J.F.M.)
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP) and Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal;
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (F.M.-S.); (V.C.)
| |
Collapse
|
17
|
Luo J, Li H, Deziel NC, Huang H, Zhao N, Ma S, Ni X, Udelsman R, Zhang Y. Genetic susceptibility may modify the association between cell phone use and thyroid cancer: A population-based case-control study in Connecticut. ENVIRONMENTAL RESEARCH 2020; 182:109013. [PMID: 31918310 PMCID: PMC7061309 DOI: 10.1016/j.envres.2019.109013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 05/17/2023]
Abstract
Emerging studies have provided evidence on the carcinogenicity of radiofrequency radiation (RFR) from cell phones. This study aims to test the genetic susceptibility on the association between cell phone use and thyroid cancer. Population-based case-control study was conducted in Connecticut between 2010 and 2011 including 440 thyroid cancer cases and 465 population-based controls with genotyping information for 823 single nucleotide polymorphisms (SNPs) in 176 DNA genes. We used multivariate unconditional logistic regression models to estimate the genotype-environment interaction between each SNP and cell phone use and to estimate the association with cell phone use in populations according to SNP variants. Ten SNPs had P < 0.01 for interaction in all thyroid cancers. In the common homozygote groups, no association with cell phone use was observed. In the variant group (heterozygotes and rare homozygotes), cell phone use was associated with an increased risk for rs11070256 (odds ratio (OR): 2.36, 95% confidence interval (CI): 1.30-4.30), rs1695147 (OR: 2.52, 95% CI: 1.30-4.90), rs6732673 (OR: 1.59, 95% CI: 1.01-2.49), rs396746 (OR: 2.53, 95% CI: 1.13-5.65), rs12204529 (OR: 2.62, 95% CI: 1.33-5.17), and rs3800537 (OR: 2.64, 95% CI: 1.30-5.36) with thyroid cancers. In small tumors, increased risk was observed for 5 SNPs (rs1063639, rs1695147, rs11070256, rs12204529 and rs3800537), In large tumors, increased risk was observed for 3 SNPs (rs11070256, rs1695147, and rs396746). Our result suggests that genetic susceptibilities modify the associations between cell phone use and risk of thyroid cancer. The findings provide more evidence for RFR carcinogenic group classification.
Collapse
Affiliation(s)
- Jiajun Luo
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Hang Li
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| | - Nicole C Deziel
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Huang Huang
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Nan Zhao
- Peking Union Medical College Hospital, Beijing, China
| | - Shuangge Ma
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Xin Ni
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing, China; Big Data and Engineering Research Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Robert Udelsman
- Endocrine Neoplasm Institute, Miami Cancer Institute, Miami, FL, USA
| | - Yawei Zhang
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA; Section of Surgical Outcomes and Epidemiology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
De U, Son JY, Jeon Y, Ha SY, Park YJ, Yoon S, Ha KT, Choi WS, Lee BM, Kim IS, Kwak JH, Kim HS. Plumbagin from a tropical pitcher plant (Nepenthes alata Blanco) induces apoptotic cell death via a p53-dependent pathway in MCF-7 human breast cancer cells. Food Chem Toxicol 2019; 123:492-500. [PMID: 30458268 DOI: 10.1016/j.fct.2018.11.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 12/16/2022]
Abstract
Plumbagin (5-hydroxy-2-methyl-1,4-naphthaquinone) has displayed antitumor activity in vitro and in animal models; however, the underlying molecular mechanisms have not been fully explored. The aim of this study was to investigate the anticancer effects of plumbagin isolated from Nepenthes alata against MCF-7 breast cancer cells. We examined the cytotoxicity, cell cycle regulation, apoptotic cell death, and generation of intracellular reactive oxygen species (ROS) in MCF-7 cells. Plumbagin exhibited potent cytotoxicity in MCF-7 cells (wild-type p53) compared to that in SK-OV-3 (null-type) human epithelial ovarian cancer cells. Specifically, plumbagin upregulated the expression of p21CIP1/WAF1 in MCF-7 cells, causing cell cycle arrest in the G2/M phase through inhibition of cyclin B1 levels. Plumbagin also significantly increased the ratio of Bax/Bcl-2 and release of cytochrome c, resulting in apoptotic cell death in MCF-7 cells. Furthermore, plumbagin dramatically increased the intracellular ROS level, whereas pretreatment with the ROS scavenger N-acetyl cysteine protected against plumbagin-induced cytotoxicity, suggesting that ROS formation plays a pivotal role in antitumor activity in MCF-7 cells. In mice bearing MCF-7 cell xenografts, plumbagin significantly reduced tumor growth and weight without apparent side effects. We therefore concluded that plumbagin exerts anticancer activity against MCF-7 cells through the generation of intracellular ROS, resulting in the induction of apoptosis via a p53-dependent pathway. This study thus identifies a new anticancer mechanism of plumbagin against p53-dependent breast cancer cells and suggests a novel strategy for overcoming of breast cancer therapy.
Collapse
Affiliation(s)
- Umasankar De
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ji Yeon Son
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yukyoung Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Song-Yi Ha
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yu Jin Park
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sungpil Yoon
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ki-Tae Ha
- School of Korean Medicine and Healthy Aging Korean Medicine Research Center, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Wahn Soo Choi
- School of Medicine, Konkuk University, Chungju, 27478, Republic of Korea
| | - Byung Mu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
19
|
Huang D, Cui L, Ahmed S, Zainab F, Wu Q, Wang X, Yuan Z. An overview of epigenetic agents and natural nutrition products targeting DNA methyltransferase, histone deacetylases and microRNAs. Food Chem Toxicol 2019; 123:574-594. [DOI: 10.1016/j.fct.2018.10.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/25/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
|
20
|
Lascano S, Lopez M, Arimondo PB. Natural Products and Chemical Biology Tools: Alternatives to Target Epigenetic Mechanisms in Cancers. CHEM REC 2018; 18:1854-1876. [PMID: 30537358 DOI: 10.1002/tcr.201800133] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/21/2022]
Abstract
DNA methylation and histone acetylation are widely studied epigenetic modifications. They are involved in numerous pathologies such as cancer, neurological disease, inflammation, obesity, etc. Since the discovery of the epigenome, numerous compounds have been developed to reverse DNA methylation and histone acetylation aberrant profile in diseases. Among them several were inspired by Nature and have a great interest as therapeutic molecules. In the quest of finding new ways to target epigenetic mechanisms, the use of chemical tools is a powerful strategy to better understand epigenetic mechanisms in biological systems. In this review we will present natural products reported as DNMT or HDAC inhibitors for anticancer treatments. We will then discuss the use of chemical tools that have been used in order to explore the epigenome.
Collapse
Affiliation(s)
- Santiago Lascano
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université de Montpellier-ENSCM, 240 avenue du Prof. E. Jeanbrau, 34296, Montpellier cedex 5, France
| | - Marie Lopez
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 CNRS-Université de Montpellier-ENSCM, 240 avenue du Prof. E. Jeanbrau, 34296, Montpellier cedex 5, France
| | - Paola B Arimondo
- Epigenetic Chemical Biology, Institut Pasteur, CNRS UMR3523, 28 rue du Docteur Roux, 75724, Paris cedex 15, France
| |
Collapse
|
21
|
Zhou L, Xu X, Liu H, Hu X, Zhang W, Ye M, Zhu X. Prognosis Analysis of Histone Deacetylases mRNA Expression in Ovarian Cancer Patients. J Cancer 2018; 9:4547-4555. [PMID: 30519361 PMCID: PMC6277648 DOI: 10.7150/jca.26780] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylases modulate the dynamic balance of histone acetylation and deacetylation in cells, which participate in epigenetic regulations. Accumulated evidence has demonstrated that histone deacetylases are associated with angiogenesis, cell proliferation and survival in a variety of human cancers. However, the expression and distinct prognostic value of histone deacetylases in ovarian cancer have not been well elucidated. In the present study, we collected the overall survival (OS), progress free survival (PFS), and histone deacetylases (HDAC1-11) mRNA expression in ovarian cancer from the Kaplan-Meier plotter online database. We investigated the relationship between histone deacetylases mRNA level and the clinicopathological parameters of the ovarian cancer patients, such as histology subtypes, clinical stages, grades and TP53 mutation. Our analysis data showed that over-expression of HDAC1, HDAC2, HDAC4, HDAC5 and HDAC11 were correlated to poor overall survival and unfavorable progress free survival in all ovarian cancer patients. Notably, the higher level of HDAC11 was associated with the worse OS and PFS for serous/ stage III+IV/ grade III/ TP53 mutation ovarian cancer patients. In conclusion, HDACs may play a crucial role in the prognosis of ovarian cancer, but it is worth noting that HDAC11 may be a biomarker for poor prognosis in ovarian cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
22
|
Ahn MY. HDAC inhibitor apicidin suppresses murine oral squamous cell carcinoma cell growth in vitro and in vivo via inhibiting HDAC8 expression. Oncol Lett 2018; 16:6552-6560. [PMID: 30405794 PMCID: PMC6202526 DOI: 10.3892/ol.2018.9468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/23/2018] [Indexed: 01/14/2023] Open
Abstract
Apicidin, a cyclic peptide histone deacetylase (HDAC) inhibitor, has been demonstrated to exhibit antitumor activity in a number of human cancer types. The present study examined the antitumor activity of apicidin in murine oral squamous cell carcinoma (OSCC) cells. Inhibition of cell proliferation and the expression of selective HDACs were determined in apicidin-treated AT-84 murine OSCC cells. A C3H mouse model with subcutaneous injection of AT-84 cells was used to assess the in vivo effect of apicidin on tumor growth. Apicidin-induced cell growth inhibition and selectively reduced HDAC8 expression in AT-84 cells. Induction of apoptosis and autophagy was observed in apicidin-treated AT-84 cells. Apicidin notably inhibited tumor growth by up to 46% relative to the control group at the end of a 14-day period in a murine tumor model. The immunohistochemistry results in tumor tissues indicated that apicidin inhibited cell proliferation and induced apoptosis and autophagy in AT-84 cell-derived tumor tissues. Overexpression of HDAC8 was observed in the nucleus and cytoplasm in tumor tissues and apicidin significantly inhibited the level of HDAC8 expression, compared with the vehicle group. These results indicated that apicidin inhibited cell proliferation through HDAC8 inhibition in murine OSCC cells in vitro and in vivo. The present study indicated that apicidin may be an effective therapeutic agent for OSCC.
Collapse
Affiliation(s)
- Mee-Young Ahn
- Department of Pharmaceutical Engineering, Division of Bio-industry, College of Medical and Life Sciences, Silla University, Busan 46958, Republic of Korea
| |
Collapse
|
23
|
Cai JY, Xu TT, Wang Y, Chang JJ, Li J, Chen XY, Chen X, Yin YF, Ni XJ. Histone deacetylase HDAC4 promotes the proliferation and invasion of glioma cells. Int J Oncol 2018; 53:2758-2768. [PMID: 30272277 DOI: 10.3892/ijo.2018.4564] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 08/07/2018] [Indexed: 11/06/2022] Open
Abstract
Glioma is the most lethal type of primary brain tumor characterized by aggressiveness and a poor prognosis. Histone deacetylase 4 (HDAC4) is frequently dysregulated in human malignancies. However, its biological functions in the development of glioma are not fully understood. The present study aimed to evaluate HDAC4 expression in human glioma and to elucidate the mechanistic role of HDAC4 in glioma. The results suggested that HDAC4 was significantly upregulated in glioma tissues and a number of glioma cell lines compared with adjacent non-tumor tissues and the non-cancerous human glial cell line SVG p12, respectively (P<0.05). The proliferation, adenosine triphosphate (ATP) levels and invasion ability were substantially enhanced in U251 cells with HDAC4 overexpression, and suppressed in U251 cells with a knockdown of HDAC4 compared with that in U251 cells transfected with the negative control. Knockdown of HDAC4 resulted in cell cycle arrest at the G0/G1 phase and induced the increase of reactive oxygen species level in U251 cells. Furthermore, HDAC4 overexpression was revealed to substantially inhibit the expression of cyclin-dependent kinase (CDK) inhibitors p21 and p27, and the expression of E-cadherin and β‑catenin in glioma U251 cells. Knockdown of HDAC4 substantially promoted the expression of CDK1 and CDK2 and vimentin in glioma U251 cells. Mechanistically, the results of the present study demonstrated that HDAC4 displayed a significant upregulation in glioma, and promoted glioma cell proliferation and invasion mediated through the repression of p21, p27, E-cadherin and β‑catenin, and the potentiation of CDK1, CDK2 and vimentin. Altogether, the present study revealed that HDAC4 overexpression was central for the tumorigenesis of glioma, which may serve as a useful prognostic biomarker and potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Jun-Yan Cai
- Department of Rehabilitation, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Tong-Tong Xu
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ye Wang
- School of Medicine, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jing-Jian Chang
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jian Li
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiao-Yang Chen
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xi Chen
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi-Fei Yin
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xue-Jun Ni
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
24
|
Andrade SN, Evangelista FCG, Seckler D, Marques DR, Freitas TR, Nunes RR, Oliveira JT, Ribeiro RIMA, Santos HB, Thomé RG, Taranto AG, Santos FV, Viana GHR, Freitas RP, Humberto JL, Sabino ADP, Hilário FF, Varotti FP. Synthesis, cytotoxic activity, and mode of action of new Santacruzamate A analogs. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2244-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Ateia IM, Sutthiboonyapan P, Kamarajan P, Jin T, Godovikova V, Kapila YL, Fenno JC. Treponema denticola increases MMP-2 expression and activation in the periodontium via reversible DNA and histone modifications. Cell Microbiol 2018; 20. [PMID: 29205773 DOI: 10.1111/cmi.12815] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022]
Abstract
Host-derived matrix metalloproteinases (MMPs) and bacterial proteases mediate destruction of extracellular matrices and supporting alveolar bone in periodontitis. The Treponema denticola dentilisin protease induces MMP-2 expression and activation in periodontal ligament (PDL) cells, and dentilisin-mediated activation of pro-MMP-2 is required for cellular fibronectin degradation. Here, we report that T. denticola regulates MMP-2 expression through epigenetic modifications in the periodontium. PDL cells were treated with epigenetic enzyme inhibitors before or after T. denticola challenge. Fibronectin fragmentation, MMP-2 expression, and activation were assessed by immunoblot, zymography, and qRT-PCR, respectively. Chromatin modification enzyme expression in T. denticola-challenged PDL cells and periodontal tissues were evaluated using gene arrays. Several classes of epigenetic enzymes showed significant alterations in transcription in diseased tissue and T. denticola-challenged PDL cells. T. denticola-mediated MMP-2 expression and activation were significantly reduced in PDL cells treated with inhibitors of aurora kinases and histone deacetylases. In contrast, DNA methyltransferase inhibitors had little effect, and inhibitors of histone acetyltransferases, methyltransferases, and demethylases exacerbated T. denticola-mediated MMP-2 expression and activation. Chronic epigenetic changes in periodontal tissues mediated by T. denticola or other oral microbes may contribute to the limited success of conventional treatment of chronic periodontitis and may be amenable to therapeutic reversal.
Collapse
Affiliation(s)
- Islam M Ateia
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontics and Oral Medicine, University of Mansoura Faculty of Dentistry, Mansoura, Egypt
| | - Pimchanok Sutthiboonyapan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontology, Chulalongkorn University Faculty of Dentistry, Bangkok, Thailand
| | - Pachiyappan Kamarajan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - Taocong Jin
- Office of Research, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Valentina Godovikova
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Yvonne L Kapila
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Ahn MY, Yoon JH. Histone deacetylase 7 silencing induces apoptosis and autophagy in salivary mucoepidermoid carcinoma cells. J Oral Pathol Med 2017; 46:276-283. [PMID: 28178760 DOI: 10.1111/jop.12560] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The overexpression of histone deacetylases (HDACs) has been observed in many cancers, and inhibition of specific HDACs has emerged as a new target for cancer therapy. We found that HDAC7 expression was selectively reduced by HDAC inhibitor apicidin in salivary mucoepidermoid carcinoma (MEC) cells. Here, we show that HDAC7 suppression has a potent antitumor effect in MEC cells. METHODS Histone deacetylases7 was knocked down using HDAC7 siRNAs, and cell proliferation was quantified. Cell cycle progression, apoptosis, and autophagy were measured by flow cytometry and immunoblotting. RESULTS Histone deacetylases 7 siRNAs inhibited cell proliferation and c-Myc expression, increased p27 expression, and caused G2/M phase cell cycle arrest in both YD-15 and Mc3 cells. HDAC7 silencing increased the sub-G1 population, Annexin V positive apoptotic cells and cleaved caspase3 levels. HDAC7 silencing induced an increase in autophagic markers, number of acidic vesicular organelles, and LC3B II levels, and decrease in p62 levels. HDAC7 siRNAs reduced the activation of ERK. HDAC7 knockdown resulted in growth inhibition through G2/M phase cell cycle arrest and induced both apoptosis and autophagy in MEC cells. CONCLUSIONS This study indicates that inhibition of HDAC7 might become a novel and effective therapeutic approach for treating to MEC.
Collapse
Affiliation(s)
- Mee-Young Ahn
- Division of Bio-industry, Major in Pharmaceutical Engineering, College of Medical and Life Sciences, Silla University, Busan, Korea
| | - Jung-Hoon Yoon
- Department of Oral & Maxillofacial Pathology, Wonkwang Bone Regeneration Research Institute, College of Dentistry, Daejeon Dental Hospital, Wonkwang University, Daejeon, Korea
| |
Collapse
|
27
|
HDAC Inhibitors and RECK Modulate Endoplasmic Reticulum Stress in Tumor Cells. Int J Mol Sci 2017; 18:ijms18020258. [PMID: 28134767 PMCID: PMC5343794 DOI: 10.3390/ijms18020258] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 01/22/2017] [Accepted: 01/23/2017] [Indexed: 01/05/2023] Open
Abstract
In the tumor microenvironment hypoxia and nutrient deprived states can induce endoplasmic reticulum (ER) stress. If ER stress is not relieved, the tumor cells may become apoptotic. Therefore, targeting ER homeostasis is a potential strategy for cancer treatment. Various chemotherapeutic agents including histone deacetylase (HDAC) inhibitors can induce ER stress to cause cell death in cancers. Some HDAC inhibitors can prevent HDAC from binding to the specificity protein 1-binding site of the promoter of reversion-inducing cysteine-rich protein with Kazal motifs (RECK) and up-regulate RECK expression. Up-regulation of RECK expression by HDAC inhibitors has been observed in various cancer types. RECK is a tumor and metastasis suppressor gene and is critical for regulating tumor cell invasiveness and metastasis. RECK also modulates ER stress via binding to and sequestering glucose-regulated protein 78 protein, so that the transmembrane sensors, such as protein kinase RNA-like ER kinase are released to activate eukaryotic translational initiation factor 2α phosphorylation and enhance ER stress. Therefore, HDAC inhibitors may directly induce ER stress or indirectly induce this stress by up-regulating RECK in cancer cells.
Collapse
|
28
|
Yang X, Xu Y, Wang T, Shu D, Guo P, Miskimins K, Qian SY. Inhibition of cancer migration and invasion by knocking down delta-5-desaturase in COX-2 overexpressed cancer cells. Redox Biol 2017; 11:653-662. [PMID: 28157665 PMCID: PMC5288391 DOI: 10.1016/j.redox.2017.01.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 12/23/2022] Open
Abstract
We recently reported that knockdown of delta-5-desaturase (a key enzyme that converts dihomo-γ-linolenic acid, DGLA, to the downstream ω-6 arachidonic acid) promotes formation of an anti-cancer byproduct 8-hydroxyoctanoic acid from cyclooxygenase (COX)-catalyzed DGLA peroxidation. 8-hydroxyoctanoic acid can exert its growth inhibitory effect on cancer cells (e.g. colon and pancreatic cancer) by serving as a histone deacetylase inhibitor. Since histone deacetylase inhibitors have been well-known to suppress cancer cell migration and invasion, we thus tested whether knockdown of delta-5-desaturase and DGLA treatment could also be used to inhibit cancer migration and invasion of colon cancer and pancreatic cancer cells. Wound healing assay, transwell assay and western blot were used to assess cell migration and invasion as well as the associated molecular mechanisms. Formation of threshold level of 8-hydroxyoctanoic acid was quantified from COX-catalyzed DGLA peroxidation in the cancer cells that overexpress COX-2 and their delta-5-desaturases were knocked down by shRNA transfection. Our results showed that knockdown of delta-5-desaturase along with DGLA supplement not only significantly inhibited cell migration, but also improved the efficacies of 5-flurouracil and gemcitabine, two frontline chemotherapy drugs currently used in the treatment of colon and pancreatic cancer, respectively. The molecular mechanism behind these observations is that 8-hydroxyoctanoic acid inhibits histone deacetylase, resulting in downregulation of cancer metastasis promotors, e.g., MMP-2 and MMP-9 as well as upregulation of cancer metastasis suppressor, e.g. E-cadherin. For the first time, we demonstrated that we could take the advantage of the common phenomenon of COX-2 overexpression in cancers to inhibit cancer cell migration and invasion. With the shifting paradigm of COX-2 cancer biology, our research outcome may provide us a novel cancer treatment strategy. High level of COX-2 could be used to inhibit cancer cell migration and invasion. 8-hydroxyoctanoic acid suppresses cancer migration and invasion via inhibiting HDAC. D5D knockdown and DGLA improves efficacy of chemotherapy to inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Yi Xu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Tao Wang
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - Dan Shu
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, and College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, and College of Medicine, Ohio State University, Columbus, OH 43210, USA
| | - Keith Miskimins
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, USA
| | - Steven Y Qian
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58108, USA.
| |
Collapse
|
29
|
Jang SH, Chen H, Gregersen PK, Diamond B, Kim SJ. Kruppel-like factor4 regulates PRDM1 expression through binding to an autoimmune risk allele. JCI Insight 2017; 2:e89569. [PMID: 28097234 DOI: 10.1172/jci.insight.89569] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A SNP identified as rs548234, which is found in PRDM1, the gene that encodes BLIMP1, is a risk allele associated with systemic lupus erythematosus (SLE). BLIMP1 expression was reported to be decreased in women with the PRDM1 rs548234 risk allele compared with women with the nonrisk allele in monocyte-derived DCs (MO-DCs). In this study, we demonstrate that BLIMP1 expression is regulated by the binding of Kruppel-like factor 4 (KLF4) to the risk SNP. KLF4 is highly expressed in MO-DCs but undetectable in B cells, consistent with the lack of altered expression of BLIMP1 in B cells from risk SNP carriers. Female rs548234 risk allele carriers, but not nonrisk allele carriers, exhibited decreased levels of BLIMP1 in MO-DCs, showing that the regulatory function of KLF4 is influenced by the risk allele. In addition, KLF4 directly recruits histone deacetylases (HDAC4, HDAC6, and HDAC7), established negative regulators of gene expression. Finally, the knock down of KLF4 expression reversed the inhibitory effects of the risk SNP on promoter activity and BLIMP1 expression. Therefore, the binding of KLF4 and the subsequent recruitment of HDACs represent a mechanism for reduced BLIMP1 expression in MO-DCs bearing the SLE risk allele rs548234.
Collapse
Affiliation(s)
- Su Hwa Jang
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Helen Chen
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Peter K Gregersen
- Center for Genomics and Human Genetics, The Feinstein Institute for Medical Research, Manhasset, New York, New York, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases and
| | - Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Diseases and
| |
Collapse
|
30
|
Manal M, Chandrasekar M, Gomathi Priya J, Nanjan M. Inhibitors of histone deacetylase as antitumor agents: A critical review. Bioorg Chem 2016; 67:18-42. [DOI: 10.1016/j.bioorg.2016.05.005] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/29/2016] [Accepted: 05/15/2016] [Indexed: 12/11/2022]
|
31
|
Kumar S, Chaudhary AK, Kumar R, O'Malley J, Dubrovska A, Wang X, Yadav N, Goodrich DW, Chandra D. Combination therapy induces unfolded protein response and cytoskeletal rearrangement leading to mitochondrial apoptosis in prostate cancer. Mol Oncol 2016; 10:949-65. [PMID: 27106131 DOI: 10.1016/j.molonc.2016.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/13/2016] [Accepted: 03/23/2016] [Indexed: 02/07/2023] Open
Abstract
Development of therapeutic resistance is responsible for most prostate cancer (PCa) related mortality. Resistance has been attributed to an acquired or selected cancer stem cell phenotype. Here we report the histone deacetylase inhibitor apicidin (APC) or ER stressor thapsigargin (TG) potentiate paclitaxel (TXL)-induced apoptosis in PCa cells and limit accumulation of cancer stem cells. TXL-induced responses were modulated in the presence of TG with increased accumulation of cells at G1-phase, rearrangement of the cytoskeleton, and changes in cytokine release. Cytoskeletal rearrangement was associated with modulation of the cytoplasmic and mitochondrial unfolded protein response leading to mitochondrial dysfunction and release of proapoptotic proteins from mitochondria. TXL in combination with APC or TG enhanced caspase activation. Importantly, TXL in combination with TG induced caspase activation and apoptosis in X-ray resistant LNCaP cells. Increased release of transforming growth factor-beta (TGF-β) was observed while phosphorylated β-catenin level was suppressed with TXL combination treatments. This was accompanied by a decrease in the CD44(+)CD133(+) cancer stem cell-like population, suggesting treatment affects cancer stem cell properties. Taken together, combination treatment with TXL and either APC or TG induces efficient apoptosis in both proliferating and cancer stem cells, suggesting this therapeutic combination may overcome drug resistance and recurrence in PCa.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse, Dresden, Germany; German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Xinjiang Wang
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
32
|
Huang C, Sun Y, Shen M, Zhang X, Gao P, Duan Y. Altered Cell Cycle Arrest by Multifunctional Drug-Loaded Enzymatically-Triggered Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2016; 8:1360-1370. [PMID: 26698111 DOI: 10.1021/acsami.5b10241] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
cRGD-targeting matrix metalloproteinase (MMP)-sensitive nanoparticles [PLGA-PEG1K-cRGD/PLGA-peptide-PEG5K (NPs-cRGD)] were successfully developed. Au-Pt(IV) nanoparticles, PTX, and ADR were encapsulated into NPs-RGD separately. The effects of the drug-loaded nanoparticles on the cell cycle were investigated. Here, we showed that higher cytotoxicity of drug-loaded nanoparticles was related to the cell cycle arrest, compared to that of free drugs. The NPs-cRGD studied here did not disrupt cell cycle progression. The cell cycle of Au-Pt(IV)@NPs-cRGD showed a main S phase arrest in all phases of the cell cycle phase, especially in G0/G1 phase. PTX@NPs-cRGD and ADR@NPs-cRGD showed a higher ratio of G2/M and S phase arrest than the free drugs, respectively. Cells in G0/G1 and S phases of the cell cycle had a higher uptake ratio of NPs-cRGD. A nutrient deprivation or an increase in the requirement of nutrients in tumor cells could promote the uptake of nanoparticles from the microenvironments. In vivo, NPs-cRGD could efficiently accumulate at tumor sites. The inhibition of tumor growth coupled with cell cycle arrest is in line with that in vitro. On the basis of our results, we propose that future studies on nanoparticle action mechanism should consider the cell cycle, which could be different from free drugs. Understanding the actions of cell cycle arrest could affect the application of nanomedicine in the clinic.
Collapse
Affiliation(s)
- Can Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Xiangyu Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Pei Gao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
33
|
Shen YF, Wei AM, Kou Q, Zhu QY, Zhang L. Histone deacetylase 4 increases progressive epithelial ovarian cancer cells via repression of p21 on fibrillar collagen matrices. Oncol Rep 2015; 35:948-54. [PMID: 26572940 DOI: 10.3892/or.2015.4423] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 10/13/2015] [Indexed: 11/05/2022] Open
Abstract
Histone deacetylase (HDAC) 4 is an emerging target in cancer therapeutics, but little is known about the function of HDAC4 in gynecologic malignancies. Therefore we investigated the mechanism of HDAC4 promoting the proliferation of epithelial ovarian cancer cells (OV). In this study, we observed that the proliferation of cells with HDAC4 inhibitor Trichostatin A (TSA) treatment was markedly decreased, Further, we showed that epithelial ovarian cancer tissues with stage III/IV had higher HDAC4 expression, compared to that with stage I/II. We examined first that the HDAC4 expression was increased in response to fibrillar collagen matrices. In addition, we found that HDAC4 was retained in the nucleus by regulation of PP1α, which regulated HDAC4 cellular fraction via phosphorylation of HDAC4. In addition, we found that HDAC4 bound to Sp1 in epithelial ovarian cancer cells. Finally, ovarian cancer cell line OVCAR3 was evaluated via gain/loss-of-function of HDAC4 by either overexpression of HDCA4 or knock-down of HDAC4 with shRNA. We examined both protein and mRNA of p21 by western blotting and qPCR. We performed analysis of colony formation in matrigel and migration by ECIS. Our results suggest that the accumulation of HDAC4 induced by fibrillar collagen matrices in the nucleus via co-localization of PP1α, leads to repression of the mRNA/protein of p21 and in turn promotes the proliferation and migration of epithelial ovarian cancer cells.
Collapse
Affiliation(s)
- Yu-Fei Shen
- State Key Laboratory of Reproductive Medicine, Department of Gynecology and Obstetrics, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| | - Ai-Min Wei
- State Key Laboratory of Reproductive Medicine, Department of Gynecology and Obstetrics, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| | - Qing Kou
- State Key Laboratory of Reproductive Medicine, Department of Gynecology and Obstetrics, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| | - Qiao-Ying Zhu
- State Key Laboratory of Reproductive Medicine, Department of Gynecology and Obstetrics, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| | - Lei Zhang
- State Key Laboratory of Reproductive Medicine, Department of Gynecology and Obstetrics, Nanjing Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Nanjing 210029, P.R. China
| |
Collapse
|
34
|
De U, Kundu S, Patra N, Ahn MY, Ahn JH, Son JY, Yoon JH, Moon HR, Lee BM, Kim HS. A New Histone Deacetylase Inhibitor, MHY219, Inhibits the Migration of Human Prostate Cancer Cells via HDAC1. Biomol Ther (Seoul) 2015; 23:434-441. [PMID: 26336583 PMCID: PMC4556203 DOI: 10.4062/biomolther.2015.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/01/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors are considered novel agents for cancer chemotherapy. We previously investigated MHY219, a new HDAC inhibitor, and its potent anticancer activity in human prostate cancer cells. In the present study, we evaluated MHY219 molecular mechanisms involved in the regulation of prostate cancer cell migration. Similar to suberanilohydroxamic acid (SAHA), MHY219 inhibited HDAC1 enzyme activity in a dose-dependent manner. MHY219 cytotoxicity was higher in LNCaP (IC50=0.67 μM) than in DU145 cells (IC50=1.10 μM) and PC3 cells (IC50=5.60 μM) after 48 h of treatment. MHY219 significantly inhibited the HDAC1 protein levels in LNCaP and DU145 cells at high concentrations. However, inhibitory effects of MHY219 on HDAC proteins levels varied based on the cell type. MHY219 significantly inhibited LNCaP and DU145 cells migration by down-regulation of matrix metalloprotease-1 (MMP-1) and MMP-2 and induction of tissue inhibitor of metalloproteinases-1 (TIMP-1). These results suggest that MHY219 may potentially be used as an anticancer agent to block cancer cell migration through the repression of MMP-1 and MMP-2, which is related to the reduction of HDAC1.
Collapse
Affiliation(s)
- Umasankar De
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746
| | - Soma Kundu
- College of Pharmacy, Pusan National University, Busan 609-735
| | - Nabanita Patra
- College of Pharmacy, Pusan National University, Busan 609-735
| | - Mee Young Ahn
- Department of Oral & Maxillofacial Pathology, College of Dentistry, Wonkwang Bone Regeneration Institute, Daejeon Dental Hospital, Wonkwang University, Daejeon 302-120,
Republic of Korea
| | - Ji Hae Ahn
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746
| | - Ji Yeon Son
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746
| | - Jung Hyun Yoon
- College of Pharmacy, Pusan National University, Busan 609-735
| | | | - Byung Mu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746
| |
Collapse
|
35
|
Kornienko A, Evidente A, Vurro M, Mathieu V, Cimmino A, Evidente M, van Otterlo WAL, Dasari R, Lefranc F, Kiss R. Toward a Cancer Drug of Fungal Origin. Med Res Rev 2015; 35:937-67. [PMID: 25850821 PMCID: PMC4529806 DOI: 10.1002/med.21348] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although fungi produce highly structurally diverse metabolites, many of which have served as excellent sources of pharmaceuticals, no fungi-derived agent has been approved as a cancer drug so far. This is despite a tremendous amount of research being aimed at the identification of fungal metabolites with promising anticancer activities. This review discusses the results of clinical testing of fungal metabolites and their synthetic derivatives, with the goal to evaluate how far we are from an approved cancer drug of fungal origin. Also, because in vivo studies in animal models are predictive of the efficacy and toxicity of a given compound in a clinical situation, literature describing animal cancer testing of compounds of fungal origin is reviewed as well. Agents showing the potential to advance to clinical trials are also identified. Finally, the technological challenges involved in the exploitation of fungal biodiversity and procurement of sufficient quantities of clinical candidates are discussed, and potential solutions that could be pursued by researchers are highlighted.
Collapse
Affiliation(s)
- Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - Antonio Evidente
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Napoli, Italy
| | - Maurizio Vurro
- Institute of Sciences of Food Production, National Research Council, Via Amendola 122/0, 70126 Bari, Italy
| | - Véronique Mathieu
- Laboratorie de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alessio Cimmino
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Napoli, Italy
| | - Marco Evidente
- Dipartimento di Scienze Chimiche, Università di Napoli Federico II, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Napoli, Italy
| | - Willem A. L. van Otterlo
- Department of Chemistry and Polymer Science, University of Stellenbosch, Private Bag X1, Matieland 7602, South Africa
| | - Ramesh Dasari
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, Texas 78666, USA
| | - Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme; Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Robert Kiss
- Laboratorie de Cancérologie et de Toxicologie Expérimentale, Faculté de Pharmacie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
36
|
Li J, Jiang K, Zhao F. Icariin regulates the proliferation and apoptosis of human ovarian cancer cells through microRNA-21 by targeting PTEN, RECK and Bcl-2. Oncol Rep 2015; 33:2829-36. [PMID: 25845681 DOI: 10.3892/or.2015.3891] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/05/2015] [Indexed: 11/05/2022] Open
Abstract
Icariin is the main active ingredient found in the traditional Chinese medicinal plant Epimedium, and exhibits various pharmacological effects such as enhanced immune function, anticancer activity, improved cardiovascular function and endocrine adjustment. However, the effect of icariin on ovarian cancer and the related mechanism have never been investigated. In the present study, we aimed to verify whether icariin inhibits the proliferation and increases the apoptosis of human ovarian cancer cells, and its molecular mechanism in order to establish an association and identify potential therapeutic targets. In the present study, ovarian cancer A2780 cells were treated with various concentrations of icariin, and the cell viability was evaluated by 3,3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Flow cytometry and caspase-3 colorimetric assay were performed to observe apoptotic changes in the A2780 cells. qPCR analysis was used to analyze miR-21 expression in the A2780 cells. Western blot analysis was used to assess PTEN, RECK and Bcl-2 protein expression. Transfection of microRNA-21 (miR-21) and anti-miR-21 was used to investigate expression of its target genes associated with cell proliferation and apoptosis. Icariin concomitantly suppressed cell proliferation, accelerated apoptosis and increased caspase-3 activity in the A2780 cells. In the ovarian cancer A2780 cells, icariin substantially decreased the miR-21 expression level, increased PTEN and RECK protein expression levels and decreased the Bcl-2 protein expression level. Notably, miR-21 regulated the potential anticancer effects of icariin on cell proliferation and apoptosis by targeting PTEN, RECK and Bcl-2 in the ovarian cancer A2780 cells. Our results demonstrated that icariin is an excellent candidate antitumor agent which exhibits an anticancer curative effect on ovarian cancer cells. miR-21 and its target genes may play a vital role in the molecular mechanism of the anticancer effects of icariin.
Collapse
Affiliation(s)
- Jingwei Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 100004, P.R. China
| | - Kailei Jiang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 100004, P.R. China
| | - Fujie Zhao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 100004, P.R. China
| |
Collapse
|
37
|
Tan S, Liu ZP. Natural Products as Zinc-Dependent Histone Deacetylase Inhibitors. ChemMedChem 2015; 10:441-50. [DOI: 10.1002/cmdc.201402460] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Indexed: 12/21/2022]
|
38
|
Ni X, Li L, Pan G. HDAC inhibitor-induced drug resistance involving ATP-binding cassette transporters (Review). Oncol Lett 2014; 9:515-521. [PMID: 25624882 PMCID: PMC4301560 DOI: 10.3892/ol.2014.2714] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/31/2014] [Indexed: 01/04/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors are becoming a novel and promising class of antineoplastic agents that have been used for cancer therapy in the clinic. Two HDAC inhibitors, vorinostat and romidepsin, have been approved by the Food and Drug Administration to treat T-cell lymphoma. Nevertheless, similar to common anticancer drugs, HDAC inhibitors have been found to induce multidrug resistance (MDR), which is an obstacle for the success of chemotherapy. The most common cause of MDR is considered to be the increased expression of adenosine triphosphate binding cassette (ABC) transporters. Numerous studies have identified that the upregulation of ABC transporters is often observed following treatment with HDAC inhibitors, particularly the increased expression of P-glycoprotein, which leads to drug efflux, reduces intracellular drug concentration and induces MDR. The present review summarizes the key ABC transporters involved in MDR following various HDAC inhibitor treatments in a range of cancer cell lines and also explored the potential mechanisms that result in MDR, including the effect of nuclear receptors, which are the upstream regulatory factors of ABC transporters.
Collapse
Affiliation(s)
- Xuan Ni
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Li Li
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993-0002, USA
| | - Guoyu Pan
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| |
Collapse
|
39
|
Shin BS, Yoo SD, Kim TH, Bulitta JB, Landersdorfer CB, Shin JC, Choi JH, Weon KY, Joo SH, Shin S. Quantitative determination of absorption and first-pass metabolism of apicidin, a potent histone deacetylase inhibitor. Drug Metab Dispos 2014; 42:974-82. [PMID: 24644298 DOI: 10.1124/dmd.113.056713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Apicidin, a potential oral chemotherapeutic agent, possesses potent anti-histone-deacetylase activity. After oral administration, the total bioavailability of apicidin is known to be low (14.2%-19.3%). In the present study, we evaluated the factors contributing to the low bioavailability of apicidin by means of quantitative determination of absorption fraction and first-pass metabolism after oral administration. Apicidin was given to rats by five different routes: into the femoral vein, duodenum, superior mesenteric artery, portal vein, and carotid artery. Especially, the fraction absorbed (FX) and the fraction that is not metabolized in the gut wall (FG) were separated by injection of apicidin via superior mesenteric artery, which enables bypassing the permeability barrier. The FX was 45.9% ± 9.7%, the FG was 70.9% ± 8.1% and the hepatic bioavailability (FH) was 70.6% ± 12.3%, while the pulmonary first-pass metabolism was minimal (FL = 102.8% ± 7.4%), indicating that intestinal absorption was the rate-determining step for oral absorption of apicidin. The low FX was further examined in terms of passive diffusion and transporter-mediated efflux by in vitro immobilized artificial membrane (IAM) chromatographic assay and in situ single-pass perfusion method, respectively. Although the passive diffusion potential of apicidin was high (98.01%) by the IAM assay, the in situ permeability was significantly enhanced by the presence of the P-glycoprotein (P-gp) inhibitor elacrider. These data suggest that the low bioavailability of apicidin was mainly attributed to the P-gp efflux consistent with the limited FX measured in vivo experiment.
Collapse
Affiliation(s)
- Beom Soo Shin
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si, Gyeongbuk, South Korea (B.S.S., J.C.S., J.H.C., K.Y.W., S.H.J.); School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, South Korea (S.D.Y., T.H.K.); Centre for Medicine Use and Safety, Monash University, Parkville, Australia (J.B.B., C.B.L.); Department of Pharmacy, Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk, South Korea (S.S.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li XH, Huang ML, Wang SM, Wang Q. Selective Inhibition of Bicyclic Tetrapeptide Histone Deacetylase Inhibitor on HDAC4 and K562 Leukemia Cell. Asian Pac J Cancer Prev 2013; 14:7095-100. [DOI: 10.7314/apjcp.2013.14.12.7095] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
41
|
Wang S, Li X, Wei Y, Xiu Z, Nishino N. Discovery of potent HDAC inhibitors based on chlamydocin with inhibitory effects on cell migration. ChemMedChem 2013; 9:627-37. [PMID: 24285590 DOI: 10.1002/cmdc.201300372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 11/06/2013] [Indexed: 02/05/2023]
Abstract
The histone deacetylase (HDAC) family is a promising drug target class owing to the importance of these enzymes in a variety of cellular processes. Docking studies were conducted to identify novel HDAC inhibitors. Subtle modifications in the recognition domain were introduced into a series of chlamydocin analogues, and the resulting scaffolds were combined with various zinc binding domains. Remarkably, cyclo(L-Asu(NHOH)-L-A3mc6c-L-Phe-D-Pro, compound 1 b), with a methyl group at positions 3 or 5 on the aliphatic ring, exhibited better antiproliferative effects than trichostatin A (TSA) against MCF-7 and K562 cell lines. In addition to cell-cycle arrest and apoptosis, cell migration inhibition was observed in cells treated with compound 1 b. Subsequent western blot analysis revealed that the balance between matrix metalloproteinase 2 (MMP2) and tissue inhibitors of metalloproteinase 1 (TIMP1) determines the degree of metalloproteinase activity in MCF-7 cells, thereby regulating cell migration. The improved inhibitory activity imparted by altering the hydrophobic substitution pattern at the bulky cap group is a valuable approach in the development of novel HDAC inhibitors.
Collapse
Affiliation(s)
- Shimiao Wang
- School of Life Science and Biotechnology, Dalian University of Technology, 2 Linggong Road, 116024 Dalian (China)
| | | | | | | | | |
Collapse
|
42
|
Apoptosis signal-regulating kinase 1 is involved in brain-derived neurotrophic factor (BDNF)-enhanced cell motility and matrix metalloproteinase 1 expression in human chondrosarcoma cells. Int J Mol Sci 2013; 14:15459-78. [PMID: 23892595 PMCID: PMC3759868 DOI: 10.3390/ijms140815459] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/15/2013] [Accepted: 07/15/2013] [Indexed: 12/11/2022] Open
Abstract
Chondrosarcoma is the primary malignancy of bone that is characterized by a potent capacity to invade locally and cause distant metastasis, and is therefore associated with poor prognoses. Chondrosarcoma further shows a predilection for metastasis to the lungs. The brain-derived neurotrophic factor (BDNF) is a small molecule in the neurotrophin family of growth factors that is associated with the disease status and outcome of cancers. However, the effect of BDNF on cell motility in human chondrosarcoma cells is mostly unknown. Here, we found that human chondrosarcoma cell lines had significantly higher cell motility and BDNF expression compared to normal chondrocytes. We also found that BDNF increased cell motility and expression of matrix metalloproteinase-1 (MMP-1) in human chondrosarcoma cells. BDNF-mediated cell motility and MMP-1 up-regulation were attenuated by Trk inhibitor (K252a), ASK1 inhibitor (thioredoxin), JNK inhibitor (SP600125), and p38 inhibitor (SB203580). Furthermore, BDNF also promoted Sp1 activation. Our results indicate that BDNF enhances the migration and invasion activity of chondrosarcoma cells by increasing MMP-1 expression through a signal transduction pathway that involves the TrkB receptor, ASK1, JNK/p38, and Sp1. BDNF thus represents a promising new target for treating chondrosarcoma metastasis.
Collapse
|
43
|
Wang HG, Huang XD, Shen P, Li LR, Xue HT, Ji GZ. Anticancer effects of sodium butyrate on hepatocellular carcinoma cells in vitro. Int J Mol Med 2013; 31:967-74. [PMID: 23440283 DOI: 10.3892/ijmm.2013.1285] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/28/2013] [Indexed: 11/05/2022] Open
Abstract
In the present study, we investigated the anticancer effects of sodium butyrate (NaBu) on hepatocellular carcinoma (HCC) cells in vitro. As a histone deacetylase (HDAC) inhibitor, NaBu upregulated Ac-H3 and inhibited HDAC4 protein expression in a time- and dose-dependent manner. MTT assays showed that treatment with NaBu at high concentrations significantly inhibited the growth of various HCC cells. Exposure to NaBu for 24 h induced cell cycle arrest in the SMMC-7721 and HepG2 cells. NaBu also induced the apoptosis of SMMC‑7721 cells. The expression levels of cell cycle- and apoptosis-related proteins were further investigated by western blot analysis using specific antibodies. The results provided a possible mechanism responsible for the inhibitory effects of NaBu on the growth of HCC cells. To further analyze the role of NaBu in cell migration, wound healing and Transwell assays were performed, indicating that NaBu significantly inhibits cell migration/invasion in HCC cells. Transforming growth factor-β1 (TGF-β1)-induced epithelial to mesenchymal transition (EMT) has been associated with tumor cell migration and invasion. The EMT markers, E-cadherin, vimentin and N-cadherin, were regulated by TGF-β1, while NaBu inhibited this process in which HDAC4 and matrix metalloproteinase (MMP)7 may be involved. Based on our findings, we propose that NaBu may be useful as an anticancer drug for HCC therapy.
Collapse
Affiliation(s)
- Hong-Gang Wang
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, PR China
| | | | | | | | | | | |
Collapse
|