1
|
Khandan M, Khazeei Tabari MA, Rahimi SM, Hassani M, Bagheri A. The effects of flavonoid baicalein on miRNA expressions in cancer: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04078-y. [PMID: 40153015 DOI: 10.1007/s00210-025-04078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Baicalein from Scutellaria baicalensis influences miRNA expression in various cancers, affecting key signaling pathways (PI3K/AKT, Wnt/β-catenin, mTOR) and processes like tumor growth, apoptosis, and metastasis. miRNAs, as small non-coding RNAs, play crucial roles in the cancer pathogenesis-associated gene regulations. This study is aimed at systematically reviewing the effects of baicalein on miRNA expression in various cancers. A comprehensive systematic review was conducted following PRISMA guidelines to investigate the impact of baicalein on miRNA expression in cancer. Databases including PubMed, Scopus, and Web of Science were systematically searched using key search terms. Inclusion criteria encompassed studies reporting changes in miRNA expression following baicalein treatment in cancer cell lines and animal models. Data extraction and risk of bias assessment based on SYRCLE's risk of bias tool were performed to ensure methodological rigor and reliability of the findings. Fifteen studies meeting the inclusion criteria were included in the systematic review. Baicalein impacts miRNA expression in cancers like hepatocellular carcinoma, breast, cervical, ovarian, and gastric cancers, suggesting its potential as a multi-cancer therapeutic. Baicalein regulates tumor-related genes (HDAC10, MDM2, Bcl-2/Bax, and Cyclin E1) and signaling molecules (AKT, FOXO3α), affecting cell viability, apoptosis, and cell cycle, indicating targeted therapeutic potential. In vitro and in vivo studies show baicalein inhibits tumor growth, enhances apoptosis, and regulates cell proliferation, supporting its anticancer effects. Baicalein exhibits potential in modulating miRNA expression in cancer, offering avenues for therapeutic intervention. However, methodological rigor in future studies is essential to enhance the reliability and validity of findings. Comprehensive understanding of baicalein's effects on miRNA expression holds promise for developing novel cancer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Mahmoud Hassani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abouzar Bagheri
- Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
- Immunogenetics Research Center, Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Ji J, Xiong C, Yang H, Jiang Z, Zhang Y, Wang X, Yu T, Li Q, Zhu S, Zhou Y. The aryl hydrocarbon receptor: A crucial mediator in ocular disease pathogenesis and therapeutic target. Exp Eye Res 2024; 249:110144. [PMID: 39486499 DOI: 10.1016/j.exer.2024.110144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
The aryl hydrocarbon receptor (AHR) is a pivotal nuclear receptor involved in mediating cellular responses to a wide range of environmental pollutants and endogenous ligands. AHR plays a central role in regulating essential physiological processes, including xenobiotic metabolism, immune response modulation, cell cycle control, tumorigenesis, and developmental events. Recent studies have identified AHR as a critical mediator and a potential therapeutic target in the pathogenesis of ocular diseases. This review provides a thorough analysis of the various functions of AHR signalling in the ocular environment, with a specific emphasis on its effects on the retina, retinal pigment epithelium (RPE), choroid, and cornea. We provide a detailed discussion on the molecular mechanisms through which AHR integrates environmental and endogenous signals, influencing the development and progression of age-related macular degeneration (AMD), retinitis pigmentosa, uveitis, and other major ocular disorders. Furthermore, we evaluate the therapeutic potential of modulating AHR activity through novel ligands and agonists as a strategy for treating eye diseases. Understanding the molecular mechanisms of AHR in ocular tissues may facilitate the development of AHR-targeted therapies, which is crucial for addressing the pressing clinical demand for novel treatment strategies in ocular diseases.
Collapse
Affiliation(s)
- Juanjuan Ji
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chanyu Xiong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Huining Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhilin Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yun Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tianshu Yu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiong Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shikai Zhu
- Organ Transplant Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhou
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Genome Sequencing Center, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
3
|
Opitz CA, Holfelder P, Prentzell MT, Trump S. The complex biology of aryl hydrocarbon receptor activation in cancer and beyond. Biochem Pharmacol 2023; 216:115798. [PMID: 37696456 PMCID: PMC10570930 DOI: 10.1016/j.bcp.2023.115798] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
The aryl hydrocarbon receptor (AHR) signaling pathway is a complex regulatory network that plays a critical role in various biological processes, including cellular metabolism, development, and immune responses. The complexity of AHR signaling arises from multiple factors, including the diverse ligands that activate the receptor, the expression level of AHR itself, and its interaction with the AHR nuclear translocator (ARNT). Additionally, the AHR crosstalks with the AHR repressor (AHRR) or other transcription factors and signaling pathways and it can also mediate non-genomic effects. Finally, posttranslational modifications of the AHR and its interaction partners, epigenetic regulation of AHR and its target genes, as well as AHR-mediated induction of enzymes that degrade AHR-activating ligands may contribute to the context-specificity of AHR activation. Understanding the complexity of AHR signaling is crucial for deciphering its physiological and pathological roles and developing therapeutic strategies targeting this pathway. Ongoing research continues to unravel the intricacies of AHR signaling, shedding light on the regulatory mechanisms controlling its diverse functions.
Collapse
Affiliation(s)
- Christiane A Opitz
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Neurology Clinic and National Center for Tumor Diseases, 69120 Heidelberg, Germany.
| | - Pauline Holfelder
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Mirja Tamara Prentzell
- German Cancer Research Center (DKFZ), Heidelberg, Division of Metabolic Crosstalk in Cancer and the German Cancer Consortium (DKTK), DKFZ Core Center Heidelberg, 69120 Heidelberg, Germany; Faculty of Bioscience, Heidelberg University, 69120 Heidelberg, Germany
| | - Saskia Trump
- Molecular Epidemiology Unit, Berlin Institute of Health at Charité and the German Cancer Consortium (DKTK), Partner Site Berlin, a partnership between DKFZ and Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
4
|
Salama BM, Helmy MW, Fouad H, Shamaa MM, Houssen ME. The Synergistic Antitumor Effect of Decitabine and Vorinostat Combination on HepG2 Human Hepatocellular Carcinoma Cell Line via Epigenetic Modulation of Autophagy-Apoptosis Molecular Crosstalk. Curr Issues Mol Biol 2023; 45:5935-5949. [PMID: 37504291 PMCID: PMC10378248 DOI: 10.3390/cimb45070375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a worldwide health issue. Epigenetic alterations play a crucial role in HCC tumorigenesis. Using epigenetic modulators for HCC treatment confers a promising therapeutic effect. The aim of this study was to explore the effect of a decitabine (DAC) and vorinostat (VOR) combination on the crosstalk between apoptosis and autophagy in the HCC HepG2 cell line at 24 h and 72 h. Median inhibitory concentrations (IC50s) of VOR and DAC were assessed in the HepG2 cell line. The activity of caspase-3 was evaluated colorimetrically, and Cyclin D1(CCND1), Bcl-2, ATG5, ATG7, and P62 levels were assessed using ELISA at different time intervals (24 h and 72 h), while LC3IIB and Beclin-1gene expression were measured by using qRT-PCR. The synergistic effect of VOR and DAC was confirmed due to the observed combination indices (CIs) and dose reduction indices (DRIs). The combined treatment with both drugs inhibited the proliferation marker (CCND1), and enhanced apoptosis compared with each drug alone at 24 h and 72 h (via active caspase-3 upregulation and Bcl-2 downregulation). Moreover, the combination induced autophagy as an early event via upregulation of Beclin-1, LC3IIB, ATG5, and ATG7 gene expression. The initial induction of autophagy started to decrease after 72 h due to Beclin-1 downregulation, and there was decreased expression of LC3IIB compared with the value at 24 h. Herein, epigenetic modulation via the VOR/DAC combination showed an antitumor effect through the coordination of an autophagy-apoptosis crosstalk and promotion of autophagy-induced apoptosis, which ultimately led to the cellular death of HCC cancer cells.
Collapse
Affiliation(s)
- Basant M Salama
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Maged W Helmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
- Pharmacology and Toxicology Department, Clinical and Biological Sciences Division, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria 1029, Egypt
| | - Hosny Fouad
- Pharmacology Department, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Biochemistry, Clinical and Biological Science Division, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria 1029, Egypt
| | - Marium M Shamaa
- Department of Biochemistry, Clinical and Biological Science Division, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria 1029, Egypt
| | - Maha E Houssen
- Department of Biochemistry, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| |
Collapse
|
5
|
Suzuki T, Aoshima K, Yamazaki J, Kobayashi A, Kimura T. Manipulating Histone Acetylation Leads to Antitumor Effects in Hemangiosarcoma Cells. Vet Comp Oncol 2022; 20:805-816. [PMID: 35568976 DOI: 10.1111/vco.12840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/26/2022]
Abstract
Canine hemangiosarcoma (HSA) is a malignant tumor derived from endothelial cells. No effective treatment has yet been developed because of the lack of understanding of its pathogenesis. Histone acetylation, an epigenetic modification, is highly associated with cancer pathogenesis. Manipulating histone acetylation by histone deacetylase inhibitors (HDACi) or bromodomain and extraterminal domain inhibitors (BETi) is one approach to treat various cancers. However, the role of histone acetylation in HSA remains unknown. This study aimed to investigate how histone acetylation functions in HSA pathogenesis using two HDACi, suberanilohydroxamic acid (SAHA) and valproic acid (VPA), and one BETi, JQ1, in vitro and in vivo. Histone acetylation levels were high in cell lines and heterogeneous in clinical cases. SAHA and JQ1 induced apoptosis in HSA cell lines. HSA cell lines treated with SAHA and VPA upregulated inflammatory-related genes and attracted macrophage cell line RAW264 cells, which suggests that SAHA and VPA can affect immune responses. JQ1 stimulated autophagy and inhibited the cell cycle in HSA cell lines. Finally, we demonstrated that JQ1 suppressed HSA tumor cell proliferation in vivo although SAHA and VPA did not affect tumor growth. These results suggest that BETi can be alternative drugs for HSA treatment. Although further research is required, our study indicated that dysregulation of histone acetylation is likely to be involved in HSA malignancy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tamami Suzuki
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Keisuke Aoshima
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Jumpei Yamazaki
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Atsushi Kobayashi
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takashi Kimura
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
6
|
Zhu M, Zhang P, Yu S, Tang C, Wang Y, Shen Z, Chen W, Liu T, Cui Y. Targeting ZFP64/GAL-1 axis promotes therapeutic effect of nab-paclitaxel and reverses immunosuppressive microenvironment in gastric cancer. J Exp Clin Cancer Res 2022; 41:14. [PMID: 34996504 PMCID: PMC8740411 DOI: 10.1186/s13046-021-02224-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/13/2021] [Indexed: 01/03/2023] Open
Abstract
Background Chemoresistance is a main obstacle in gastric cancer (GC) treatment, but its molecular mechanism still needs to be elucidated. Here, we aim to reveal the underlying mechanisms of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) resistance in GC. Methods We performed RNA sequencing (RNA-seq) on samples from patients who were resistant or sensitive to nab-paclitaxel, and identified Zinc Finger Protein 64 (ZFP64) as critical for nab-paclitaxel resistance in GC. CCK8, flow cytometry, TUNEL staining, sphere formation assays were performed to investigate the effects of ZFP64 in vitro, while subcutaneous tumor formation models were established in nude mice or humanized mice to evaluate the biological roles of ZFP64 in vivo. Chromatin immunoprecipitation sequencing (CHIP-seq) and double-luciferase reporter gene assay were conducted to reveal the underlying mechanism of ZFP64. Results ZFP64 overexpression was linked with aggressive phenotypes, nab-paclitaxel resistance and served as an independent prognostic factor in GC. As a transcription factor, ZFP64 directly binds to Galectin-1 (GAL-1) promoter and promoted GAL-1 transcription, thus inducing stem-cell like phenotypes and immunosuppressive microenvironment in GC. Importantly, compared to treatment with nab-paclitaxel alone, nab-paclitaxel plus GAL-1 blockade significantly enhanced the anti-tumor effect in mouse models, particularly in humanized mice. Conclusions Our data support a pivotal role for ZFP64 in GC progression by simultaneously promoting cellular chemotherapy resistance and tumor immunosuppression. Treatment with the combination of nab-paclitaxel and a GAL-1 inhibitor might benefit a subgroup of GC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02224-x.
Collapse
Affiliation(s)
- Mengxuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Center of Evidence-based Medicine, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Tang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weidong Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Evidence-based Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Lam KK, Tang CL, Tan E, Wong SH, Cheah PY. KRAS mutation-independent downregulation of MAPK/PI3K signaling in colorectal cancer. Mol Oncol 2021; 16:1171-1183. [PMID: 34919787 PMCID: PMC8895447 DOI: 10.1002/1878-0261.13163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022] Open
Abstract
KRAS is a gatekeeper gene in human colorectal tumorigenesis. KRAS is ‘undruggable’; hence, efforts have been diverted to inhibit downstream RAF/MEK/ERK and PI3K/Akt signaling. Nevertheless, none of these inhibitors has progressed to clinical use despite extensive trials. We examined levels of phospho‐ERK1/2(T202/Y204) and phospho‐Akt1/2/3(S473) in human colorectal tumor compared to matched mucosa with semi‐quantitative near‐infrared western blot and confocal fluorescence immunohistochemistry imaging. Surprisingly, 75.5% (25/33) of tumors had lower or equivalent phospho‐ERK1/2 and 96.9% (31/32) of tumors had lower phospho‐Akt1/2/3 compared to matched mucosa, irrespective of KRAS mutation status. In contrast, we discovered KRAS‐dependent SOX9 upregulation in 28 of the 31 (90.3%) tumors. These observations were substantiated by analysis of the public domain transcriptomics The Cancer Genome Atlas (TCGA) and NCBI Gene Expression Omnibus (GEO) datasets and proteomics Clinical Proteomic Tumor Analysis Consortium (CPTAC) dataset. These data suggest that RAF/MEK/ERK and PI3K/Akt signaling are unlikely to be activated in most human colorectal cancer.
Collapse
Affiliation(s)
- Kuen Kuen Lam
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | - Choong Leong Tang
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | - Emile Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore
| | | | - Peh Yean Cheah
- Department of Colorectal Surgery, Singapore General Hospital, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore
| |
Collapse
|
8
|
Adam RS, Blomberg I, Ten Hoorn S, Bijlsma MF, Vermeulen L. The recurring features of molecular subtypes in distinct gastrointestinal malignancies-A systematic review. Crit Rev Oncol Hematol 2021; 164:103428. [PMID: 34284100 DOI: 10.1016/j.critrevonc.2021.103428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/26/2022] Open
Abstract
In colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC) and gastric cancer (GC) multiple studies of inter-tumor heterogeneity have identified molecular subtypes, which correlate with clinical features. Our aim was to investigate the attributes of molecular subtypes across three different gastrointestinal cancer types. We performed a systematic search for publications on molecular subtypes or classifications in PDAC and GC and compared the described subtypes with the established consensus molecular subtypes of CRC. Examining the characteristics of subtypes across CRC, PDAC and GC resulted in four categories of subtypes. We describe uniting and distinguishing features within a mesenchymal, an epithelial, an immunogenic and a metabolic and digestive subtype category. We conclude that molecular subtypes of CRC, PDAC and GC display relevant overlap in molecular features and clinical outcomes. This finding encourages quantitative studies on subtypes across different cancer types and could lead to a paradigm shift in future treatment strategies.
Collapse
Affiliation(s)
- Ronja S Adam
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Ilse Blomberg
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Sanne Ten Hoorn
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Mir IH, Guha S, Behera J, Thirunavukkarasu C. Targeting molecular signal transduction pathways in hepatocellular carcinoma and its implications for cancer therapy. Cell Biol Int 2021; 45:2161-2177. [PMID: 34270844 DOI: 10.1002/cbin.11670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/22/2021] [Accepted: 07/11/2021] [Indexed: 12/27/2022]
Abstract
Hepatocellular carcinoma is a substantial health concern. It is currently the third dominating cause of mortality associated with cancer worldwide. The development of hepatocellular carcinoma is an intricate process that encompasses the impairment of genetic, epigenetic, and signal transduction mechanisms contributing to an aberrant metabolic system, enabling tumorigenesis. Throughout the past decade, research has led to the revelation of molecular pathways implicated in the progression of this notorious disorder. The altered signal transduction pathways, such as the mitogen-activated protein kinase pathway, phosphoinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathway, WNT/β-catenin pathway, hepatocyte growth factor/c-MET pathway, and just another kinase/signal transducers and activators of transcription signaling pathway is of much therapeutic significance, as targeting them may avail to revert, retard or avert hepatocarcinogenesis. The present review article sums up the contemporary knowledge of such signaling mechanisms, including their therapeutic targets and betokens that novel and efficacious therapies can be developed only by the keen understanding of their character in hepatocarcinogenesis. In additament, we address the role of consequential therapeutic agents and preclinical nondrug therapies known for combating hepatocarcinogenesis.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Shreyoshi Guha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Jajnasenee Behera
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | |
Collapse
|
10
|
Sodium Valproate, a Histone Deacetylase Inhibitor, Provokes Reactive Oxygen Species-Mediated Cytotoxicity in Human Hepatocellular Carcinoma Cells. J Gastrointest Cancer 2021; 52:138-144. [PMID: 32006341 DOI: 10.1007/s12029-020-00370-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Sodium valproate (SV), a novel class of histone deacetylases (HDACs) inhibitors commonly used as an antiepileptic drug. HDAC inhibitors are known to possess anticancer potentials. In this study, we investigated the cytotoxic potential of SV in human hepatocellular carcinoma (HepG2 cells) cell line. METHODS MTT assay was used to analyze cytotoxicity. Intracellular ROS and cytochrome c expression were analyzed by fluorescence microscopy. Morphology-related apoptosis was analyzed by dual staining with acridine orange/ethidium bromide. Caspase 3 protein expression was investigated by Western blotting analysis. RESULTS Sodium valproate treatments in HepG2 cells caused significant and dose-dependent cytotoxicity. Intracellular ROS was remarkably increased in the cells which are treated with SV and caused early and late apoptosis as evidenced by dual staining. SV-treated cells expressed cytochrome c and caspase 3 protein expression. CONCLUSION These results suggest the cytotoxic potentials of SV in HepG2 cells. This study may give an important clue for the inclusion of SV as an adjuvant along with standard anticancer agents after necessary in vivo and clinical studies.
Collapse
|
11
|
Koufaris C, Kirmizis A. Identification of NAA40 as a Potential Prognostic Marker for Aggressive Liver Cancer Subtypes. Front Oncol 2021; 11:691950. [PMID: 34150665 PMCID: PMC8208081 DOI: 10.3389/fonc.2021.691950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/17/2021] [Indexed: 11/30/2022] Open
Abstract
Liver hepatocellular carcinoma (LIHC) is a leading cause of cancer-related mortality. In this study we initially interrogated the Cancer Genome Atlas (TCGA) dataset to determine the implication of N-terminal acetyltransferases (NATs), a family of enzymes that modify the N-terminus of the majority of eukaryotic proteins, in LIHC. This examination unveiled NAA40 as the NAT family member with the most prominent upregulation and significant disease prognosis for this cancer. Focusing on this enzyme, which selectively targets histone proteins, we show that its upregulation occurs from early stages of LIHC and is not specifically correlated with any established risk factors such as viral infection, obesity or alcoholic disease. Notably, in silico analysis of TCGA and other LIHC datasets found that expression of this epigenetic enzyme is associated with high proliferating, poorly differentiating and more aggressive LIHC subtypes. In particular, NAA40 upregulation was preferentially linked to mutational or non-mutational P53 functional inactivation. Accordingly, we observed that high NAA40 expression was associated with worse survival specifically in liver cancer patients with inactivated P53. These findings define NAA40 as a NAT with potentially oncogenic functions in LIHC and uncover its prognostic value for aggressive LIHC subtypes.
Collapse
|
12
|
Sanaei M, Kavoosi F. Effect of vorinostat on INK4 family and HDACs 1, 2, and 3 in pancreatic cancer and hepatocellular carcinoma. Res Pharm Sci 2021; 16:260-268. [PMID: 34221059 PMCID: PMC8216159 DOI: 10.4103/1735-5362.314824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/11/2020] [Accepted: 03/13/2021] [Indexed: 11/18/2022] Open
Abstract
Background and purpose: In mammalian cells, several distinct surveillance systems, named cell cycle checkpoints, can interrupt normal cell-cycle progression. The cyclin-dependent kinases are negatively regulated by proteins of cyclin-dependent kinases inhibitors comprising INK4 and Cip/Kip families. Histone deacetylation induced by histone deacetylases (HDACs) inactivates the INK4 and Cip/Kip families lead to cancer induction. HDAC inhibitors (HDACIs) have been indicated to be potent inducers of differentiation, growth arrest, and apoptotic induction. Vorinostat (suberoylanilide hydroxamic acid, SAHA), as an HDACI, is reported to be useful in various cancers. Previously, we reported the effect of trichostatin A on hepatocellular carcinoma and also vorinostat on colon cancer cell lines. The current study was aimed to investigate the effect of vorinostat on p16INK4a, p14ARF, p15INK4b, and class I HDACs 1, 2, and 3 gene expression, cell growth inhibition, and apoptosis induction in pancreatic cancer AsPC-1 and hepatocellular carcinoma LCL-PI 11 cell lines. Experimental approach: The AsPC-1 and LCL-PI 11 cell lines were cultured and treated with vorinostat. To determine, viability, apoptosis, and the relative expression level of p16INK4a, p14ARF, p15INK4b, class I HDACs 1, 2, and 3 genes, MTT assay, cell apoptosis assay, and RT-qPCR were performed, respectively. Findings/Results: Vorinostat significantly inhibited cell growth, induced apoptosis, increased p16INK4a, p14ARF, p15INK4b, and decreased class I HDACs 1, 2, and 3 gene expression. Conclusion and implications: Vorinostat can reactivate the INK4 family through inhibition of class I HDACs 1, 2, and 3 genes activity.
Collapse
Affiliation(s)
- Masumeh Sanaei
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, I.R. Iran
| | - Fraidoon Kavoosi
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, I.R. Iran
| |
Collapse
|
13
|
Hsu WC, Ramesh S, Shibu MA, Chen MC, Wang TF, Day CH, Chen RJ, Padma VV, Li CC, Tseng YC, Huang CY. Platycodin D reverses histone deacetylase inhibitor resistance in hepatocellular carcinoma cells by repressing ERK1/2-mediated cofilin-1 phosphorylation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153442. [PMID: 33412494 DOI: 10.1016/j.phymed.2020.153442] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/22/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Chemoresistance remains the main obstacle in hepatocellular carcinoma (HCC) therapy. Despite significant advances in HCC therapy, HCC still has a poor prognosis. Thus, there is an urgent need to identify a treatment target to reverse HCC chemotherapy resistance. Platycodon grandiflorus (PG) is a perennial herb that has been used as food and traditional Chinese medicine for thousands of years in Northeast Asia. Platycodin D (PD), a main active triterpenoid saponin found in the root of PG, has been reported to possess anticancer properties in several cancer cell lines, including HCC; however, the reversal effect of this molecule on HCC chemoresistance remains largely unknown. PURPOSE This study aimed to investigate the role and the mechanism of PD-mediated reversal of the histone deacetylase inhibitor (HDACi) resistance in HCC cells. METHODS Human HCC cells (HA22T) and HDACi-resistant (HDACi-R) cells were used. Cell viability was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Combination index was used to calculate the synergism potential. Expression of ERK1/2 (total/phospho), cofilin-1 (total/phospho) and apoptosis-related protein was determined using western blotting. Mitochondrial membrane potential was assessed using the JC-1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolocarbocyanine iodide) probe. Apoptosis was detected using the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Mitochondrial reactive oxygen species generation was measured using the MitoSOX Red fluorescent probe. RESULTS We found that PD treatment inhibited cell viability both in HA22T HCC and HDACi-R cells. Inhibition of ERK1/2 by PD98059 could reverse drug resistance in HDACi-R cells treated with PD98059 and PD. Nevertheless, pre-treatment with U46619, an ERK1/2 activator, rescued PD-induced apoptosis by decreasing levels of apoptosis-related proteins in HCC cells. The combined treatment of PD with apicidin a powerful HDACi, dramatically enhanced the apoptotic effect in HDACi-R cells. CONCLUSION For the first time, we showed that PD reversed HDACi resistance in HCC by repressing ERK1/2-mediated cofilin-1 phosphorylation. Thus, PD can potentially be a treatment target to reverse HCC chemotherapy resistance in future therapeutic trials.
Collapse
Affiliation(s)
- Wei-Chung Hsu
- Department of Radiation Oncology, Chung-Kang Branch, Cheng-Ching General Hospital, Taichung 40764, Taiwan; Department of Occupational Therapy, Asia University, Taichung 41354, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; Department of Microbiology, PRIST Deemed to be University, Thanjavur 614 904, Tamil Nadu, India
| | - Marthandam Asokan Shibu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Ming-Cheng Chen
- Department of Surgery, Division of Colorectal Surgery, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; School of Medicine, Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan
| | | | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - V Vijaya Padma
- Department of Biotechnology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Chi-Cheng Li
- School of Medicine, Tzu Chi University, 701, Section 3, Chung-Yang Road, Hualien 97004, Taiwan; Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Yu-Chen Tseng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taichung Armed Forces General Hospital, Taichung, Taiwan; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; Center of Stem Cell & Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; Department of Biological Science and Technology, Asia University, Taichung, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
14
|
Imamura Y, Watanabe M, Oki E, Morita M, Baba H. Esophagogastric junction adenocarcinoma shares characteristics with gastric adenocarcinoma: Literature review and retrospective multicenter cohort study. Ann Gastroenterol Surg 2021; 5:46-59. [PMID: 33532680 PMCID: PMC7832959 DOI: 10.1002/ags3.12406] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/23/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of esophagogastric junction (EGJ) adenocarcinoma has been gradually increasing in Asia, just like in Western countries a few decades ago. Despite recent advances in next-generation sequencing and multimodal treatments, EGJ adenocarcinoma is still an aggressive malignancy with poor outcomes. Clinically, EGJ adenocarcinoma can be separated into Barrett's adenocarcinoma and cardiac adenocarcinoma, with frequent similarities observed. Barrett's adenocarcinoma is likely to be of gastric origin in terms of its premalignant background, risk factors, and stem cell regulators. Recent comprehensive genomic analyses suggest that immunotherapy may be essential for high-level microsatellite instability (MSI-H)- and Epstein-Barr virus (EBV)-associated subtypes, and against the immunosuppressive phenotype in genomically stable (GS) subtypes, in the treatment of EGJ and gastric adenocarcinoma. Although the chromosomal instability (CIN) subtype dominates EGJ adenocarcinoma, there is still a need to investigate the other molecular subtypes and their targets. Because of the distinctive characteristics of tumor location of EGJ adenocarcinoma, we also described the results of a multicenter cohort study of EGJ adenocarcinoma, comparing Siewert type I (distal esophagus), II (cardia of the stomach), and III (subcardia) tumors. We show that type I tumors were frequently accompanied by Barrett's esophagus (78%, P < .0001), with a significantly unfavorable outcome (multivariate EGJ-cancer-specific mortality hazard ratio = 1.81, 95% CI, 1.06-2.97; P = .031). In addition, over half (56%) of these cases experienced disease recurrence in the lymph nodes. Our findings suggest that Barrett's adenocarcinoma may be an aggressive phenotype of EGJ adenocarcinoma due to the potential risk of tumor spread through the complex lympho-vascular network of the esophagus.
Collapse
Affiliation(s)
- Yu Imamura
- Department of Gastroenterological SurgeryCancer Institute Hospital of Japanese Foundation of Cancer ResearchTokyoJapan
| | - Masayuki Watanabe
- Department of Gastroenterological SurgeryCancer Institute Hospital of Japanese Foundation of Cancer ResearchTokyoJapan
| | - Eiji Oki
- Department of Surgery and ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Masaru Morita
- Department of Gastroenterological SurgeryKyushu Cancer CenterNational Hospital OrganizationFukuokaJapan
| | - Hideo Baba
- Department of Gastroenterological SurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
15
|
Choudhary M, Malek G. The Aryl Hydrocarbon Receptor: A Mediator and Potential Therapeutic Target for Ocular and Non-Ocular Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21186777. [PMID: 32947781 PMCID: PMC7555571 DOI: 10.3390/ijms21186777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, which senses environmental, dietary or metabolic signals to mount a transcriptional response, vital in health and disease. As environmental stimuli and metabolic products have been shown to impact the central nervous system (CNS), a burgeoning area of research has been on the role of the AHR in ocular and non-ocular neurodegenerative diseases. Herein, we summarize our current knowledge, of AHR-controlled cellular processes and their impact on regulating pathobiology of select ocular and neurodegenerative diseases. We catalogue animal models generated to study the role of the AHR in tissue homeostasis and disease pathogenesis. Finally, we discuss the potential of targeting the AHR pathway as a therapeutic strategy, in the context of the maladies of the eye and brain.
Collapse
Affiliation(s)
- Mayur Choudhary
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| | - Goldis Malek
- Department of Ophthalmology, Duke University School of Medicine, 2351 Erwin Road, P.O. Box 3802, Durham, NC 27705, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27705, USA
- Correspondence: (M.C.); (G.M.)
| |
Collapse
|
16
|
Akhtar S, Hourani S, Therachiyil L, Al-Dhfyan A, Agouni A, Zeidan A, Uddin S, Korashy HM. Epigenetic Regulation of Cancer Stem Cells by the Aryl Hydrocarbon Receptor Pathway. Semin Cancer Biol 2020; 83:177-196. [PMID: 32877761 DOI: 10.1016/j.semcancer.2020.08.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022]
Abstract
Compelling evidence has demonstrated that tumor bulk comprises distinctive subset of cells generally referred as cancer stem cells (CSCs) that have been proposed as a strong sustainer and promoter of tumorigenesis and therapeutic resistance. These distinguished properties of CSCs have raised interest in understanding the molecular mechanisms that govern the maintenance of these cells. Numerous experimental and epidemiological studies have demonstrated that exposure to environmental toxins such as the polycyclic aromatic hydrocarbons (PAHs) is strongly involved in cancer initiation and progression. The PAH-induced carcinogenesis is shown to be mediated through the activation of a cytosolic receptor, aryl hydrocarbon receptor (AhR)/Cytochrome P4501A pathway, suggesting a possible direct link between AhR and CSCs. Several recent studies have investigated the role of AhR in CSCs self-renewal and maintenance, however the molecular mechanisms and particularly the epigenetic regulations of CSCs by the AhR/CYP1A pathway have not been reviewed before. In this review, we first summarize the crosstalk between AhR and cancer genetics, with a particular emphasis on the mechanisms relevant to CSCs such as Wnt/β-catenin, Notch, NF-κB, and PTEN-PI3K/Akt signaling pathways. The second part of this review discusses the recent advances and studies highlighting the epigenetic mechanisms mediated by the AhR/CYP1A pathway that control CSC gene expression, self-renewal, and chemoresistance in various human cancers. Furthermore, the review also sheds light on the importance of targeting the epigenetic pathways as a novel therapeutic approach against CSCs.
Collapse
Affiliation(s)
- Sabah Akhtar
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Shireen Hourani
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Lubna Therachiyil
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Abdullah Al-Dhfyan
- Stem Cell & Tissue Re-Engineering, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Asad Zeidan
- Department of Biomedical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Hesham M Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
17
|
Xiao Q, Liu H, Wang HS, Cao MT, Meng XJ, Xiang YL, Zhang YQ, Shu F, Zhang QG, Shan H, Jiang GM. Histone deacetylase inhibitors promote epithelial-mesenchymal transition in Hepatocellular Carcinoma via AMPK-FOXO1-ULK1 signaling axis-mediated autophagy. Am J Cancer Res 2020; 10:10245-10261. [PMID: 32929346 PMCID: PMC7481427 DOI: 10.7150/thno.47045] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most frequent cause of cancer-related deaths globally because of high metastasis and recurrence rates. Elucidating the molecular mechanisms of HCC recurrence and metastasis and developing effective targeted therapies are expected to improve patient survival. The promising anti-cancer agents for the treatment of hematological malignancies, histone deacetylase inhibitors (HDIs), have limited effects against epithelial cell-derived cancers, including HCC, the mechanisms involved have not been elucidated. Herein, we studied the molecular mechanisms underlying HDI-induced epithelial-mesenchymal transition (EMT) involving FOXO1-mediated autophagy. Methods: The biological functions of HDIs in combination with autophagy inhibitors were examined both in vitro and in vivo. Cell autophagy was assessed using the generation of mRFP-GFP-LC3-expressing cells and fluorescent LC3 puncta analysis, Western blotting, and electron microscopy. An orthotopic hepatoma model was established in mice for the in vivo experiments. Results: Our study provided novel mechanistic insights into HDI-induced EMT mediated by the autophagy AMPK-FOXO1-ULK1-Snail signaling axis. We demonstrated that autophagy served as a pro-metastasis mechanism in HDI-treated hepatoma cells. HDIs induced autophagy via a FOXO1-dependent pathway, and FOXO1 inhibition promoted HDI-mediated apoptosis in hepatoma cells. Thus, our findings provided novel insights into the molecular mechanisms underlying HDI-induced EMT involving FOXO1-mediated autophagy and demonstrated that a FOXO1 inhibitor exerted a synergistic effect with an HDI to inhibit cell growth and metastasis in vitro and in vivo. Conclusion: We demonstrated that HDIs triggers FOXO1-dependent autophagy, which ultimately promotes EMT, limiting the clinical outcome of HDI-based therapies. Our study suggests that the combination of an HDI and a FOXO1 inhibitor is an effective therapeutic strategy for the treatment of HCC.
Collapse
|
18
|
ElHady AK, Shih SP, Chen YC, Liu YC, Ahmed NS, Keeton AB, Piazza GA, Engel M, Abadi AH, Abdel-Halim M. Extending the use of tadalafil scaffold: Development of novel selective phosphodiesterase 5 inhibitors and histone deacetylase inhibitors. Bioorg Chem 2020; 98:103742. [PMID: 32199305 DOI: 10.1016/j.bioorg.2020.103742] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/27/2020] [Accepted: 03/07/2020] [Indexed: 12/17/2022]
Abstract
Herein we present the synthesis and characterization of a novel chemical series of tadalafil analogues that display different pharmacological profiles. Compounds that have the 6R, 12aR configuration and terminal carboxylic acid group at the side chain arising from the piperazinedione nitrogen were potent PDE5 inhibitors, with compound 11 having almost equal potency to tadalafil and superior selectivity over PDE11, the most common off-target for tadalafil. Modifying the stereochemistry into 6S, 12aS configuration and adopting the hydroxamic acid moiety as a terminal group gave rise to compounds that only inhibited HDAC. Dual PDE5/HDAC inhibition could be achieved with compounds having 6R, 12aR configuration and hydroxamic acid moiety as a terminal group. The anticancer activity of the synthesized compounds was evaluated against a diverse number of cell lines of different origin. The compounds elicited anticancer activity against cell lines belonging to lymphoproliferative cancer as well as solid tumors. Despite the previous reports suggesting anticancer activity of PDE5 inhibitors, the growth inhibitory activity of the compounds seemed to be solely dependent on HDAC inhibition. Compound 26 (pan HDAC IC50 = 14 nM, PDE5 IC50 = 46 nM) displayed the most potent anticancer activity in the present series and was shown to induce apoptosis in Molt-4 cells. HDAC isoform selectivity testing for compound 26 showed that it is more selective for HDAC6 and 8 over HDAC1 by more than 20-fold.
Collapse
Affiliation(s)
- Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Shou-Ping Shih
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 804, Taiwan; Doctoral Degree Program in Marine Biotechnology, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115, Taiwan
| | - Yu-Cheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 40402, Taiwan
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Nermin S Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Adam B Keeton
- Department of Oncologic Sciences and Pharmacology, Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36608, USA
| | - Gary A Piazza
- Department of Oncologic Sciences and Pharmacology, Drug Discovery Research Center, Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36608, USA
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University, Campus C2.3, D-66123 Saarbrücken, Germany
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt.
| |
Collapse
|
19
|
Li Y, Li Z, Jia Y, Ding B, Yu J. In Vitro Anti-hepatoma Activities of Notoginsenoside R1 Through Downregulation of Tumor Promoter miR-21. Dig Dis Sci 2020; 65:1364-1375. [PMID: 31559550 DOI: 10.1007/s10620-019-05856-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Notoginsenoside R1 (NG-R1) is the predominant active ingredient and a novel triterpene saponin compound extracted from the roots of Panax notoginseng. To date, to the best of our knowledge, there are no previous studies concerning the effect of NG-R1 on hepatocellular carcinoma (HCC). AIMS To investigate the effects of NG-R1 on HCC cell growth, apoptosis, and invasion and to explore the underlying mechanisms. METHODS Cell viability and lactate dehydrogenase (LDH) release were evaluated by cell counting kit-8 and LDH assay, respectively. Apoptosis was assessed using flow cytometry analysis and caspase-3/7 activity assay. Cell invasion was detected by Transwell invasion assay and western blot analysis of matrix metallopeptidase (MMP)-2 and MMP-9. The effects of NG-R1 on miR-21 expression and phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway were examined by qRT-PCR and western blot, respectively. RESULTS NG-R1 inhibited the viability, increased LDH release and caspase-3/7 activity, induced apoptosis, and suppressed invasion in HCC cells. NG-R1 reduced miR-21 expression in HCC cells. miR-21 overexpression significantly attenuated the effects of NG-R1 on the viability, LDH release, apoptosis, caspase-3/7 activity, and invasion of HCC cells. We further demonstrated that NG-R1 inhibited the activation of the PI3K/Akt pathway in HCC cells, which was abolished by miR-21 overexpression. CONCLUSIONS NG-R1 exerted anti-hepatoma activity through inactivation of the PI3K/Akt pathway by downregulating miR-21, contributing to further understanding of the anti-tumor activities of NG-R1 in HCC.
Collapse
Affiliation(s)
- Yuan Li
- Department of General Surgery, Nanyang First People's Hospital, No. 12 Renmin Road, Nanyang, 473012, China
| | - Zhong Li
- Department of General Surgery, Nanyang First People's Hospital, No. 12 Renmin Road, Nanyang, 473012, China
| | - Yunhao Jia
- Department of General Surgery, Nanyang First People's Hospital, No. 12 Renmin Road, Nanyang, 473012, China
| | - Bo Ding
- Department of General Surgery, Nanyang First People's Hospital, No. 12 Renmin Road, Nanyang, 473012, China
| | - Jinsong Yu
- Department of General Surgery, Nanyang First People's Hospital, No. 12 Renmin Road, Nanyang, 473012, China.
| |
Collapse
|
20
|
A short guide to histone deacetylases including recent progress on class II enzymes. Exp Mol Med 2020; 52:204-212. [PMID: 32071378 PMCID: PMC7062823 DOI: 10.1038/s12276-020-0382-4] [Citation(s) in RCA: 247] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/27/2019] [Indexed: 01/07/2023] Open
Abstract
The interaction between histones and DNA is important for eukaryotic gene expression. A loose interaction caused, for example, by the neutralization of a positive charge on the histone surface by acetylation, induces a less compact chromatin structure, resulting in feasible accessibility of RNA polymerase and increased gene expression. In contrast, the formation of a tight chromatin structure due to the deacetylation of histone lysine residues on the surface by histone deacetylases enforces the interaction between the histones and DNA, which minimizes the chance of RNA polymerases contacting DNA, resulting in decreased gene expression. Therefore, the balance of the acetylation of histones mediated by histone acetylases (HATs) and histone deacetylases (HDACs) is an issue of transcription that has long been studied in relation to posttranslational modification. In this review, current knowledge of HDACs is briefly described with an emphasis on recent progress in research on HDACs, especially on class IIa HDACs. Targeting specific structural and functional features of enzymes involved in regulating the interactions between DNA and the histone proteins associated with it could lead to the development of more effective cancer therapeutics. Histone deacetylases (HDACs), enzymes which remove acetyl groups from histones, make the histones wrap more tightly around the DNA so that it becomes inaccessible to the initial steps in gene expression. Drugs that target these enzymes have shown limited efficacy due to lack of specificity and off-target toxicity. Jeong-Sun Kim at Chonnam National University, Gwangju, and Suk-Youl Park at Pohang Accelerator Laboratory, Pohang University of Science and Technology, South Korea, review the latest knowledge about class II HDACs. They suggest that their unique structural features and low enzymatic activity are important features to consider when designing new, more selective HDAC inhibitors.
Collapse
|
21
|
Liao B, Sun Q, Yuan Y, Yin Y, Qiao J, Jiang P. Histone deacetylase inhibitor MGCD0103 causes cell cycle arrest, apoptosis, and autophagy in liver cancer cells. J Cancer 2020; 11:1915-1926. [PMID: 32194803 PMCID: PMC7052879 DOI: 10.7150/jca.34091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/01/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Liver cancer is a common cause of cancer-related death all over the world. MGCD0103, a histone deacetylase inhibitor, exerts antitumor effect on various cancers. However, its role in liver cancer remains unclear. Methods: The effect of MGCD0103 on HepG2 and Huh7 cells was verified by several experiments such as cell viability assay, colony formation assay, cell cycle analysis, apoptosis analysis, reactive oxygen species (ROS) assay, western blotting, immunohistochemistry, and xenograft assay. Results: Cell viability and colony formation assays showed that MGCD0103 inhibited the proliferation of liver cancer cells in vitro. Flow cytometry and western blotting analysis demonstrated that MGCD0103 induced G2/M phase arrest and mitochondrial-related apoptosis. A pan-caspase inhibitor and ROS scavenger inhibited apoptosis induced by MGCD0103. What's more, MGCD0103 led to autophagy associated with cell death and an autophagy inhibitor inhibited apoptosis and autophagy induced by MGCD0103. Ultimately, MGCD0103 attenuated tumor growth but did not show significant systemic toxicity in animal model. Conclusions: MGCD0103 suppressed the growth of liver cancer cells in vitro and in vivo. It could serve as a novel therapeutic approach for liver cancer.
Collapse
Affiliation(s)
- Bo Liao
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Quan Sun
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yufeng Yuan
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuchun Yin
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jianguo Qiao
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ping Jiang
- Department of Hepatopancreatobiliary Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
22
|
The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 2019; 19:184-197. [PMID: 30718831 DOI: 10.1038/s41577-019-0125-8] [Citation(s) in RCA: 747] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.
Collapse
|
23
|
Dai Q, Zhang C, Yuan Z, Sun Q, Jiang Y. Current discovery strategies for hepatocellular carcinoma therapeutics. Expert Opin Drug Discov 2019; 15:243-258. [PMID: 31809618 DOI: 10.1080/17460441.2020.1696769] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Qiuzi Dai
- Department of Chemistry, Tsinghua University, Beijing, PR China
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
- Shenzhen Bay Laboratory, Shenzhen, PR China
| | - Cunlong Zhang
- Shenzhen Bay Laboratory, Shenzhen, PR China
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, Shenzhen Kivita Innovative Drug Discovery Institute, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
| | - Zigao Yuan
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
- Shenzhen Bay Laboratory, Shenzhen, PR China
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, Shenzhen Kivita Innovative Drug Discovery Institute, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
| | - Qinsheng Sun
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
- Shenzhen Bay Laboratory, Shenzhen, PR China
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, Shenzhen Kivita Innovative Drug Discovery Institute, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
| | - Yuyang Jiang
- National & Local United Engineering Lab for Personalized Anti-tumor Drugs, The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, the Graduate School at Shenzhen, Tsinghua University, Shenzhen, PR China
- Shenzhen Bay Laboratory, Shenzhen, PR China
- Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing, P. R. China
| |
Collapse
|
24
|
Tsai CH, Li CH, Liao PL, Chang YW, Cheng YW, Kang JJ. Aza-PBHA, a potent histone deacetylase inhibitor, inhibits human gastric-cancer cell migration via PKCα-mediated AHR-HDAC interactions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118564. [PMID: 31672612 DOI: 10.1016/j.bbamcr.2019.118564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022]
Abstract
Recently, histone deacetylase inhibitors (HDACi) have become widely used in anti-cancer treatment; however, due to acquired drug resistance and their relatively low specificity, they are largely ineffective against late-stage cancer. Thus, it is critical to elucidate the molecular mechanisms underlying these issues, so as to identify novel therapeutic targets to prevent late-stage cancer progression and resistance acquisition. The present study investigated the Aryl hydrocarbon receptor (AHR), that has been shown to mediate histone acetylation by regulating histone deacetylase (HDAC) activity during HDACi treatment in human gastric-cancer cell lines (i.e. AGS and NCI-N87 cells). The potent HDACi, Aza-PBHA, was thus shown to upregulate AHR expression in both AGS and NCI-N87 cell lines, and to increase histone acetylation levels by facilitating AHR/HDAC interactions. Conversely, AHR knockdown increased HDAC activity. Aza-PBHA also increased PKCα phosphorylation and membrane translocation; however, interestingly, PKCα inhibition reduced the Aza-PBHA-increased AHR and histone acetylation levels, and inhibited the formation of the AHR/HDAC complex, likely upregulating Aza-PBHA-inhibited cell migration. Thus, our results suggest that Aza-PBHA treatment increased AHR levels to suppress HDAC activity, and inhibited cell migration by activating PKCα activation. These findings support the use of drugs to control AHR-related epigenetic regulation as a promising potential method to prevent acquired resistance to cancer treatments.
Collapse
Affiliation(s)
- Chi-Hao Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Ching-Hao Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan.
| | - Po-Lin Liao
- Institute of Food Safety and Health Assessment, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wei Chang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yu-Wen Cheng
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, College of Pharmacy, Taipei Medical University, Taiwan; Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taiwan.
| | - Jaw-Jou Kang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Faculty of Pharmacy, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
25
|
Zhang C, Huang C, Sui X, Zhong X, Yang W, Hu X, Li Y. Association between gene methylation and HBV infection in hepatocellular carcinoma: A meta-analysis. J Cancer 2019; 10:6457-6465. [PMID: 31772678 PMCID: PMC6856736 DOI: 10.7150/jca.33005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
Gene methylation is an epigenetic alteration in hepatocellular carcinoma (HCC), and hepatitis B virus (HBV) plays a crucial role in carcinogenesis of HCC. However, the association between gene methylation and HBV infection in HCC remains unclear. In our study, we conducted a comprehensive meta-analysis to evaluate the association. A total of 1,148 studies were initially retrieved from some literature database. After a four-step filtration, we obtained 69 case-control studies in this meta-analysis. Our results showed six genes (p16, RASSF1A, GSTP1, APC, p15 and SFRP1) in HBV-positive carcinoma tissues, one gene (GSTP1) in HBV-positive adjacent tissues and two gene (p16 and APC) in HBV-positive carcinoma serums, which were significantly hypermethylated. Subgroup meta-analysis by geographical populations revealed that GSTP1 methylation was significantly higher in HBV-positive carcinoma tissues in China and Japan. In addition, p16 and RASSF1A methylation was significantly higher in China but not in Japan. Our study indicated that HBV infection could induce DNA methylation in HCC and DNA methylation could lead to the development of HBV-related HCC.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changxin Huang
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xinbing Sui
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xueqing Zhong
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wenjun Yang
- Department of Pathology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiangrong Hu
- Department of Pathology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yongqiang Li
- Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Yu L, Xie R, Tian T, Zheng L, Tang L, Cai S, Ma Z, Yang T, Han B, Yang Q. Suberoylanilide hydroxamic acid upregulates histone acetylation and activates endoplasmic reticulum stress to induce apoptosis in HepG2 liver cancer cells. Oncol Lett 2019; 18:3537-3544. [PMID: 31516571 PMCID: PMC6732958 DOI: 10.3892/ol.2019.10705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/26/2019] [Indexed: 01/07/2023] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor that has demonstrated clinical activity against various solid tumors. The aim of the present study was to explore the effects of SAHA on the apoptosis of HepG2 liver cancer cells, as well as the potential mechanisms involved in histone acetylation and endoplasmic reticulum (ER) stress. HepG2 cells were treated with various doses of SAHA (0, 1, 6 and 12 µM), and apoptosis was measured by flow cytometry. The levels of ER stress-associated molecules, including 78 kDa glucose-regulated protein (GRP78), PRKR-like endoplasmic reticulum kinase (PERK), phosphorylated (p)-PERK, activating transcription factor 4 (ATF4) and C/EBP-homologous protein (CHOP), were quantitated by western blot analysis and reverse transcription-quantitative PCR assay. The expression levels of acetylated histone H4 (acH4, acH4 lysine (K)5 and acH4K12) were detected by western blot analysis. The effects of SAHA on the acetylation of H4 in the promoter regions of GRP78, ATF4 and CHOP were evaluated by chromatin immunoprecipitation assays. Following treatment with higher doses of SAHA (6 and 12 µM) for 48 h, the proliferation of HepG2 cells was significantly suppressed. SAHA induced dose-dependent apoptosis and increased both protein and mRNA expression levels of GRP78, ATF4 and CHOP in HepG2 cells. The protein expression of PERK was markedly decreased by treatment with SAHA, whereas the p-PERK expression level was notably increased, which resulted in increased p-PERK/PERK ratio. Furthermore, the acetylation levels of H4 in the promoter regions of GRP78, ATF4 and CHOP were significantly increased in HepG2 cells exposed to 6 µM SAHA for 36 h. Thus, SAHA induces apoptosis in HepG2 cells by activating the ER stress-mediated apoptotic signaling pathway, at least partially by enhancing the acetylation of histone H4 on the promoter regions of ER-stress associated genes, including GRP78, ATF4 and CHOP.
Collapse
Affiliation(s)
- Lei Yu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathology, Hospital of Maternal and Childhood Health, Guiyang, Guizhou 550000, P.R. China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Tian Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lu Zheng
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lei Tang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Shuang Cai
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zihua Ma
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Qin Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
27
|
Park SY, Kim GS, Hwang HJ, Nam TH, Park HS, Song J, Jang TH, Lee YC, Kim JS. Structural basis of the specific interaction of SMRT corepressor with histone deacetylase 4. Nucleic Acids Res 2019; 46:11776-11788. [PMID: 30321390 PMCID: PMC6294515 DOI: 10.1093/nar/gky926] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 10/04/2018] [Indexed: 11/22/2022] Open
Abstract
Modification of chromatin and related transcription factors by histone deacetylases (HDACs) is one of the major strategies for controlling gene expression in eukaryotes. The HDAC domains of class IIa HDACs repress the respective target genes by interacting with the C-terminal region of the silencing mediator for retinoid and thyroid receptor (SMRT) repression domain 3 (SRD3c). However, latent catalytic activity suggests that their roles as deacetylases in gene regulation are unclear. Here, we found that two conserved GSI-containing motifs of SRD3c are critical for HDAC4 binding. Two SMRT peptides including these motifs commonly form a β-hairpin structure in the cleft and block the catalytic entry site of HDAC4. They interact mainly with class IIa HDAC-specific residues of HDAC4 in a closed conformation. Structure-guided mutagenesis confirmed critical interactions between the SMRT peptides and HDAC4 and –5 as well as the contribution of the Arg1369 residue in the first motif for optimal binding to the two HDACs. These results indicate that SMRT binding does not activate the cryptic deacetylase activity of HDAC4 and explain how class IIa HDACs and the SMRT-HDAC3 complex are coordinated during gene regulation.
Collapse
Affiliation(s)
- Suk-Youl Park
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea.,Pohang Accelerator Laboratory, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Gwang Sik Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyo-Jeong Hwang
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Taek-Hyun Nam
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hee-Sae Park
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jaeyoung Song
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Tae-Ho Jang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Young Chul Lee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jeong-Sun Kim
- Department of Chemistry, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
28
|
Inhibition of protein phosphatase 1 stimulates noncanonical ER stress eIF2α activation to enhance fisetin-induced chemosensitivity in HDAC inhibitor-resistant hepatocellular carcinoma cells. Cancers (Basel) 2019; 11:cancers11070918. [PMID: 31261976 PMCID: PMC6678694 DOI: 10.3390/cancers11070918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common fatal type of malignant tumor that has highly metastatic and recurrent properties. Fisetin is a natural flavonoid found in various vegetables and fruits which exhibits anti-cancer and anti-inflammatory properties, as well as other effects. Thus, we hypothesized that fisetin can act as an adjuvant therapy in cancer or drug-resistant cancer cells, and further investigated the molecular mechanisms underlying the development of drug-resistance in HCC cells. We found that fisetin effectively inhibited the cell viability of not only parental cells but also histone deacetylase inhibitors-resistant (HDACis-R) cells and enhanced the chemosensitivity of HCC cells. Interestingly, fisetin did not induce cell apoptosis through the activation of the endoplasmic reticulum (ER) stress sensor of protein kinase R (PKR)-like endoplasmic reticulum kinase, but rather through the non-canonical pathway of the protein phosphatase 1 (PP1)-mediated suppression of eIF2α phosphorylation. Moreover, fisetin-induced cell apoptosis was reversed by treatment with PP1 activator or eIF2α siRNA in HCC cells. Based on these observations, we suggest that PP1-eIF2α pathways are significantly involved in the effect of fisetin on HCC apoptosis. Thus, fisetin may act as a novel anticancer drug and new chemotherapy adjuvant which can improve the efficacy of chemotherapeutic agents and diminish their side-effects.
Collapse
|
29
|
Jin G, Liu Y, Zhang J, Bian Z, Yao S, Fei B, Zhou L, Yin Y, Huang Z. A panel of serum exosomal microRNAs as predictive markers for chemoresistance in advanced colorectal cancer. Cancer Chemother Pharmacol 2019; 84:315-325. [PMID: 31089750 DOI: 10.1007/s00280-019-03867-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chemoresistance is a common problem for cancer treatment worldwide. Circulating exosomal microRNAs (miRNAs) have been considered as promising biomarkers of cancers. However, few studies have assessed the relationship between serum/plasma exosomal microRNAs and chemoresistance in colorectal cancer (CRC). METHODS Based on previous microarray analysis, we selected 30 miRNAs which are aberrantly expressed during CRC progression and then detected their expression levels in three pairs of oxaliplatin/5-fluorouracil-resistant CRC cell lines and the corresponding secreted exosomes. Six candidate exosomal miRNAs were identified for further evaluating potential value in predicting chemotherapeutic effect in advanced CRC patients. Finally, the molecular mechanisms of these miRNAs in drug resistance were explored by bioinformatics preliminarily. RESULTS We observed that the expression of 14 miRNAs was significantly higher in three drug-resistant CRC cells comparing with their parental cells. Among these miRNAs, miR-21-5p, miR-1246, miR-1229-5p, miR-135b, miR-425 and miR-96-5p are also up-regulated in exosomes from culture media of resistant cells. Clinical sample analysis confirmed that the expression levels of miR-21-5p, miR-1246, miR-1229-5p and miR-96-5p in serum exosomes were significantly higher in chemoresistant patients in contrast with chemosensitive controls. ROC curve showed that the combination of the four miRNAs had an area of under the curve (AUC) of 0.804 (P < 0.05). In addition, GO analysis and KEGG pathway analysis revealed that these miRNAs were enriched in PI3K-Akt signaling pathway, FoxO signaling pathway and autophagy pathway. CONCLUSIONS Our study demonstrates that a panel of serum exosomal miRNAs containing miR-21-5p, miR-1246, miR-1229-5p and miR-96-5p could significantly distinguish the chemotherapy-resistant group from advanced colorectal cancer patients. Targeting these miRNAs may promote chemosensitivity to oxaliplatin and 5-fluorouracil, and might be promising strategy for CRC treatment.
Collapse
Affiliation(s)
- Guoying Jin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China.,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yuhang Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China.,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jia Zhang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China.,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zehua Bian
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China.,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Surui Yao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China.,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Bojian Fei
- Department of Surgical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Leyuan Zhou
- Department of Radiation Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, 214062, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China. .,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, Jiangsu, China. .,Cancer Epigenetics Program, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
30
|
Role of Gut Microbiota in Hepatocarcinogenesis. Microorganisms 2019; 7:microorganisms7050121. [PMID: 31060311 PMCID: PMC6560397 DOI: 10.3390/microorganisms7050121] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/23/2019] [Accepted: 05/03/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC), one of the leading causes of death worldwide, has a causal nexus with liver injury, inflammation, and regeneration that accumulates over decades. Observations from recent studies have accounted for the involvement of the gut–liver axis in the pathophysiological mechanism responsible for HCC. The human intestine nurtures a diversified colony of microorganisms residing in the host ecosystem. The intestinal barrier is critical for conserving the normal physiology of the gut microbiome. Therefore, a rupture of this barrier or dysbiosis can cause the intestinal microbiome to serve as the main source of portal-vein endotoxins, such as lipopolysaccharide, in the progression of hepatic diseases. Indeed, increased bacterial translocation is a key sign of HCC. Considering the limited number of clinical studies on HCC with respect to the microbiome, we focus on clinical as well as animal studies involving the gut microbiota, with the current understandings of the mechanism by which the intestinal dysbiosis promotes hepatocarcinogenesis. Future research might offer mechanistic insights into the specific phyla targeting the leaky gut, as well as microbial dysbiosis, and their metabolites, which represent key pathways that drive HCC-promoting microbiome-mediated liver inflammation and fibrosis, thereby restoring the gut barrier function.
Collapse
|
31
|
Qin J, Wen B, Liang Y, Yu W, Li H. Histone Modifications and their Role in Colorectal Cancer (Review). Pathol Oncol Res 2019; 26:2023-2033. [PMID: 31055775 PMCID: PMC7471167 DOI: 10.1007/s12253-019-00663-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
The development of colorectal cancer is a complex and multistep process mediated by a variety of factors including the dysregulation of genetic and epigenetic under the influence of microenvironment. It is evident that epigenetics that affects gene activity and expression has been recognized as a critical role in the carcinogenesis. Aside from DNA methylation, miRNA level, and genomic imprinting, histone modification is increasingly recognized as an essential mechanism underlying the occurrence and development of colorectal cancer. Aberrant regulation of histone modification like acetylation, methylation and phosphorylation levels on specific residues is implicated in a wide spectrum of cancers, including colorectal cancer. In addition, as this process is reversible and accompanied by a plethora of deregulated enzymes, inhibiting those histone-modifying enzymes activity and regulating its level has been thought of as a potential path for tumor therapy. This review provides insight into the basic information of histone modification and its application in the colorectal cancer treatment, thereby offering new potential targets for treatment of colorectal cancer.
Collapse
Affiliation(s)
- Jingchun Qin
- Institute of Spleen and Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Bin Wen
- Institute of Spleen and Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| | - Yuqi Liang
- Institute of Spleen and Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Weitao Yu
- Lianyungang Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Huixuan Li
- Institute of Spleen and Stomach, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| |
Collapse
|
32
|
Wu M, Liu Z, Li X, Zhang A, Lin D, Li N. Analysis of potential key genes in very early hepatocellular carcinoma. World J Surg Oncol 2019; 17:77. [PMID: 31043166 PMCID: PMC6495517 DOI: 10.1186/s12957-019-1616-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/14/2019] [Indexed: 02/07/2023] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the major pathological type of primary liver cancer, one of the leading causes of cancer death worldwide. In addition, the long-term survival rates of HCC still remain low. Therefore, we attempted to identify the potential key genes in the occurrence of HCC by comparing the expression profiles of very early HCC tissue samples with that of chronic cirrhotic tissue samples by integrating the bioinformatics analysis in this study. Methods Gene expression profiles of 19 very early HCC and 19 cirrhotic tissue samples were selected from GSE63898. Differentially expressed genes (DEGs) were also identified by using online tool GEO2R. Furthermore, the GO and KEGG enrichment analysis of the DGEs were conducted on DAVID datasets. Then a protein–protein interaction (PPI) network was constructed and the modules were analyzed based on STRING database and Cytoscape software. The hub genes were screened by applying the cytoHubba plugin and then analyzed with the Kaplan Meier plotter. Results A total of 118 DEGs were identified between very early HCC and cirrhotic tissue samples. These DGEs were strongly associated with several biological processes, such as negative regulation of growth and p53 signaling pathway. A PPI network was constructed and top eight hub genes, including CDKN3, CDK1, CCNB1, TOP2A, CCNA2, CCNB2, PRC1, and RRM2, were determined. High expressions of CDK1, CCNB1, TOP2A, CCNA2, PRC1, RRM2, CDKN3, and CCNB2 were associated with poorer overall survivals (OS) in HCC patients. Conclusion We had compared the expression profiles between the very early HCC and cirrhotic tissue samples by using bioinformatics analysis tools, which might help us better to understand the molecular mechanism of the initiation of HCC and even to find novel targets for HCC therapy.
Collapse
Affiliation(s)
- Min Wu
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China
| | - Zhaobo Liu
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China
| | - Xin Li
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China
| | - Aiying Zhang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China
| | - Dongdong Lin
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China
| | - Ning Li
- Department of General Surgery, Beijing Youan Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai, Feng-tai District, Beijing, 100069, China.
| |
Collapse
|
33
|
Li YL, Rao MJ, Zhang NY, Wu LW, Lin NM, Zhang C. BAY 87-2243 sensitizes hepatocellular carcinoma Hep3B cells to histone deacetylase inhibitors treatment via GSK-3β activation. Exp Ther Med 2019; 17:4547-4553. [PMID: 31186678 DOI: 10.3892/etm.2019.7500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with some of the highest cancer-associated mortality rates. Histone deacetylase (HDAC) inhibitors anti-HCC activities have been shown to promote Snail-induced metastasis. In the present study, it was shown that BAY 87-2243, a hypoxia-inducible transcription factor-1α inhibitor, could enhance the anti-HCC effects of HDAC inhibitors, including trichostatin A and vorinostat. In addition, BAY 87-2243 plus HDAC inhibitors exhibited synergistic cytotoxicity and induced significant cell death in Hep3B cells. Additionally, BAY 87-2243 combined with HDAC inhibitors-treated Hep3B cells formed fewer and smaller colonies as compared with either the control or single agent-treated cells. Furthermore, glycogen synthase kinase-3β might be involved in the enhanced cell death induced by BAY 87-2243 plus HDAC inhibitors. The present data also indicated that BAY 87-2243 combined with HDAC inhibitors could suppress the migration of Hep3B cells, and BAY 87-2243 could reverse the HDAC inhibitor-induced Snail activation in Hep3B cells. In conclusion, BAY 87-2243 combined with HDAC inhibitors might be an attractive chemotherapy strategy for HCC therapy.
Collapse
Affiliation(s)
- Yang-Ling Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Ming-Jun Rao
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China.,Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, P.R. China
| | - Ning-Yu Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| | - Lin-Wen Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Neng-Ming Lin
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China.,Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, P.R. China
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| |
Collapse
|
34
|
Ferreira RG, Cardoso MV, de Souza Furtado KM, Espíndola KMM, Amorim RP, Monteiro MC. Epigenetic alterations caused by aflatoxin b1: a public health risk in the induction of hepatocellular carcinoma. Transl Res 2019; 204:51-71. [PMID: 30304666 DOI: 10.1016/j.trsl.2018.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/27/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023]
Abstract
Aflatoxin B1 (AFB1) is currently the most commonly studied mycotoxin due to its great toxicity, its distribution in a wide variety of foods such as grains and cereals and its involvement in the development of + (hepatocellular carcinoma; HCC). HCC is one of the main types of liver cancer, and has become a serious public health problem, due to its high incidence mainly in Southeast Asia and Africa. Studies show that AFB1 acts in synergy with other risk factors such as hepatitis B and C virus leading to the development of HCC through genetic and epigenetic modifications. The genetic modifications begin in the liver through the biomorphic AFB1, the AFB1-exo-8.9-Epoxy active, which interacts with DNA to form adducts of AFB1-DNA. These adducts induce mutation in codon 249, mediated by a transversion of G-T in the p53 tumor suppressor gene, causing HCC. Thus, this review provides an overview of the evidence for AFB1-induced epigenetic alterations and the potential mechanisms involved in the development of HCC, focusing on a critical analysis of the importance of severe legislation in the detection of aflatoxins.
Collapse
Affiliation(s)
- Roseane Guimarães Ferreira
- Neurosciences and Cell Biology Post-Graduation Program, Biological Sciences Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil.
| | - Magda Vieira Cardoso
- Pharmaceutical Science Post-Graduation Program, Health Science Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil.
| | | | | | | | - Marta Chagas Monteiro
- Neurosciences and Cell Biology Post-Graduation Program, Pharmaceutical Science Post-Graduation Program, Health Science Institute, Federal University of Pará/UFPA, Belém, Pará, Brazil.
| |
Collapse
|
35
|
Patrizi B, Siciliani de Cumis M. TCDD Toxicity Mediated by Epigenetic Mechanisms. Int J Mol Sci 2018; 19:ijms19124101. [PMID: 30567322 PMCID: PMC6320947 DOI: 10.3390/ijms19124101] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Dioxins are highly toxic and persistent halogenated organic pollutants belonging to two families i.e., Polychlorinated Dibenzo-p-Dioxins (PCDDs) and Polychlorinated Dibenzo Furans (PCDFs). They can cause cancer, reproductive and developmental issues, damage to the immune system, and can deeply interfere with the endocrine system. Dioxins toxicity is mediated by the Aryl-hydrocarbon Receptor (AhR) which mediates the cellular metabolic adaptation to these planar aromatic xenobiotics through the classical transcriptional regulation pathway, including AhR binding of ligand in the cytosol, translocation of the receptor to the nucleus, dimerization with the AhR nuclear translocator, and the binding of this heterodimeric transcription factor to dioxin-responsive elements which regulate the expression of genes involved in xenobiotic metabolism. 2,3,7,8-TCDD is the most toxic among dioxins showing the highest affinity toward the AhR receptor. Beside this classical and well-studied pathway, a number of papers are dealing with the role of epigenetic mechanisms in the response to environmental xenobiotics. In this review, we report on the potential role of epigenetic mechanisms in dioxins-induced cellular response by inspecting recent literature and focusing our attention on epigenetic mechanisms induced by the most toxic 2,3,7,8-TCDD.
Collapse
Affiliation(s)
- Barbara Patrizi
- National Institute of Optics-National Research Council (INO-CNR), Via Madonna del Piano 10, 50019 Sesto Fiorentino, Italy.
- European Laboratory for Non-Linear Spectroscopy (LENS), Via Nello Carrara 1, 50019 Sesto Fiorentino, Italy.
| | | |
Collapse
|
36
|
Yang L, Chang Y, Cao P. CCR7 preservation via histone deacetylase inhibition promotes epithelial-mesenchymal transition of hepatocellular carcinoma cells. Exp Cell Res 2018; 371:231-237. [PMID: 30107147 DOI: 10.1016/j.yexcr.2018.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/16/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
The effects of Histone deacetylase (HDAC) inhibition on epithelial-mesenchymal transition (EMT) differs in various types of cancers. However, its function in hepatocellular carcinoma (HCC) is not well-explored. In this study, we investigated the effect of HDAC inhibition on EMT in HCC cells by using trichostatin A (TSA) and valproic acid (VPA). The results showed that TSA/VPA significantly induced EMT phenotype, as demonstrated by the decreased level of E-cadherin, increased level of N-cadherin, vimentin, Twist and snail, and enhanced capacity of cell migration and invasion. In addition, CCR7 was speculated and confirmed as a function target of HDAC inhibition. CCR7 promotes the progression of HCC and is associated with poor survival. Knockdown of CCR7 significantly attenuated the effect of TSA on EMT. Moreover, our results demonstrated that HDAC inhibition up-regulates CCR7 via reversing the promoter hypoacetylation and increasing CCR7 transcription. Taken together, our study has identified the function of HDAC in EMT of HCC and suggested a novel mechanism through which TSA/VPA exerts its carcinogenic roles in HCC. HDAC inhibitors require careful caution before their application as new anticancer drugs.
Collapse
Affiliation(s)
- Lingling Yang
- Department of Gastroenterology, Baoji Central hospital, Baoji 721008, China
| | - Yanxiang Chang
- Department of Oncology, the First Affiliated Hospital of Xi'an Medical University, Xi'an 710003, China
| | - Peilong Cao
- Department of Pathology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
37
|
Anticancer effect of histone deacetylase inhibitor scriptaid as a single agent for hepatocellular carcinoma. Biosci Rep 2018; 38:BSR20180360. [PMID: 29945926 PMCID: PMC6043718 DOI: 10.1042/bsr20180360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 12/23/2022] Open
Abstract
Recurrence is one of the major causes of poor prognosis for patients with hepatocellular carcinoma (HCC), and drug resistance is closely associated with disease recurrence. Histone deacetylase (HDAC) inhibitor scriptaid functions as an anticancer agent in many different types of tumors, but its possible roles in HCC progression have not been explored to date. Herein, we show that HDAC inhibitor scriptaid decreases HCC cell proliferation and induces cell cycle G2/M-phase arrest in a dose-dependent manner. Furthermore, scriptaid triggered HCC cell death via transcriptional activation of p21 and subsequent elevated global H3Ac levels. Importantly, we found that scriptaid showed robust antitumor activity against HCC. Thus, our findings indicate that HDAC inhibitor scriptaid could be an important potential candidate for treatment of HCC patients.
Collapse
|
38
|
Li S, Yang E, Shen L, Niu D, Breitzig M, Tan LC, Wu X, Huang M, Sun H, Wang F. The novel truncated isoform of human manganese superoxide dismutase has a differential role in promoting metastasis of lung cancer cells. Cell Biol Int 2018; 42:1030-1040. [DOI: 10.1002/cbin.10972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 04/06/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Shuaiguang Li
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Enze Yang
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Lianghua Shen
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Dewei Niu
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Mason Breitzig
- University of South Florida; 12901 Bruce B Downs Blvd, MDC 19 Tampa 33612 Florida
| | - Lee Charles Tan
- University of South Florida; 12901 Bruce B Downs Blvd, MDC 19 Tampa 33612 Florida
| | - Xiaocong Wu
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Meiyan Huang
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Hanxiao Sun
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| | - Feng Wang
- Institute of Genomic Medicine; College of Pharmacy, Jinan University; Guangzhou 510632 China
- Guangdong Provincial Key Laboratory of Pharmacodynamics Constituents of TCM and New Drugs Research; Jinan University; Guangzhou 510632 China
| |
Collapse
|