1
|
Ahmadpour Youshanlui M, Yari A, Bahojb Mahdavi SZ, Amini M, Baradaran B, Ahangar R, Pourbagherian O, Mokhtarzadeh AA. BRD4 expression and its regulatory interaction with miR-26a-3p, DLG5-AS1, and JMJD1C-AS1 lncRNAs in gastric cancer progression. Discov Oncol 2024; 15:356. [PMID: 39152304 PMCID: PMC11329449 DOI: 10.1007/s12672-024-01230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024] Open
Abstract
Gastric cancer remains a significant health challenge despite advancements in diagnosis and treatment. Early detection is critical to reducing mortality, necessitating the investigation of molecular mechanisms underlying gastric cancer progression. This study focuses on BRD4 expression and its correlation with miR-26a-3p, DLG5-AS1, and JMJD1C-AS1 lncRNAs in gastric cancer. Analysis of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets revealed significant upregulation of BRD4 in gastric cancer tissues compared to normal tissues, correlating negatively with miR-26a-3p and positively with DLG5-AS1 and JMJD1C-AS1 lncRNAs. Quantitative RT-PCR confirmed these findings in 25 gastric cancer tissue samples and 25 normal samples. BRD4's overexpression was associated with reduced survival rates and older patient age. MiR-26a-3p, a known tumor suppressor, showed decreased expression in gastric cancer tissues, with ROC analysis suggesting it, alongside BRD4, as a potential diagnostic biomarker. Additionally, bioinformatics predicted miR-26a-3p's interaction with BRD4 mRNA. Upregulated lncRNAs DLG5-AS1 and JMJD1C-AS1 likely act as competing endogenous RNAs, sponging miR-26a-3p, thus promoting BRD4 dysregulation. These lncRNAs have not been previously studied in gastric cancer. The findings propose a novel BRD4/lncRNA/miRNA regulatory axis in gastric cancer, highlighting the potential of BRD4, DLG5-AS1, and JMJD1C-AS1 as biomarkers for early diagnosis. Further studies with larger sample sizes and in vivo and in vitro experiments are needed to elucidate this regulatory mechanism's role in gastric cancer progression.
Collapse
Affiliation(s)
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Ahangar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Pourbagherian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
2
|
Yongprayoon V, Wattanakul N, Khomate W, Apithanangsiri N, Kasitipradit T, Nantajit D, Tavassoli M. Targeting BRD4: Potential therapeutic strategy for head and neck squamous cell carcinoma (Review). Oncol Rep 2024; 51:74. [PMID: 38606512 DOI: 10.3892/or.2024.8733] [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: 02/08/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024] Open
Abstract
As a member of BET (bromodomain and extra-terminal) protein family, BRD4 (bromodomain‑containing protein 4) is a chromatin‑associated protein that interacts with acetylated histones and actively recruits regulatory proteins, leading to the modulation of gene expression and chromatin remodeling. The cellular and epigenetic functions of BRD4 implicate normal development, fibrosis and inflammation. BRD4 has been suggested as a potential therapeutic target as it is often overexpressed and plays a critical role in regulating gene expression programs that drive tumor cell proliferation, survival, migration and drug resistance. To address the roles of BRD4 in cancer, several drugs that specifically target BRD4 have been developed. Inhibition of BRD4 has shown promising results in preclinical models, with several BRD4 inhibitors undergoing clinical trials for the treatment of various cancers. Head and neck squamous cell carcinoma (HNSCC), a heterogeneous group of cancers, remains a health challenge with a high incidence rate and poor prognosis. Conventional therapies for HNSCC often cause adverse effects to the patients. Targeting BRD4, therefore, represents a promising strategy to sensitize HNSCC to chemo‑ and radiotherapy allowing de‑intensification of the current therapeutic regime and subsequent reduced side effects. However, further studies are required to fully understand the underlying mechanisms of action of BRD4 in HNSCC in order to determine the optimal dosing and administration of BRD4‑targeted drugs for the treatment of patients with HNSCC.
Collapse
Affiliation(s)
- Voraporn Yongprayoon
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Napasporn Wattanakul
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Winnada Khomate
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Nathakrit Apithanangsiri
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Tarathip Kasitipradit
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Danupon Nantajit
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Mahvash Tavassoli
- Centre for Host Microbiome Interactions, King's College London, London SE1 1UL, UK
| |
Collapse
|
3
|
Hua R, Chu Q, Guo F, Chen Q, Li M, Zhou X, Zhu Y. DNM3OS Enhances the Apoptosis and Senescence of Spermatogonia Associated with Nonobstructive Azoospermia by Providing miR-214-5p and Decreasing E2F2 Expression. Anal Cell Pathol (Amst) 2023; 2023:1477658. [PMID: 38152068 PMCID: PMC10752680 DOI: 10.1155/2023/1477658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023] Open
Abstract
Background Nonobstructive azoospermia (NOA) is a complex disease characterized by the spermatogenic dysfunction of testicular tissues. The roles played by long noncoding RNAs (lncRNAs) in NOA pathogenesis have not been extensively studied. Methods Microarray assays were performed on samples of testicular biopsy tissue obtained from patients with NOA for the purpose of identifying differentially expressed lncRNAs and messenger RNA (mRNA) transcripts, and the results were verified by quantitative real-time polymerase chain reaction. Mouse-derived GC-1 spermatogonia (spg) cells undergoing treatment with Adriamycin (ADR) were used to investigate the biological functions of the selected lncRNAs in vitro. The target microRNAs (miRNAs) of lncRNAs and the target mRNAs of miRNAs were predicted by a bioinformatics analysis. Functional studies performed using the CCK-8 assay, EdU incorporation assay, apoptosis detection, and senescence-associated β-galactosidase (SA-β-Gal) staining were conducted using GC-1 spg cells. Results Totals of 2,652 lncRNAs and 2,625 mRNAs were found to be differentially expressed in the testicular tissue of NOA patients when compared with patients in a control group. Dynamin 3 opposite strand (DNM3OS) was a provider of pe-miR-214-5p that positively regulates miR-214-5p expression in GC-1 spg cells. The E2 factor (E2F) family of transcription factor 2 (E2F2) was initially predicted and subsequently verified to be a downstream gene of miR-214-5p. E2F2 expression was upregulated after DNM3OS knockdown in ADR-treated GC-1 spg cells. Moreover, knockdown of either DNM3OS or miR-214-5p significantly alleviated ADR-induced decreases in cellular activity and proliferation, as well as increases in apoptosis and senescence of mouse spermatogonial GC-1 spg cells. Conclusions DNM3OS was found to regulate the apoptosis and senescence of spermatogonia by providing miR-214-5p and decreasing E2F2 expression, suggesting it as a novel target for gene therapy of male infertility.
Collapse
Affiliation(s)
- Rui Hua
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingjun Chu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feiyan Guo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinjie Chen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Maocai Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuan Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongtong Zhu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Qu G, Yang G, Chen D, Tang C, Xu Y. E2F2 serves as an essential prognostic biomarker and therapeutic target for human renal cell carcinoma by presenting "E2F2/miR-16-5p/SPTLC1" schema. Transl Oncol 2023; 34:101699. [PMID: 37300925 DOI: 10.1016/j.tranon.2023.101699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a common malignant tumor of the urinary system with high mortality and morbidity. Although E2F2, a classical transcription factor implicated in cell cycle, has been shown to foster tumorigenesis in several human cancers, it could not draw a satisfy answer referring to precise downstream signaling axis in RCC development yet. METHODS Based on the publicly available data from TCGA database, expression patterns of E2F2, SPTLC1 and miR-16-5p were identified, either with the ability to predict the prognosis of patients with RCC, which was further validated in 38 paired RCC tissues and matched adjacent tissues by RT-qPCR and Western blot, respectively. Their cellular biofunctions were evaluated using MTT, EdU, Colony formation and transwell assays. Chromatin immunoprecipitation (ChIP) and luciferase reporter assay were employed to certain the exquisite core transcription regulatory circuitry of E2F2/miR-16-5p/SPTLC1 in RCC progression, which was also determined in xenograft tumor model. RESULTS Consistent with the public TCGA database, E2F2 was significantly increased in RCC tissues and cells, indicating shorter overall survival. Mechanistically, E2F2 served as a transcriptional activator of miR-16-5p, thus accounting for its negative regulation on SPTLC1 expression. E2F2 knockdown-mediated suppressive biofunctions on RCC cells were rescued by miR-16-5p mimics, while this effect was abolished again by SPTLC1 overexpression. Role of E2F2 on RCC tumorigenesis via the miR-16-5p/SPTLC1 axis was verified both in vitro and in vivo. CONCLUSION E2F2 promoted RCC progression via the miR-16-5p/SPTLC1 axis, which may represent a novel prognostic and therapeutic biomarker for RCC.
Collapse
Affiliation(s)
- GenYi Qu
- Department of Urology, ZhuZhou central hospital, ZhuZhou, Hunan Province 412000, China
| | - Guang Yang
- Department of Urology, ZhuZhou central hospital, ZhuZhou, Hunan Province 412000, China
| | - Dan Chen
- Department of Urology, ZhuZhou central hospital, ZhuZhou, Hunan Province 412000, China
| | - Cheng Tang
- Department of Urology, ZhuZhou central hospital, ZhuZhou, Hunan Province 412000, China
| | - Yong Xu
- Department of Urology, ZhuZhou central hospital, ZhuZhou, Hunan Province 412000, China.
| |
Collapse
|
5
|
Yang Z, Liu Y, Cheng Q, Chen T. Targeting super enhancers for liver disease: a review. PeerJ 2023; 11:e14780. [PMID: 36726725 PMCID: PMC9885865 DOI: 10.7717/peerj.14780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/03/2023] [Indexed: 01/28/2023] Open
Abstract
Background Super enhancers (SEs) refer to the ultralong regions of a gene accompanied by multiple transcription factors and cofactors and strongly drive the expression of cell-type-related genes. Recent studies have demonstrated that SEs play crucial roles in regulating gene expression related to cell cycle progression and transcription. Aberrant activation of SEs is closely related to the occurrence and development of liver disease. Liver disease, especially liver failure and hepatocellular carcinoma (HCC), constitutes a major class of diseases that seriously endanger human health. Currently, therapeutic strategies targeting SEs can dramatically prevent disease progression and improve the prognosis of animal models. The associated new approaches to the treatment of related liver disease are relatively new and need systematic elaboration. Objectives In this review, we elaborate on the features of SEs and discuss their function in liver disease. Additionally, we review their application prospects in clinical practice in the future. The article would be of interest to hepatologists, molecular biologists, clinicians, and all those concerned with targeted therapy and prognosis of liver disease. Methodology We searched three bibliographic databases (Web of Science Core Collection, Embase, PubMed) from 01/1981 to 06/2022 for peer-reviewed scientific publications focused on (1) gene treatment of liver disease; (2) current status of SE research; and (3) targeting SEs for liver disease. We included English language original studies only. Results The number of published studies considering the role of enhancers in liver disease is considerable. Since SEs were just defined in 2013, the corresponding data on SEs are scarce: approximately 50 papers found in bibliographic databases on the correlation between enhancers (or SEs) and liver disease. Remarkably, half of these papers were published in the past three years, indicating the growing interest of the scientific community in this issue. Studies have shown that treatments targeting components of SEs can improve outcomes in liver disease in animal and clinical trials. Conclusions The treatment of liver disease is facing a bottleneck, and new treatments are needed. Therapeutic regimens targeting SEs have an important role in the treatment of liver disease. However, given the off-target effect of gene therapy and the lack of clinical trials, the available experimental data are still fragmented and controversial.
Collapse
|
6
|
Baek M, Chai JC, Choi HI, Yoo E, Binas B, Lee YS, Jung KH, Chai YG. Comprehensive transcriptome profiling of BET inhibitor-treated HepG2 cells. PLoS One 2022; 17:e0266966. [PMID: 35486664 PMCID: PMC9053788 DOI: 10.1371/journal.pone.0266966] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 11/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and poor prognosis. Emerging evidence suggests that epigenetic alterations play a crucial role in HCC, suggesting epigenetic inhibition as a promising therapeutic approach. Indeed, the bromodomain and extra-terminal (BET) inhibitors inhibit the proliferation and invasion of various cancers but still lack a strong mechanistic rationale. Here, we identified the differentially expressed mRNAs (DEmRNAs) and lncRNAs (DElncRNAs) in human HCC cell line HepG2 treated with the BET inhibitors, JQ1, OTX015, or ABBV-075. We analyzed the correlation between DEmRNAs and DElncRNAs in common for the three inhibitors based on their expression profiles and performed functional annotation pathway enrichment analysis. Most of these shared DEmRNAs and DElncRNAs, including some novel transcripts, were downregulated, indicating decreased proliferation/adhesion and increased apoptosis/inflammation. Our study suggests that BET proteins play a crucial role in regulating cancer progression-related genes and provide a valuable resource for novel putative biomarkers and therapeutic targets in HCC.
Collapse
Affiliation(s)
- Mina Baek
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
- Institute of Natural Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Jin Choul Chai
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hae In Choi
- Department of Bionanotechnology, Hanyang University, Seoul, Republic of Korea
| | - Eunyoung Yoo
- Department of Bionanotechnology, Hanyang University, Seoul, Republic of Korea
| | - Bert Binas
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
| | - Young Seek Lee
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- * E-mail: (YGC); (KHJ); (YSL)
| | - Kyoung Hwa Jung
- Department of Biopharmaceutical System, Gwangmyeong Convergence Technology Campus of Korea Polytechnic II, Incheon, Republic of Korea
- * E-mail: (YGC); (KHJ); (YSL)
| | - Young Gyu Chai
- Department of Molecular and Life Science, Hanyang University, Ansan, Republic of Korea
- Department of Bionanotechnology, Hanyang University, Seoul, Republic of Korea
- * E-mail: (YGC); (KHJ); (YSL)
| |
Collapse
|
7
|
Xie G, Zhu A, Gu X. Converged DNA Damage Response Renders Human Hepatocellular Carcinoma Sensitive to CDK7 Inhibition. Cancers (Basel) 2022; 14:cancers14071714. [PMID: 35406486 PMCID: PMC8996977 DOI: 10.3390/cancers14071714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. HCC has a dismal five-year mortality estimate of >95%, ranking as the fourth leading cause of cancer-related mortality worldwide. Despite the recent progression in the treatment of HCC with multikinase inhibitors, immunotherapy, and antiangiogenic monoclonal antibodies, among other newly emerging therapeutics, the efficacy has varied among patients, making HCC a high priority for developing novel targeted therapeutic agents. CDK7 has been exploited as a therapeutic target in HCC. In the present study, we demonstrated that HCC cells were highly susceptible to THZ1, a selective covalent CDK7 inhibitor. We further discovered that transcription factor MYC-promoted cell proliferation renders cancer cells hypersensitive to apoptotic cell death with THZ1 treatment. Our findings indicate that targeting CDK7 with THZ1 may be a new plausible strategy for treating HCC, in which MYC plays crucial roles in cell proliferation and tumor growth. Abstract Hepatocellular carcinoma (HCC) is a lethal malignancy with high mortality. The inhibition of cyclin-dependent kinase 7 (CDK7) activity has shown therapeutic efficacy in HCC. However, the underlying molecular mechanisms remain elusive. Here, we show that three HCC lines, HepG2, Hep3B, and SK-Hep-1, were highly susceptible to the CDK7 inhibitor THZ1. In mouse models, THZ1 effectively reduced HepG2 tumor growth and tumor weight. THZ1 arrested cell cycle and triggered MYC-related apoptosis in HepG2. To evaluate how MYC protein levels affected THZ1-induced apoptotic cell death, we overexpressed MYC in HepG2 and found that exogenously overexpressed MYC promoted cell cycle progression and increased cells in the S phase. THZ1 drastically engendered the apoptosis of MYC-overexpressing HepG2 cells in the S and G2/M phases. Importantly, transcription-inhibition-induced apoptosis is associated with DNA damage, and exogenous MYC expression further enhanced the THZ1-induced DNA damage response in MYC-overexpressing HepG2 cells. Consistently, in the HepG2 xenografts, THZ1 treatment was associated with DNA-damage-induced cell death. Together, our data indicate that the converged effect of MYC-promoted cell cycle progression and CDK7 inhibition by THZ1 confers the hypersensitivity of HCC to DNA-damage-induced cell death. Our findings may suggest a new therapeutic strategy of THZ1 against HCC.
Collapse
Affiliation(s)
- Guiqin Xie
- Department of Oral Pathology, Howard University, 600 W. Street NW, Washington, DC 20059, USA;
- Cancer Center, Howard University, 600 W. Street NW, Washington, DC 20059, USA
- Correspondence: (G.X.); (X.G.)
| | - Ailin Zhu
- Department of Oral Pathology, Howard University, 600 W. Street NW, Washington, DC 20059, USA;
| | - Xinbin Gu
- Department of Oral Pathology, Howard University, 600 W. Street NW, Washington, DC 20059, USA;
- Cancer Center, Howard University, 600 W. Street NW, Washington, DC 20059, USA
- Correspondence: (G.X.); (X.G.)
| |
Collapse
|
8
|
Sun HY, Du ST, Li YY, Deng GT, Zeng FR. Bromodomain and extra-terminal inhibitors emerge as potential therapeutic avenues for gastrointestinal cancers. World J Gastrointest Oncol 2022; 14:75-89. [PMID: 35116104 PMCID: PMC8790409 DOI: 10.4251/wjgo.v14.i1.75] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 08/11/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal (GI) cancers, including colorectal cancer, pancreatic cancer, liver cancer and gastric cancer, are severe social burdens due to high incidence and mortality rates. Bromodomain and extra-terminal (BET) proteins are epigenetic readers consisting of four conserved members (BRD2, BRD3, BRD4 and BRDT). BET family perform pivotal roles in tumorigenesis through transcriptional regulation, thereby emerging as potential therapeutic targets. BET inhibitors, disrupting the interaction between BET proteins and acetylated lysines, have been reported to suppress tumor initiation and progression in most of GI cancers. In this review, we will demonstrate how BET proteins participate in the GI cancers progression and highlight the therapeutic potential of targeting BET proteins for GI cancers treatment.
Collapse
Affiliation(s)
- Hui-Yan Sun
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Song-Tao Du
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Department of Colorectal Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Ya-Yun Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Guang-Tong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Fu-Rong Zeng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
9
|
Zhou Y, Zhang J, Li W, Zhang D, Wang Z, Zhai Y, Yu H, Li Z. Integrative investigation of the TF-miRNA coregulatory network involved in the inhibition of breast cancer cell proliferation by resveratrol. FEBS Open Bio 2021; 12:379-393. [PMID: 34856073 PMCID: PMC8804603 DOI: 10.1002/2211-5463.13344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 01/18/2023] Open
Abstract
Resveratrol is a polyphenol with antiaging and anticancer effects. Most previous studies used a single analysis to determine the key functions of resveratrol in inhibiting cancer progression. However, most of the signal transmission pathways in biological processes are multilevel. We used bioinformatics to elucidate the mechanism of resveratrol inhibition of breast cancer development. The mRNA expression profile of GSE25412 from the National Center for Biotechnology Information (NCBI) and the microRNA (miRNA) expression profile of PubMed identifier (PMID) 26890143 were integrated. De novo motifs were used to obtain predicted transcription factor (TF) motifs for differentially expressed genes. The regulatory effect of resveratrol on key nodes in the comprehensive analysis results was verified. The TF–miRNA–mRNA interaction network based on the STITCH and miRDB databases showed that resveratrol exerted a dual inhibitory effect by activating inhibitory TFs to block the cell cycle and inhibit miRNAs from upregulating apoptosis. However, these two processes did not work completely independently. TP53 is the dominant hub gene associated with the cell cycle and apoptosis throughout the TF–miRNA network. Kaplan–Meier plotter analysis found that resveratrol‐induced expression changes in key RNAs, such as E2F2, JUN, FOS, BRCA1, CDK1, CDKN1A, TNF, and hsa‐miR‐34a‐5p, significantly improved the prognosis of breast cancer patients, which was further verified using real‐time quantitative PCR (qPCR) and western blotting. This study constructed a TF–miRNA regulatory network with TP53 and E2F as the main central genes to elucidate the molecular mechanism of resveratrol in the treatment of breast cancer.
Collapse
Affiliation(s)
- Yongfeng Zhou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| | - Jing Zhang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| | - Daoyu Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| | - Zhengzhu Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| | - Yanhui Zhai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
| |
Collapse
|
10
|
Adams AC, Macy AM, Kang P, Castro-Ochoa KF, Wijeratne EMK, Xu YM, Liu MX, Charos A, Bosenberg MW, Gunatilaka AAL, Sertil AR, Hastings KT. Physachenolide C induces complete regression of established murine melanoma tumors via apoptosis and cell cycle arrest. Transl Oncol 2021; 15:101259. [PMID: 34735896 PMCID: PMC8571524 DOI: 10.1016/j.tranon.2021.101259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/10/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
PCC and LG-134 had direct cytotoxicity in murine melanoma cell lines (IC50 values ranged from 0.19–1.8 µM). PCC treatment induced apoptosis of tumor cells both in vitro and in vivo. PCC treatment induced G0-G1 cell cycle arrest of melanoma cells. PCC treatment caused complete regression of established melanoma tumors in all mice. 17β-hydroxywithanolides have the potential to improve melanoma therapeutic outcome.
Melanoma is an aggressive skin cancer that metastasizes to other organs. While immune checkpoint blockade with anti-PD-1 has transformed the treatment of advanced melanoma, many melanoma patients fail to respond to anti-PD-1 therapy or develop acquired resistance. Thus, effective treatment of melanoma still represents an unmet clinical need. Our prior studies support the anti-cancer activity of the 17β-hydroxywithanolide class of natural products, including physachenolide C (PCC). As single agents, PCC and its semi-synthetic analog demonstrated direct cytotoxicity in a panel of murine melanoma cell lines, which share common driver mutations with human melanoma; the IC50 values ranged from 0.19–1.8 µM. PCC treatment induced apoptosis of tumor cells both in vitro and in vivo. In vivo treatment with PCC alone caused the complete regression of established melanoma tumors in all mice, with a durable response in 33% of mice after discontinuation of treatment. T cell-mediated immunity did not contribute to the therapeutic efficacy of PCC or prevent tumor recurrence in YUMM2.1 melanoma model. In addition to apoptosis, PCC treatment induced G0-G1 cell cycle arrest of melanoma cells, which upon removal of PCC, re-entered the cell cycle. PCC-induced cycle cell arrest likely contributed to the in vivo tumor recurrence in a portion of mice after discontinuation of treatment. Thus, 17β-hydroxywithanolides have the potential to improve the therapeutic outcome for patients with advanced melanoma.
Collapse
Affiliation(s)
- Anngela C Adams
- University of Arizona College of Medicine Phoenix, 425 N. 5th St., Phoenix, AZ 85004, United States.
| | - Anne M Macy
- University of Arizona College of Medicine Phoenix, 425 N. 5th St., Phoenix, AZ 85004, United States.
| | - Paul Kang
- Mel and Enid Zuckerman College of Public Health, 714 E. Van Buren St., Phoenix, AZ 85006, United States.
| | - Karla F Castro-Ochoa
- University of Arizona College of Medicine Phoenix, 425 N. 5th St., Phoenix, AZ 85004, United States.
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Rd., Tucson, AZ 85706, United States.
| | - Ya-Ming Xu
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Rd., Tucson, AZ 85706, United States.
| | - Manping X Liu
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Rd., Tucson, AZ 85706, United States.
| | - Alexandra Charos
- Department of Dermatology, Yale University, 333 Cedar St., New Haven, CT 06520, United States.
| | - Marcus W Bosenberg
- Department of Dermatology, Yale University, 333 Cedar St., New Haven, CT 06520, United States.
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, University of Arizona, 250 E. Valencia Rd., Tucson, AZ 85706, United States; University of Arizona Cancer Center, University of Arizona, 1515 N. Campbell Ave., Tucson, AZ 85724, United States.
| | - Aparna R Sertil
- University of Arizona College of Medicine Phoenix, 425 N. 5th St., Phoenix, AZ 85004, United States.
| | - K Taraszka Hastings
- University of Arizona College of Medicine Phoenix, 425 N. 5th St., Phoenix, AZ 85004, United States; University of Arizona Cancer Center, University of Arizona, 1515 N. Campbell Ave., Tucson, AZ 85724, United States.
| |
Collapse
|
11
|
Shi C, Jug R, Bean SM, Jeck WR, Guy CD. Primary hepatic neoplasms arising in cirrhotic livers can have a variable spectrum of neuroendocrine differentiation. Hum Pathol 2021; 116:63-72. [PMID: 34310982 DOI: 10.1016/j.humpath.2021.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/25/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022]
Abstract
Primary hepatic neoplasms with neuroendocrine differentiation are extremely rare. Their clinicopathological features and molecular genetic basis are largely unknown. We identified four cases of primary hepatic neoplasms with neuroendocrine differentiation. Electronic medical records were reviewed for clinical history, imaging findings, laboratory results, and follow-up. Pathology slides, immunohistochemistry, and ancillary studies were reviewed. There were two females and two males with age ranging from 52 to 74 years. There was one amphicrine carcinoma with tumor cells simultaneously demonstrating both hepatocellular and neuroendocrine differentiation, one mixed hepatocellular-neuroendocrine carcinoma (NEC) with hepatocellular component intermingled with neuroendocrine component, one small cell NEC, and one well-differentiated neuroendocrine tumor. Next- generation sequencing of the mixed hepatocellular-NEC and small cell NEC showed molecular/genetic alterations commonly seen in hepatocellular carcinoma (HCC). All four cases arose in a background of cirrhosis. Primary hepatic neoplasms arising in cirrhotic livers can have a spectrum of neuroendocrine differentiation. Presence of a NEC component may be an indicator of aggressiveness. In addition, primary hepatic carcinomas with neuroendocrine differentiation likely share the same molecular pathways as HCC.
Collapse
Affiliation(s)
- Chanjuan Shi
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Rachel Jug
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sarah M Bean
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - William R Jeck
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
12
|
Choi HI, An GY, Baek M, Yoo E, Chai JC, Lee YS, Jung KH, Chai YG. BET inhibitor suppresses migration of human hepatocellular carcinoma by inhibiting SMARCA4. Sci Rep 2021; 11:11799. [PMID: 34083693 PMCID: PMC8175750 DOI: 10.1038/s41598-021-91284-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and poorly responsive cancers worldwide. Bromodomain and extraterminal (BET) inhibitors, such as JQ1 and OTX-015, inhibit BET protein binding to acetylated residues in histones. However, the physiological mechanisms and regulatory processes of BET inhibition in HCC remain unclear. To explore BET inhibitors’ potential role in the molecular mechanisms underlying their anticancer effects in HCC, we analyzed BET inhibitor-treated HCC cells’ gene expression profiles with RNA-seq and bioinformatics analysis. BET inhibitor treatment significantly downregulated genes related to bromodomain-containing proteins 4 (BRD4), such as ACSL5, SLC38A5, and ICAM2. Importantly, some cell migration-related genes, including AOC3, CCR6, SSTR5, and SCL7A11, were significantly downregulated. Additionally, bioinformatics analysis using Ingenuity Knowledge Base Ingenuity Pathway Analysis (IPA) revealed that SMARCA4 regulated migration response molecules. Furthermore, knockdown of SMARCA4 gene expression by siRNA treatment significantly reduced cell migration and the expression of migration-related genes. In summary, our results indicated that BET inhibitor treatment in HCC cell lines reduces cell migration through the downregulation of SMARCA4.
Collapse
Affiliation(s)
- Hae In Choi
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Ga Yeong An
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Mina Baek
- Institute of Natural Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea.,Department of Molecular and Life Science, Hanyang University, Ansan, 15588, Republic of Korea
| | - Eunyoung Yoo
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Jin Choul Chai
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Seek Lee
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Kyoung Hwa Jung
- Convergence Technology Campus of Korea Polytechnic II, Incheon, 21417, Republic of Korea. .,Department of Biopharmaceutical System, Gwangmyeong Convergence Technology Campus of Korea Polytechnic II, Gwangmyeong , 14222, Republic of Korea.
| | - Young Gyu Chai
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea. .,Department of Molecular and Life Science, Hanyang University, Ansan, 15588, Republic of Korea.
| |
Collapse
|
13
|
Abstract
E2F transcription factor 2 (E2F2) is a member of the E2F family of transcription factors. The classical view is that some E2Fs act as "activators" and others "inhibitors" of cell cycle gene expression. However, the so-called "activator" E2F2 is particularly enigmatic, with seemingly contradictory roles in the cell cycle, proliferation, apoptosis, inflammation, and cell migration and invasion. How can we rationalize the apparently opposing functions of E2F2 in different situations? This is difficult because different methods of studying E2F2 have yielded conflicting results, so extrapolating mechanisms from an observed endpoint is challenging. This review will attempt to summarize and clarify these issues. This review focuses on genetic studies that have helped elucidate the biological functions of E2F2 and that have enhanced our understanding of how E2F2 is integrated into pathways controlling the cell cycle, proliferation, apoptosis, inflammation, and cell migration and invasion. This review will also discuss the function of E2F2 in cancer and other diseases. This review provides a strong basis for further research on the biological function and clinical potential of E2F2.
Collapse
Affiliation(s)
- Luwen Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Shiguan Wang
- Medical College, Shandong University, Ji'nan, China
| | - Yihang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| |
Collapse
|
14
|
Circular RNA circE2F2 promotes malignant progression of ovarian cancer cells by upregulating the expression of E2F2 protein via binding to HuR protein. Cell Signal 2021; 84:110014. [PMID: 33894314 DOI: 10.1016/j.cellsig.2021.110014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/20/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Ovarian cancer (OC) is a gynecological malignancy with a poor prognosis and low survival rate. E2F2 is a transcription activator that plays an indispensable role in cell proliferation and cell cycle progression. The preliminary analysis indicated that the E2F2 gene could produce three circular RNAs (circRNAs). This study aimed to investigate whether these circRNAs would be involved in OC tumorigenesis. The results showed that one of the circRNAs (termed circE2F2) was significantly upregulated in OC tissues and cell lines, and high circE2F2 expression was associated with poor survival in OC patients. The knockdown of circE2F2 in OC cells suppressed cell proliferation, migration, invasion, and cellular glucose metabolism. In circE2F2-deficient cells, the half-life of the E2F2 mRNA was significantly shorter than that in the control group, indicating that sufficient circE2F2 expression could strengthen the stability of the E2F2 mRNA. Further analysis revealed that circE2F2 could bind to RNA-binding protein Hu antigen R (HuR). Moreover, circE2F2 enhanced the stability of the E2F2 mRNA via binding to the HuR protein. Also, E2F2 overexpression significantly enhanced the mobility, invasiveness, and glucose metabolism of OC cells with insufficient circE2F2 expression, suggesting that circE2F2 induced OC cell growth and metastasis by upregulating E2F2. In conclusion, circE2F2 promoted OC cell proliferation, metastasis, and glucose metabolism by stabilizing the E2F2 mRNA via binding to the HuR protein. These findings suggest a novel regulatory mechanism for the oncogenic effects of circE2F2, E2F2, and HuR on ovarian carcinogenesis.
Collapse
|
15
|
González-Romero F, Mestre D, Aurrekoetxea I, O'Rourke CJ, Andersen JB, Woodhoo A, Tamayo-Caro M, Varela-Rey M, Palomo-Irigoyen M, Gómez-Santos B, de Urturi DS, Núñez-García M, García-Rodríguez JL, Fernández-Ares L, Buqué X, Iglesias-Ara A, Bernales I, De Juan VG, Delgado TC, Goikoetxea-Usandizaga N, Lee R, Bhanot S, Delgado I, Perugorria MJ, Errazti G, Mosteiro L, Gaztambide S, Martinez de la Piscina I, Iruzubieta P, Crespo J, Banales JM, Martínez-Chantar ML, Castaño L, Zubiaga AM, Aspichueta P. E2F1 and E2F2-Mediated Repression of CPT2 Establishes a Lipid-Rich Tumor-Promoting Environment. Cancer Res 2021; 81:2874-2887. [PMID: 33771899 DOI: 10.1158/0008-5472.can-20-2052] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/15/2020] [Accepted: 03/24/2021] [Indexed: 11/16/2022]
Abstract
Lipid metabolism rearrangements in nonalcoholic fatty liver disease (NAFLD) contribute to disease progression. NAFLD has emerged as a major risk for hepatocellular carcinoma (HCC), where metabolic reprogramming is a hallmark. Identification of metabolic drivers might reveal therapeutic targets to improve HCC treatment. Here, we investigated the contribution of transcription factors E2F1 and E2F2 to NAFLD-related HCC and their involvement in metabolic rewiring during disease progression. In mice receiving a high-fat diet (HFD) and diethylnitrosamine (DEN) administration, E2f1 and E2f2 expressions were increased in NAFLD-related HCC. In human NAFLD, E2F1 and E2F2 levels were also increased and positively correlated. E2f1 -/- and E2f2 -/- mice were resistant to DEN-HFD-induced hepatocarcinogenesis and associated lipid accumulation. Administration of DEN-HFD in E2f1 -/- and E2f2 -/- mice enhanced fatty acid oxidation (FAO) and increased expression of Cpt2, an enzyme essential for FAO, whose downregulation is linked to NAFLD-related hepatocarcinogenesis. These results were recapitulated following E2f2 knockdown in liver, and overexpression of E2f2 elicited opposing effects. E2F2 binding to the Cpt2 promoter was enhanced in DEN-HFD-administered mouse livers compared with controls, implying a direct role for E2F2 in transcriptional repression. In human HCC, E2F1 and E2F2 expressions inversely correlated with CPT2 expression. Collectively, these results indicate that activation of the E2F1-E2F2-CPT2 axis provides a lipid-rich environment required for hepatocarcinogenesis. SIGNIFICANCE: These findings identify E2F1 and E2F2 transcription factors as metabolic drivers of hepatocellular carcinoma, where deletion of just one is sufficient to prevent disease. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2874/F1.large.jpg.
Collapse
Affiliation(s)
- Francisco González-Romero
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Daniela Mestre
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain.,BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Igor Aurrekoetxea
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain.,BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Colm J O'Rourke
- Department of Health and Medical Sciences, Biotech Research and Innovation Center, University of Copenhagen, Denmark
| | - Jesper B Andersen
- Department of Health and Medical Sciences, Biotech Research and Innovation Center, University of Copenhagen, Denmark
| | - Ashwin Woodhoo
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Derio, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Miguel Tamayo-Caro
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Derio, Spain
| | - Marta Varela-Rey
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), CIBERehd Derio, Spain
| | | | - Beatriz Gómez-Santos
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Diego Sáenz de Urturi
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Maitane Núñez-García
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Juan L García-Rodríguez
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Larraitz Fernández-Ares
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain.,BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Xabier Buqué
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain.,BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Ainhoa Iglesias-Ara
- Department of Genetic, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of Basque Country UPV/EHU, Leioa, Spain
| | | | - Virginia Gutierrez De Juan
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), CIBERehd Derio, Spain
| | - Teresa C Delgado
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), CIBERehd Derio, Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), CIBERehd Derio, Spain
| | | | | | - Igotz Delgado
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country UPV/EHU, CIBERehd, San Sebastian, Spain.,Department of Medicine, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Gaizka Errazti
- BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Lorena Mosteiro
- BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| | - Sonia Gaztambide
- BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain.,Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, CIBERDEM, CIBERER, Leioa, Spain
| | | | - Paula Iruzubieta
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Santander, Spain
| | - Jesus M Banales
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.,Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country UPV/EHU, CIBERehd, San Sebastian, Spain
| | - Maria L Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), CIBERehd Derio, Spain
| | - Luis Castaño
- BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain.,Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, CIBERDEM, CIBERER, Leioa, Spain
| | - Ana M Zubiaga
- Department of Genetic, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country UPV/EHU, Leioa, Spain. .,BioCruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERehd, Barakaldo, Spain
| |
Collapse
|
16
|
Yuan S, He G, Li L. Hsa-miR-155 regulates the cell cycle and barrier function of corneal endothelial cells through E2F2. Am J Transl Res 2021; 13:1505-1515. [PMID: 33841674 PMCID: PMC8014374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
This study was aimed to determine the role of has-miR-155 and E2F2 on corneal endothelial cells. Real-time quantitative PCR and Western blot assays were carried out to determine the levels of has-miR-155 and E2F2, and Flow cytometry assay was conducted to detect cell cycle. In addition, Targetscan7.2 was adopted to analyze the internal connection between hsa-miR-155 and E2F2, and a dual luciferase reporter gene assay to determine predicted site between has-miR-155 and E2F2. Increased hsa-miR-155 resulted in decreased E2F2, while decreased hsa-miR-155 increased the level of E2F2. In addition, both increased hsa-miR-155 and decreased E2F2 led to an increase in S-phase cells and a decrease in G1-phase cells. Also, they induced an increase in the activity of barrier-related proteins MLCK and ZO-1, an up-regulation of Cyclin D1 and Cyclin E1, and a down-regulation of apoptosis proteins (Caspase 3/Bax/Bim/Bid) whereas decreased hsa-miR-155 led to an opposite change in cells, and decreased E2F2 could offset cell changes caused by increased has-miR-155. In conclusion, Has-miR-155 regulates the cell cycle of corneal endothelial cells and improves their barrier function by down regulating E2F2.
Collapse
Affiliation(s)
- Shuyi Yuan
- Department of Optometry Center, Tianjin Eye HospitalTianjin 300022, China
| | - Guanghui He
- Department of Vitreoretinopathy, Tianjin Eye HospitalTianjin 300022, China
| | - Lihua Li
- Department of Optometry Center, Tianjin Eye HospitalTianjin 300022, China
| |
Collapse
|
17
|
Liu Y, Xue M, Cao D, Qin L, Wang Y, Miao Z, Wang P, Hu X, Shen J, Xiong B. Multi-omics characterization of WNT pathway reactivation to ameliorate BET inhibitor resistance in liver cancer cells. Genomics 2021; 113:1057-1069. [PMID: 33667649 DOI: 10.1016/j.ygeno.2021.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/17/2021] [Accepted: 02/28/2021] [Indexed: 01/10/2023]
Abstract
The Bromodomain and Extra-terminal domain (BET) proteins are promising targets in treating cancers. Although BET inhibitors have been in clinical trials, they are limited by lacking of suitable biomarkers to indicate drug responses in different cancers. Here we identify DHRS2, ETV4 and NOTUM as potential biomarkers to indicate drug resistance in liver cancer cells of a recently discovered BET inhibitor, Hjp-6-171. Furthermore, we confirm that reactivation of WNT pathway, the target of NOTUM, contributes to the drug sensitivity restoration in Hjp-6-171 resistant cells. Specially, combinations of Hjp-6-171 and a GSK3β inhibitor CHIR-98014 show remarkable therapeutic effects in vitro and in vivo. Integrating RNA-seq and ChIP-seq data, we reveal the expression signature of β-catenin regulated genes is contrary in sensitive cells to that in resistant cells. We propose WNT signaling molecules such as β-catenin and ETV4 to be candidate biomarkers to indicate BET inhibitor responses in liver cancer patients.
Collapse
Affiliation(s)
- Yuwei Liu
- SARI center for stem cell and nanomedicine, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mengzhu Xue
- SARI center for stem cell and nanomedicine, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Danyan Cao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lihuai Qin
- Center for chemical biology and drug discovery, department of pharmacological sciences, Tisch cancer institute, Icahn School of medicine at Mount Sinai, New York 10029, USA
| | - Ying Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zehong Miao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Peng Wang
- Bio-Med Big Data Center, Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Xin Hu
- Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Jingkang Shen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Xiong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
18
|
Jühling F, Hamdane N, Crouchet E, Li S, El Saghire H, Mukherji A, Fujiwara N, Oudot MA, Thumann C, Saviano A, Roca Suarez AA, Goto K, Masia R, Sojoodi M, Arora G, Aikata H, Ono A, Tabrizian P, Schwartz M, Polyak SJ, Davidson I, Schmidl C, Bock C, Schuster C, Chayama K, Pessaux P, Tanabe KK, Hoshida Y, Zeisel MB, Duong FHT, Fuchs BC, Baumert TF. Targeting clinical epigenetic reprogramming for chemoprevention of metabolic and viral hepatocellular carcinoma. Gut 2021; 70:157-169. [PMID: 32217639 PMCID: PMC7116473 DOI: 10.1136/gutjnl-2019-318918] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related mortality with chronic viral hepatitis and non-alcoholic steatohepatitis (NASH) as major aetiologies. Treatment options for HCC are unsatisfactory and chemopreventive approaches are absent. Chronic hepatitis C (CHC) results in epigenetic alterations driving HCC risk and persisting following cure. Here, we aimed to investigate epigenetic modifications as targets for liver cancer chemoprevention. DESIGN Liver tissues from patients with NASH and CHC were analysed by ChIP-Seq (H3K27ac) and RNA-Seq. The liver disease-specific epigenetic and transcriptional reprogramming in patients was modelled in a liver cell culture system. Perturbation studies combined with a targeted small molecule screen followed by in vivo and ex vivo validation were used to identify chromatin modifiers and readers for HCC chemoprevention. RESULTS In patients, CHC and NASH share similar epigenetic and transcriptomic modifications driving cancer risk. Using a cell-based system modelling epigenetic modifications in patients, we identified chromatin readers as targets to revert liver gene transcription driving clinical HCC risk. Proof-of-concept studies in a NASH-HCC mouse model showed that the pharmacological inhibition of chromatin reader bromodomain 4 inhibited liver disease progression and hepatocarcinogenesis by restoring transcriptional reprogramming of the genes that were epigenetically altered in patients. CONCLUSION Our results unravel the functional relevance of metabolic and virus-induced epigenetic alterations for pathogenesis of HCC development and identify chromatin readers as targets for chemoprevention in patients with chronic liver diseases.
Collapse
Affiliation(s)
- Frank Jühling
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Nourdine Hamdane
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Emilie Crouchet
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Shen Li
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Houssein El Saghire
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Atish Mukherji
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Marine A Oudot
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Christine Thumann
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Antonio Saviano
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Armando Andres Roca Suarez
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Kaku Goto
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Ricard Masia
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mozhdeh Sojoodi
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gunisha Arora
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsushi Ono
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Parissa Tabrizian
- Recanati/Miller Transplantation Institute, Mount Sinai Medical Center, New York, New York, USA
| | - Myron Schwartz
- Recanati/Miller Transplantation Institute, Mount Sinai Medical Center, New York, New York, USA
| | - Stephen J Polyak
- Department of Global Health, University of Washington, Seattle, Washington, USA,Department of Laboratory Medicine, University of Washington, Seattle, Washington, USA
| | - Irwin Davidson
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, Illkirch, France
| | - Christian Schmidl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria,Regensburg Centre for Interventional Immunology (RCI), Regensburg, Germany
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Catherine Schuster
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Patrick Pessaux
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Kenneth K Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mirjam B Zeisel
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | - François HT Duong
- Université de Strasbourg, Strasbourg, France,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
| | - Bryan C Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas F Baumert
- Université de Strasbourg, Strasbourg, France .,Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France.,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
19
|
Zeng Z, Cao Z, Tang Y. Increased E2F2 predicts poor prognosis in patients with HCC based on TCGA data. BMC Cancer 2020; 20:1037. [PMID: 33115417 PMCID: PMC7594443 DOI: 10.1186/s12885-020-07529-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Background The E2F family of transcription factor 2 (E2F2) plays an important role in the development and progression of various tumors, but its association with hepatocellular carcinoma (HCC) remains unknown. Our study aimed to investigate the role and clinical significance of E2F2 in HCC. Methods HCC raw data were extracted from The Cancer Genome Atlas (TCGA). Wilcoxon signed-rank test, Kruskal-Wallis test and logistic regression were applied to analyze the relationship between the expression of E2F2 and clinicopathologic characteristics. Cox regression and Kaplan-Meier were employed to evaluate the correlation between clinicopathologic features and survival. The biological function of E2F2 was annotated by Gene Set Enrichment Analysis (GSEA). Results The expression of E2F2 was increased in HCC samples. The expression of elevated E2F2 in HCC samples was prominently correlated with histologic grade (OR = 2.62 for G3–4 vs. G1–2, p = 1.80E-05), clinical stage (OR = 1.74 for III-IV vs. I-II, p = 0.03), T (OR = 1.64 for T3–4 vs.T1–2, p = 0.04), tumor status (OR = 1.88 for with tumor vs. tumor free, p = 3.79E-03), plasma alpha fetoprotein (AFP) value (OR = 3.18 for AFP ≥ 400 vs AFP<20, p = 2.16E-04; OR = 2.50 for 20 ≤ AFP<400 vs AFP<20, p = 2.56E-03). Increased E2F2 had an unfavorable OS (p = 7.468e− 05), PFI (p = 3.183e− 05), DFI (p = 0.001), DSS (p = 4.172e− 05). Elevated E2F2 was independently bound up with OS (p = 0.004, hazard ratio [HR] = 2.4 (95% CI [1.3–4.2])), DFI (P = 0.029, hazard ratio [HR] = 2.0 (95% CI [1.1–3.7])) and PFI (P = 0.005, hazard ratio [HR] = 2.2 (95% CI [1.3–3.9])). GSEA disclosed that cell circle, RNA degradation, pyrimidine metabolism, base excision repair, aminoacyl tRNA biosynthesis, DNA replication, p53 signaling pathway, nucleotide excision repair, ubiquitin-mediated proteolysis, citrate cycle TCA cycle were notably enriched in E2F2 high expression phenotype. Conclusions Elevated E2F2 can be a promising independent prognostic biomarker and therapeutic target for HCC. Additionally, cell cycle, pyrimidine metabolism, DNA replication, p53 signaling pathway, ubiquitin-mediated proteolysis, the citrate cycle TCA cycle may be the key pathway by which E2F2 participates in the initial and progression of HCC.
Collapse
Affiliation(s)
- Zhili Zeng
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Zebiao Cao
- Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Ying Tang
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, no.12, Airport Road, Sanyuanli Street, Baiyun District, Guangzhou, 510405, Guangdong, China.
| |
Collapse
|
20
|
BRD4/8/9 are prognostic biomarkers and associated with immune infiltrates in hepatocellular carcinoma. Aging (Albany NY) 2020; 12:17541-17567. [PMID: 32927435 PMCID: PMC7521508 DOI: 10.18632/aging.103768] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Bromodomain (BRD)-containing proteins are a class of epigenetic readers with unique recognition for N-acetyl-lysine in histones and functions of gene transcription and chromatin modification, known to be critical in various cancers. However, little is known about the roles of distinct BRD-containing protein genes in hepatocellular carcinoma (HCC). Most recently, we investigated the transcriptional and survival data of BRD1, BRD2, BRD3, BRD4, BRD7, BRD8, BRD9 in HCC patients through ONCOMINE, UALCAN, Human Protein Atlas, GEPIA, cBioPortal, STRING, TIMER databases. BRD1/2/3/4/7/8/9 were over-expressed in HCC and were significantly associated with clinical cancer stages and pathological tumor grades. High mRNA expressions of BRD4/8/9 were promising candidate biomarkers in HCC patients. The rate of sequence alternations in BRD1/2/3/4/7/8/9 was relatively high (52%) in HCC patients, and the genetic alternations were correlated with shorter overall survival and disease-free survival in HCC patients. Additionally, the mRNA expression levels of individual BRD genes were significantly positively associated with the immune infiltrating levels of B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells. And the associations between BRD1/2/3/4/7/8/9 and diverse immune marker sets showed a significance. Overall, these results indicated that BRD4/8/9 could be potential prognostic markers and druggable epigenetic targets in HCC patients.
Collapse
|
21
|
Camero S, Camicia L, Marampon F, Ceccarelli S, Shukla R, Mannarino O, Pizer B, Schiavetti A, Pizzuti A, Tombolini V, Marchese C, Dominici C, Megiorni F. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett 2020; 479:71-88. [PMID: 32200036 DOI: 10.1016/j.canlet.2020.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The antitumour effects of OTX015, a first-in-class BET inhibitor (BETi), were investigated as a single agent or in combination with ionizing radiation (IR) in preclinical in vitro models of rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma. Herein, we demonstrated the upregulation of BET Bromodomain gene expression in RMS tumour biopsies and cell lines compared to normal skeletal muscle. In vitro experiments showed that OTX015 significantly reduced RMS cell proliferation by altering cell cycle modulators and apoptotic related proteins due to the accumulation of DNA breaks that cells are unable to repair. Interestingly, OTX015 also impaired migration capacity and tumour-sphere architecture by downregulating pro-stemness genes and was able to potentiate ionizing radiation effects by reducing the expression of different drivers of tumour dissemination and resistance mechanisms, including the GNL3 gene, that we correlated for the first time with the RMS phenotype. In conclusion, our research sheds further light on the molecular events of OTX015 action against RMS cells and indicates this novel BETi as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with RMS.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Lucrezia Camicia
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Olga Mannarino
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP, UK.
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Antonio Pizzuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Carlo Dominici
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
22
|
Sun X, Ge X, Xu Z, Chen D. Identification of circular RNA-microRNA-messenger RNA regulatory network in hepatocellular carcinoma by integrated analysis. J Gastroenterol Hepatol 2020; 35:157-164. [PMID: 31222831 DOI: 10.1111/jgh.14762] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/03/2019] [Accepted: 06/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) is the most common types of hepatic malignancies. This study aimed to better understand the pathogenesis of HCC and may help facilitate the improvement of the diagnostic of HCC. METHODS The mRNA and miRNA expression profiles of HCC, which was retrieved from The Cancer Genome Atlas database, and the circRNA expression profiles of HCC, which was retrieved from Gene Expression Omnibus database, were included in this study to perform an integrated analysis. The differentially expressed mRNAs (DEmRNAs), differentially expressed miRNAs (DEmiRNAs), and differentially expressed circRNAs (DEcircRNAs) were identified, and competing endogenous RNA (ceRNA) (DEcircRNA-DEmiRNA-DEmRNA) regulatory network was conducted. Functional annotation of host gene of DEcircRNAs and DEmRNAs in ceRNA regulatory network was performed. Quantitative real-time polymerase chain reaction validation of the expression of the selected DEmRNAs, DEmiRNAs, and DEcircRNAs was performed. RESULTS A total of 2982 DEmRNAs, 144 DEmiRNAs, and 264 DEcircRNAs were obtained. The ceRNA network contained 61 circRNA-miRNA pairs and 1149 miRNA-mRNA pairs, including 48 circRNAs, 30 miRNAs, and 1149 mRNAs. Functional annotation of DEmRNAs in ceRNA regulatory network revealed that these DEmRNAs were significantly enriched in tryptophan metabolism, fatty acid metabolism, and pathways in cancer. Except for ARNT2 and hsa-miR-214-3p, expression of the others in the quantitative real-time polymerase chain reaction results was consistent with that in our integrated analysis, generally. CONCLUSION We speculate that hsa_circRNA_104268/hsa-miR-214-3p/E2F2, hsa_circRNA_104168/hsa-miR-139-5p/HRAS, and hsa_circRNA_104769/hsa-miR-93-5p/JUN interaction pairs may play a vital role in HCC. This study expected to provide a novel insight into the pathogenesis and therapy of HCC from the circRNA-miRNA-mRNA network view.
Collapse
Affiliation(s)
- Xiangjun Sun
- Department of Hepatobiliary Surgery, Linyi People's Hospital, Linyi, China
| | - Xinfeng Ge
- Department of General Surgery, The People's Hospital of Linshu, Linshu County, China
| | - Zhiyong Xu
- Department of General Surgery, The People's Hospital of Linshu, Linshu County, China
| | - Dongfeng Chen
- Department of General Surgery, Linyi People's Hospital, Linyi, China
| |
Collapse
|
23
|
Murakami S, Li R, Nagari A, Chae M, Camacho CV, Kraus WL. Distinct Roles for BET Family Members in Estrogen Receptor α Enhancer Function and Gene Regulation in Breast Cancer Cells. Mol Cancer Res 2019; 17:2356-2368. [PMID: 31551256 DOI: 10.1158/1541-7786.mcr-19-0393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/28/2019] [Accepted: 09/17/2019] [Indexed: 01/23/2023]
Abstract
The bromodomain family member proteins (BRD; BET proteins) are key coregulators for estrogen receptor alpha (ERα)-mediated transcriptional enhancers. The use of BRD-selective inhibitors has gained much attention as a potential treatment for various solid tumors, including ER-positive breast cancers. However, the roles of individual BET family members have largely remained unexplored. Here, we describe the role of BRDs in estrogen (E2)-dependent gene expression in ERα-positive breast cancer cells. We observed that chemical inhibition of BET family proteins with JQ1 impairs E2-regulated gene expression and growth in breast cancer cells. In addition, RNAi-mediated depletion of each BET family member (BRDs 2, 3, and 4) revealed partially redundant roles at ERα enhancers and for target gene transcription. Furthermore, we found a unique role of BRD3 as a molecular sensor of total BET family protein levels and activity through compensatory control of its own protein levels. Finally, we observed that BRD3 is recruited to a subset of ERα-binding sites (ERBS) that are enriched for active enhancer features, located in clusters of ERBSs likely functioning as "super enhancers," and associated with highly E2-responsive genes. Collectively, our results illustrate a critical and specific role for BET family members in ERα-dependent gene transcription. IMPLICATIONS: BRD3 is recruited to and controls the activity of a subset ERα transcriptional enhancers, providing a therapeutic opportunity to target BRD3 with BET inhibitors in ERα-positive breast cancers.
Collapse
Affiliation(s)
- Shino Murakami
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas.,Program in Genetics, Development and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rui Li
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas.,Program in Genetics, Development and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Anusha Nagari
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Minho Chae
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cristel V Camacho
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas.,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - W Lee Kraus
- The Laboratory of Signaling and Gene Expression, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas. .,The Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas.,Program in Genetics, Development and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
24
|
Hung YH, Hsu MC, Chen LT, Hung WC, Pan MR. Alteration of Epigenetic Modifiers in Pancreatic Cancer and Its Clinical Implication. J Clin Med 2019; 8:jcm8060903. [PMID: 31238554 PMCID: PMC6617267 DOI: 10.3390/jcm8060903] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022] Open
Abstract
The incidence of pancreatic cancer has considerably increased in the past decade. Pancreatic cancer has the worst prognosis among the cancers of the digestive tract because the pancreas is located in the posterior abdominal cavity, and most patients do not show clinical symptoms for early detection. Approximately 55% of all patients are diagnosed with pancreatic cancer only after the tumors metastasize. Therefore, identifying useful biomarkers for early diagnosis and screening high-risk groups are important to improve pancreatic cancer therapy. Recent emerging evidence has suggested that genetic and epigenetic alterations play a crucial role in the molecular aspects of pancreatic tumorigenesis. Here, we summarize recent progress in our understanding of the epigenetic alterations in pancreatic cancer and propose potential synthetic lethal strategies to target these genetic defects to treat this deadly disease.
Collapse
Affiliation(s)
- Yu-Hsuan Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Ming-Chuan Hsu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan.
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Mei-Ren Pan
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| |
Collapse
|
25
|
Tsang FHC, Law CT, Tang TCC, Cheng CLH, Chin DWC, Tam WSV, Wei L, Wong CCL, Ng IOL, Wong CM. Aberrant Super-Enhancer Landscape in Human Hepatocellular Carcinoma. Hepatology 2019; 69:2502-2517. [PMID: 30723918 DOI: 10.1002/hep.30544] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 01/18/2019] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) cells exploit an aberrant transcriptional program to sustain their infinite growth and progression. Emerging evidence indicates that the continuous and robust transcription of oncogenes in cancer cells is often driven by super-enhancers (SEs). In this study, we systematically compared the SE landscapes between normal liver and HCC cells and revealed that the cis-acting SE landscape was extensively reprogrammed during liver carcinogenesis. HCC cells acquired SEs at multiple prominent oncogenes to drive their vigorous expression. We identified sphingosine kinase 1 (SPHK1) as an SE-associated oncogene, and we used this gene as an example to illustrate the impact of SEs on the activation of oncogenes in HCC. Concurrently, we also showed that the critical components of the trans-acting SE complex, namely, cyclin-dependent kinase 7 (CDK7), bromodomain-containing protein 4 (BRD4), E1A binding protein P300 (EP300), and mediator complex subunit 1 (MED1), were frequently overexpressed in human HCCs and were associated with the poor prognosis of patients with HCC. Using the CRISPR/Cas9 gene-editing system and specific small-molecule inhibitors, we further demonstrated that HCC cells were highly sensitive to perturbations of the SE complex. The inactivation of CDK7, BRD4, EP300, and MED1 selectively repressed the expression of SE-associated oncogenes in HCC. Finally, we demonstrated that THZ1, which is a small-molecule inhibitor of CDK7, exerted a prominent anticancer effect in both in vitro and in vivo HCC models. Conclusion: The SE landscape and machinery were significantly altered in human HCCs. HCC cells are highly susceptible to perturbations of the SE complex due to the resulting selective suppression of SE-associated oncogenes. Our results suggest that targeting SE complex is a promising therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Felice Ho-Ching Tsang
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cheuk-Ting Law
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tsz-Ching Chloe Tang
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carol Lai-Hung Cheng
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Don Wai-Ching Chin
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wing-Sum Vincy Tam
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lai Wei
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Carmen Chak-Lui Wong
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-Lin Ng
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chun-Ming Wong
- State Key Laboratory of Liver Research (The University of Hong Kong) and Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Hong SH, You JS. SOX9 is controlled by the BRD4 inhibitor JQ1 via multiple regulation mechanisms. Biochem Biophys Res Commun 2019; 511:746-752. [PMID: 30833074 DOI: 10.1016/j.bbrc.2019.02.135] [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: 02/19/2019] [Accepted: 02/25/2019] [Indexed: 01/10/2023]
Abstract
SOX9 is a key transcription factor during cell differentiation, sex determination, and tumorigenesis. However, the detailed mechanisms of its targeting strategy remain elusive. To investigate possibilities of targeting SOX9 with epigenetic drugs and the precise underlying mechanisms, two human cancer cell lines were chosen as model systems, which showed high SOX9 expression and anti-tumorigenic effects upon loss of SOX9. Histone acetylation-related screening of a small panel of epigenetic drugs revealed that the bromodomain reader inhibitor JQ1 dramatically downregulated SOX9 through multiple regulation steps, namely, transcription, BRD4-SOX9 protein-protein interaction, and further protein stability. These findings suggest that BRD4 inhibition is a novel therapeutic strategy for diseases characterized by SOX9 overexpression.
Collapse
Affiliation(s)
- Seong Hwi Hong
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 05029, South Korea
| | - Jueng Soo You
- Department of Biochemistry, School of Medicine, Konkuk University, Seoul, 05029, South Korea; Research Institute of Medical Science, Konkuk University School of Medicine, South Korea.
| |
Collapse
|
27
|
Choi JE, Verhaegen ME, Yazdani S, Malik R, Harms PW, Mangelberger D, Tien J, Cao X, Wang Y, Cieślik M, Gurkan J, Yazdani M, Jing X, Juckette K, Su F, Wang R, Zhou B, Apel IJ, Wang S, Dlugosz AA, Chinnaiyan AM. Characterizing the Therapeutic Potential of a Potent BET Degrader in Merkel Cell Carcinoma. Neoplasia 2019; 21:322-330. [PMID: 30797188 PMCID: PMC6384317 DOI: 10.1016/j.neo.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/21/2022]
Abstract
Studies on the efficacy of small molecule inhibitors in Merkel cell carcinoma (MCC) have been limited and largely inconclusive. In this study, we investigated the therapeutic potential of a potent BET degrader, BETd-246, in the treatment of MCC. We found that MCC cell lines were significantly more sensitive to BETd-246 than to BET inhibitor treatment. Therapeutic targeting of BET proteins resulted in a loss of "MCC signature" genes but not MYC expression as previously described irrespective of Merkel cell polyomavirus (MCPyV) status. In MCPyV+ MCC cells, BETd-246 alone suppressed downstream targets in the MCPyV-LT Ag axis. We also found enrichment of HOX and cell cycle genes in MCPyV- MCC cell lines that were intrinsically resistant to BETd-246. Our findings uncover a requirement for BET proteins in maintaining MCC lineage identity and point to the potential utility of BET degraders for treating MCC.
Collapse
MESH Headings
- Acetanilides/pharmacology
- Antigens, Viral, Tumor/genetics
- Antigens, Viral, Tumor/metabolism
- Carcinoma, Merkel Cell/drug therapy
- Carcinoma, Merkel Cell/etiology
- Carcinoma, Merkel Cell/metabolism
- Carcinoma, Merkel Cell/pathology
- Cell Cycle/genetics
- Cell Line, Tumor
- Dose-Response Relationship, Drug
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Genes, Homeobox
- Heterocyclic Compounds, 3-Ring/pharmacology
- Humans
- Merkel cell polyomavirus/physiology
- Polyomavirus Infections/complications
- Polyomavirus Infections/virology
- Proteins/antagonists & inhibitors
- Proteins/metabolism
- Proteolysis
- Skin Neoplasms/drug therapy
- Skin Neoplasms/etiology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Transcriptome
Collapse
Affiliation(s)
- Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Cancer Biology Program, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Sahr Yazdani
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rohit Malik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Paul W Harms
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | | | - Jean Tien
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieślik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Gurkan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mishaal Yazdani
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xiaojun Jing
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kristin Juckette
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Bing Zhou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ingrid J Apel
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
He L, Meng D, Zhang SH, Zhang Y, Deng Z, Kong LB. microRNA-608 inhibits human hepatocellular carcinoma cell proliferation via targeting the BET family protein BRD4. Biochem Biophys Res Commun 2018; 501:1060-1067. [PMID: 29777702 DOI: 10.1016/j.bbrc.2018.05.108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 12/16/2022]
Abstract
Over-expression of the bromodomain and extraterminal (BET) family protein BRD4 is associated with hepatocellular carcinoma (HCC) progression. In the present study, we indentified a novel putative anti-BRD4 microRNA: microRNA-608 ("miR-608"). In HepG2 cells and primary human HCC cells, over-expression of miR-608, using a lentiviral construct, induced BRD4 downregulation and proliferation inhibition. Conversely, transfection of the miR-608 inhibitor increased BRD4 expression to promote HepG2 cell proliferation. Our results suggest that BRD4 is the primary target gene of miR-608 in HepG2 cells. shRNA-mediated knockdown or CRSIPR/Cas9-mediated knockout of BRD4 mimicked and overtook miR-608's actions in HepG2 cells. Furthermore, introduction of a 3'-untranslated region (3'-UTR) mutant BRD4 (UTR-A1718G) blocked miR-608-induced c-Myc downregulation and proliferation inhibition in HepG2 cells. In vivo, HepG2 xenograft tumor growth was significantly inhibited after expressing miR-608 or BRD4 CRSIPR/Cas9-KO construct. Importantly, BRD4 mRNA was upregulated in human HCC tissues, which was correlated with downregulation of miR-608. Together, we conclude that miR-608 inhibits HCC cell proliferation possibly via targeting BET family protein BRD4.
Collapse
Affiliation(s)
- Ling He
- Liver and Cholecyst Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dijuan Meng
- Nursing College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shi-Hu Zhang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengming Deng
- Liver and Cholecyst Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lian-Bao Kong
- Liver and Cholecyst Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
29
|
Fang ZQ, Li MC, Zhang YQ, Liu XG. MiR-490-5p inhibits the metastasis of hepatocellular carcinoma by down-regulating E2F2 and ECT2. J Cell Biochem 2018; 119:8317-8324. [PMID: 29932246 DOI: 10.1002/jcb.26876] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/21/2018] [Indexed: 01/07/2023]
Abstract
We intended to evaluate miR-490-5p expression in hepatocellular carcinoma (HCC) tissues and detect the potential targets of miR-490-5p. In vitro experiments were conducted to further investigate the biological function of miR-490-5p on HCC cell metastasis. We investigated the abnormally expressed miRNAs in HCC tissues, and the miR-490-5p expression level was detected by qRT-PCR. E2F2 and ECT2 were proved to be the potential targets of miR-490-5p by luciferase reporter assay. The expression levels of E2F2 and ECT2 were determined using Western blot. Transwell assay was used to analyse the impact of miR-490-5p on metastasis of HCC cells. Four high-expressed miRNAs, and seven low-expressed miRNAs, including miR-490-5p, were detected in HCC tissues. The expression level of miR-490-5p was connected with the tumor size, tumor node metastasis (TNM) stage, and survival ratio of HCC patients. E2F2 and ECT2 were the targets of miR-490-5p, and miR-490-5p inhibited HCC cell metastasis through down-regulating the expressions of E2F2 and ECT2. The over-expressed miR-490-5p could restrain the metastasis of HCC cells by down-regulating E2F2 and ECT2 expression levels.
Collapse
Affiliation(s)
- Zheng-Qing Fang
- School of Nursing, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Mao-Chun Li
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu-Qin Zhang
- Department of Medicine, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui, China
| | - Xiang-Guo Liu
- College of Chinese and Western Medicine (School of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
30
|
Xu K, Chen D, Qian D, Zhang S, Zhang Y, Guo S, Ma Z, Wang S. AZD5153, a novel BRD4 inhibitor, suppresses human thyroid carcinoma cell growth in vitro and in vivo. Biochem Biophys Res Commun 2018; 499:531-537. [PMID: 29596834 DOI: 10.1016/j.bbrc.2018.03.184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/24/2018] [Indexed: 01/23/2023]
Abstract
The development of novel anti-papillary thyroid carcinoma agents is urgent. AZD5153 is a novel and specific Bromodomain-containing protein 4 (BRD4) inhibitor. Here, we show that AZD5153 dose-dependently inhibited survival, proliferation and cell cycle progression in TPC-1 cells and primary human thyroid carcinoma cells. Yet, it was non-cytotoxic to the primary thyroid epithelial cells. AZD5153 induced caspase-3/-9 and apoptosis activation in TPC-1 cells and primary cancer cells. Its cytotoxicity in TPC-1 cells was significantly attenuated with co-treatment of the caspase inhibitors. BRD4 expression was elevated in TPC-1 and primary human thyroid carcinoma cells, but was low in the thyroid epithelial cells. BRD4-regulated proteins, including c-Myc, Bcl-2 and cyclin D1, were significantly downregulated following AZD5153 treatment in TPC-1 and primary cancer cells. In vivo, oral administration of AZD5153 at well-tolerated doses significantly inhibited TPC-1 xenograft growth in severe combined immunodeficient (SCID) mice. BRD4-dependent proteins, Myc, Bcl-2 and cyclin D1, were also downregulated in AZD5153-treated tumor tissues. Collectively, the results suggest that targeting BRD4 by AZD5153 inhibits human thyroid carcinoma cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Kun Xu
- Department of Breast Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dexuan Chen
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dong Qian
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shihu Zhang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Song Guo
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaoqun Ma
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Shui Wang
- Department of Breast Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
31
|
Wen DY, Lin P, Pang YY, Chen G, He Y, Dang YW, Yang H. Expression of the Long Intergenic Non-Protein Coding RNA 665 (LINC00665) Gene and the Cell Cycle in Hepatocellular Carcinoma Using The Cancer Genome Atlas, the Gene Expression Omnibus, and Quantitative Real-Time Polymerase Chain Reaction. Med Sci Monit 2018; 24:2786-2808. [PMID: 29728556 PMCID: PMC5956974 DOI: 10.12659/msm.907389] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Long non-coding RNAs (lncRNAs) have a role in physiological and pathological processes, including cancer. The aim of this study was to investigate the expression of the long intergenic non-protein coding RNA 665 (LINC00665) gene and the cell cycle in hepatocellular carcinoma (HCC) using database analysis including The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO), and quantitative real-time polymerase chain reaction (qPCR). Material/Methods Expression levels of LINC00665 were compared between human tissue samples of HCC and adjacent normal liver, clinicopathological correlations were made using TCGA and the GEO, and qPCR was performed to validate the findings. Other public databases were searched for other genes associated with LINC00665 expression, including The Atlas of Noncoding RNAs in Cancer (TANRIC), the Multi Experiment Matrix (MEM), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interaction (PPI) networks. Results Overexpression of LINC00665 in patients with HCC was significantly associated with gender, tumor grade, stage, and tumor cell type. Overexpression of LINC00665 in patients with HCC was significantly associated with overall survival (OS) (HR=1.47795%; CI: 1.046–2.086). Bioinformatics analysis identified 469 related genes and further analysis supported a hypothesis that LINC00665 regulates pathways in the cell cycle to facilitate the development and progression of HCC through ten identified core genes: CDK1, BUB1B, BUB1, PLK1, CCNB2, CCNB1, CDC20, ESPL1, MAD2L1, and CCNA2. Conclusions Overexpression of the lncRNA, LINC00665 may be involved in the regulation of cell cycle pathways in HCC through ten identified hub genes.
Collapse
Affiliation(s)
- Dong-Yue Wen
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Peng Lin
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yu-Yan Pang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yun He
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yi-Wu Dang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Hong Yang
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
32
|
Russell JO, Ko S, Saggi HS, Singh S, Poddar M, Shin D, Monga SP. Bromodomain and Extraterminal (BET) Proteins Regulate Hepatocyte Proliferation in Hepatocyte-Driven Liver Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1389-1405. [PMID: 29545201 DOI: 10.1016/j.ajpath.2018.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 12/20/2022]
Abstract
Bromodomain and extraterminal (BET) proteins recruit key components of basic transcriptional machinery to promote gene expression. Aberrant expression and mutations in BET genes have been identified in many malignancies. Small molecule inhibitors of BET proteins such as JQ1 have shown efficacy in preclinical cancer models, including affecting growth of hepatocellular carcinoma. BET proteins also regulate cell proliferation in nontumor settings. We recently showed that BET proteins regulate cholangiocyte-driven liver regeneration. Here, we studied the role of BET proteins in hepatocyte-driven liver regeneration in partial hepatectomy (PHx) and acetaminophen-induced liver injury models in mice and zebrafish. JQ1 was injected 2 or 16 hours after PHx in mice to determine effect on hepatic injury, regeneration, and signaling. Mice treated with JQ1 after PHx displayed increased liver injury and a near-complete inhibition of hepatocyte proliferation. Levels of Ccnd1 mRNA and Cyclin D1 protein were reduced in animals injected with JQ1 16 hours after PHx and were even further reduced in animals injected with JQ1 2 hours after PHx. JQ1-treated zebrafish larvae after acetaminophen-induced injury also displayed notably impaired hepatocyte proliferation. In both models, Wnt signaling was prominently suppressed by JQ1. Our results show that BET proteins regulate hepatocyte proliferation-driven liver regeneration, and Wnt signaling is particularly sensitive to BET protein inhibition.
Collapse
Affiliation(s)
- Jacquelyn O Russell
- Department of Pathology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania
| | - Sungjin Ko
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Department of Developmental Biology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania
| | - Harvinder S Saggi
- Department of Pathology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania
| | - Minakshi Poddar
- Department of Pathology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania
| | - Donghun Shin
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Department of Developmental Biology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania.
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania; Pittsburgh Liver Research Center, University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine Pittsburgh, Pennsylvania.
| |
Collapse
|
33
|
OTX015 (MK-8628), a novel BET inhibitor, exhibits antitumor activity in non-small cell and small cell lung cancer models harboring different oncogenic mutations. Oncotarget 2018; 7:84675-84687. [PMID: 27835869 PMCID: PMC5354535 DOI: 10.18632/oncotarget.13181] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 10/14/2016] [Indexed: 12/11/2022] Open
Abstract
Inhibitors targeting epigenetic control points of oncogenes offer a potential mean of blocking tumor progression in small cell and non-small cell lung carcinomas (SCLC, NSCLC). OTX015 (MK-8628) is a BET inhibitor selectively blocking BRD2/3/4. OTX015 was evaluated in a panel of NSCLC or SCLC models harboring different oncogenic mutations. Cell proliferation inhibition and cell cycle arrest were seen in sensitive NSCLC cells. MYC and MYCN were downregulated at both the mRNA and protein levels. In addition, OTX015-treatment significantly downregulated various stemness cell markers, including NANOG, Musashi-1, CD113 and EpCAM in H3122-tumors in vivo. Conversely, in SCLC models, weak antitumor activity was observed with OTX015, both in vitro and in vivo. No predictive biomarkers of OTX015 activity were identified in a large panel of candidate genes known to be affected by BET inhibition. In NSCLC models, OTX015 was equally active in both EML4-ALK positive and negative cell lines, whereas in SCLC models the presence of functional RB1 protein, which controls cell progression at G1, may be related to the final biological outcome of OTX015. Gene expression profiling in NSCLC and SCLC cell lines showed that OTX015 affects important genes and pathways with a very high overlapping between both sensitive and resistant cell lines. These data support the rationale for the OTX015 Phase Ib (NCT02259114) in solid tumors, where NSCLC patients with rearranged ALK gene or KRAS-positive mutations are currently being treated.
Collapse
|
34
|
Xiang T, Bai JY, She C, Yu DJ, Zhou XZ, Zhao TL. Bromodomain protein BRD4 promotes cell proliferation in skin squamous cell carcinoma. Cell Signal 2018; 42:106-113. [DOI: 10.1016/j.cellsig.2017.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/16/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022]
|
35
|
Zhu H, Mao JH, Wang Y, Gu DH, Pan XD, Shan Y, Zheng B. Dual inhibition of BRD4 and PI3K-AKT by SF2523 suppresses human renal cell carcinoma cell growth. Oncotarget 2017; 8:98471-98481. [PMID: 29228703 PMCID: PMC5716743 DOI: 10.18632/oncotarget.21432] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022] Open
Abstract
Bromodomain-containing protein 4 (BRD4) and PI3K-AKT are both important for renal cell carcinoma (RCC) development and progression. SF2523 is a BRD4 and PI3K-AKT dual inhibitor. The present study demonstrated that SF2523 was cytotoxic and anti-proliferative to established RCC cell lines (786-O and A498) and primary human RCC cells. SF2523 induced activation of caspase and apoptosis in RCC cells. Further, SF2523 disrupted RCC cell cycle progression and inhibited cell migration in vitro. At the signaling level, SF2523 in-activated PI3K-AKT-mTOR, and downregulated BRD4-dependent proteins, Bcl-2 and Myc, in RCC cells. Remarkably, SF2523 was more efficient than Wortmannin (the PI3K inhibitor) and JQ1 (the BRD4 specific inhibitor) in killing RCC cells. In vivo, SF2523 administration at well-tolerated doses suppressed 786-O xenograft tumor growth in severe combined immunodeficient (SCID) mice. Together, our results suggest that concurrent blockage of BRD4 and PI3K-AKT signalings by SF2523 efficiently inhibits RCC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Hua Zhu
- The Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China.,The Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Jia-Hui Mao
- Department of Pathophysiology, Nantong University School of Medicine, Nantong, China
| | - Yin Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Dong-Hua Gu
- The Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xiao-Dong Pan
- The Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Yuxi Shan
- The Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bing Zheng
- The Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
36
|
Sahni JM, Gayle SS, Webb BM, Weber-Bonk KL, Seachrist DD, Singh S, Sizemore ST, Restrepo NA, Bebek G, Scacheri PC, Varadan V, Summers MK, Keri RA. Mitotic Vulnerability in Triple-Negative Breast Cancer Associated with LIN9 Is Targetable with BET Inhibitors. Cancer Res 2017; 77:5395-5408. [PMID: 28807940 DOI: 10.1158/0008-5472.can-17-1571] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/12/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancers (TNBC) are highly aggressive, lack FDA-approved targeted therapies, and frequently recur, making the discovery of novel therapeutic targets for this disease imperative. Our previous analysis of the molecular mechanisms of action of bromodomain and extraterminal protein inhibitors (BETi) in TNBC revealed these drugs cause multinucleation, indicating BET proteins are essential for efficient mitosis and cytokinesis. Here, using live cell imaging, we show that BET inhibition prolonged mitotic progression and induced mitotic cell death, both of which are indicative of mitotic catastrophe. Mechanistically, the mitosis regulator LIN9 was a direct target of BET proteins that mediated the effects of BET proteins on mitosis in TNBC. Although BETi have been proposed to function by dismantling super-enhancers (SE), the LIN9 gene lacks an SE but was amplified or overexpressed in the majority of TNBCs. In addition, its mRNA expression predicted poor outcome across breast cancer subtypes. Together, these results provide a mechanism for cancer selectivity of BETi that extends beyond modulation of SE-associated genes and suggest that cancers dependent upon LIN9 overexpression may be particularly vulnerable to BETi. Cancer Res; 77(19); 5395-408. ©2017 AACR.
Collapse
Affiliation(s)
- Jennifer M Sahni
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Sylvia S Gayle
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Bryan M Webb
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | | | - Darcie D Seachrist
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - Salendra Singh
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Steven T Sizemore
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Nicole A Restrepo
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio
| | - Gurkan Bebek
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio
| | - Peter C Scacheri
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Vinay Varadan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Matthew K Summers
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Ruth A Keri
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio.,Department General Medical Sciences-Oncology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
37
|
Wang L, Wu X, Wang R, Yang C, Li Z, Wang C, Zhang F, Yang P. BRD4 inhibition suppresses cell growth, migration and invasion of salivary adenoid cystic carcinoma. Biol Res 2017; 50:19. [PMID: 28545522 PMCID: PMC5445403 DOI: 10.1186/s40659-017-0124-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
Background Bromodomain-containing protein 4 (BRD4) inhibition is a new therapeutic strategy for many malignancies. In this study, we aimed to explore the effect of BRD4 inhibition by JQ1 on in vitro cell growth, migration and invasion of salivary adenoid cystic carcinoma (SACC). Methods The human normal epithelial cells and SACC cells (ACC-LM and ACC-83) were treated with JQ1 at concentrations of 0, 0.1, 0.5 or 1 μM. Cell Counting Kit-8 (CCK-8) assay was performed to evaluate cell proliferation. Cell apoptosis and cell cycle distribution was evaluated by Flow cytometry. Immunofluorescence staining was used to examine the expression of BRD4 in SACC cells. The quantitative real-time polymerase chain reaction (qRT-PCR) assay and western blot assay were performed to examine messenger RNA (mRNA) and protein levels in SACC cells. Wound-healing assay and transwell assay were used to evaluate the activities of migration and invasion of SACC cells. Results JQ1 exhibits no adverse effects on proliferation, cell cycle and cell apoptosis of the normal human epithelial cells, while suppressed proliferation and cell cycle, and induced apoptosis of SACC cells, down-regulated the mRNA and protein levels of BRD4 in SACC cells, meanwhile reduced protein expressions of c-myc and BCL-2, two known target genes of BRD4. Moreover, JQ1 inhibited SACC cell migration and invasion by regulating key epithelial–mesenchymal transition (EMT) characteristics including E-cadherin, Vimentin and Twist. Conclusions BRD4 is an important transcription factor in SACC and BRD4 inhibition by JQ1 may be a new strategy for SACC treatment.
Collapse
Affiliation(s)
- Limei Wang
- Department of Periodontology, School of Stomatology, Shandong University, 44-1 West Wen Hua Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, Shandong, China
| | - Xiuyin Wu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, Shandong, China.,Department of Stomatology, Laiwu City People's Hospital, Laiwu, 271100, Shandong, China
| | - Ruolin Wang
- Department of Periodontology, School of Stomatology, Shandong University, 44-1 West Wen Hua Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, Shandong, China
| | - Chengzhe Yang
- Department of Oral & Maxillofacial Surgery, Qilu Hospital, and Institute of Stomatology, Shandong University, Jinan, 250012, Shandong, China
| | - Zhi Li
- Department of Periodontology, School of Stomatology, Shandong University, 44-1 West Wen Hua Road, Jinan, 250012, Shandong, People's Republic of China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, Shandong, China
| | - Cunwei Wang
- Department of Prosthodontics, School of Stomatology, Shandong University, Jinan, 250012, Shandong, China
| | - Fenghe Zhang
- Department of Oral & Maxillofacial Surgery, School of Stomatology, Shandong University, 44-1 West Wen Hua Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Pishan Yang
- Department of Periodontology, School of Stomatology, Shandong University, 44-1 West Wen Hua Road, Jinan, 250012, Shandong, People's Republic of China. .,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Jinan, 250012, Shandong, China.
| |
Collapse
|
38
|
Bromodomains in Protozoan Parasites: Evolution, Function, and Opportunities for Drug Development. Microbiol Mol Biol Rev 2017; 81:81/1/e00047-16. [PMID: 28077462 DOI: 10.1128/mmbr.00047-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Parasitic infections remain one of the most pressing global health concerns of our day, affecting billions of people and producing unsustainable economic burdens. The rise of drug-resistant parasites has created an urgent need to study their biology in hopes of uncovering new potential drug targets. It has been established that disrupting gene expression by interfering with lysine acetylation is detrimental to survival of apicomplexan (Toxoplasma gondii and Plasmodium spp.) and kinetoplastid (Leishmania spp. and Trypanosoma spp.) parasites. As "readers" of lysine acetylation, bromodomain proteins have emerged as key gene expression regulators and a promising new class of drug target. Here we review recent studies that demonstrate the essential roles played by bromodomain-containing proteins in parasite viability, invasion, and stage switching and present work showing the efficacy of bromodomain inhibitors as novel antiparasitic agents. In addition, we performed a phylogenetic analysis of bromodomain proteins in representative pathogens, some of which possess unique features that may be specific to parasite processes and useful in future drug development.
Collapse
|
39
|
Abstract
Aberrations in the epigenetic landscape are a hallmark of cancer. Alterations in enzymes that are “writers,” “erasers,” or “readers” of histone modification marks are common. Bromodomains are “readers” that bind acetylated lysines in histone tails. Their most important function is the regulation of gene transcription by the recruitment of different molecular partners. Moreover, proteins containing bromodomains are also epigenetic regulators, although little is known about the specific function of these domains. In recent years, there has been increasing interest in developing small molecules that can target specific bromodomains. First, this has helped clarify biological functions of bromodomain-containing proteins. Secondly, it opens a new front for combatting cancer. In this review we will describe the structures and mechanisms associated with Bromodomain and Extra-Terminal motif (BET) inhibitors and non-BET inhibitors, their current status of development, and their promising role as anti-cancer agents.
Collapse
Affiliation(s)
- Montserrat Pérez-Salvia
- a Cancer Epigenetics and Biology Program (PEBC) , Bellvitge Biomedical Research Institute (IDIBELL) , Barcelona , Catalonia , Spain
| | - Manel Esteller
- a Cancer Epigenetics and Biology Program (PEBC) , Bellvitge Biomedical Research Institute (IDIBELL) , Barcelona , Catalonia , Spain.,b Department of Physiological Sciences II, School of Medicine , University of Barcelona , Barcelona , Catalonia , Spain.,c Institució Catalana de Recerca i Estudis Avançats (ICREA) , Barcelona , Catalonia , Spain
| |
Collapse
|
40
|
Singh AR, Joshi S, Burgoyne AM, Sicklick JK, Ikeda S, Kono Y, Garlich JR, Morales GA, Durden DL. Single Agent and Synergistic Activity of the "First-in-Class" Dual PI3K/BRD4 Inhibitor SF1126 with Sorafenib in Hepatocellular Carcinoma. Mol Cancer Ther 2016; 15:2553-2562. [PMID: 27496136 DOI: 10.1158/1535-7163.mct-15-0976] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 07/08/2016] [Indexed: 01/30/2023]
Abstract
Deregulated PI3K/AKT/mTOR, Ras/Raf/MAPK, and c-Myc signaling pathways are of prognostic significance in hepatocellular carcinoma (HCC). Sorafenib, the only drug clinically approved for patients with advanced HCC, blocks the Ras/Raf/MAPK pathway but it does not inhibit the PI3K/AKT/mTOR pathway or c-Myc activation. Hence, there is an unmet medical need to identify potent PI3K/BRD4 inhibitors, which can be used either alone or in combination with sorafenib to treat patients with advanced HCC. Herein, we show that SF1126 (pan PI3K/BRD4 inhibitor) as single agent or in combination with sorafenib inhibited proliferation, cell cycle, apoptosis, and multiple key enzymes in PI3K/AKT/mTOR and Ras/Raf/MAPK pathway in Hep3B, HepG2, SK-Hep1, and Huh7 HCC cell lines. We demonstrate that the active moiety of the SF1126 prodrug LY294002 binds to and blocks BRD4 interaction with the acetylated histone-H4 chromatin mark protein and displaced BRD4 coactivator protein from the transcriptional start site of MYC in Huh7 and SK-Hep1 HCC cell lines. Moreover, SF1126 blocked expression levels of c-Myc in HCC cells. Treatment of SF1126 either alone or in combination with sorafenib showed significant antitumor activity in vivo Our results establish that SF1126 is a dual PI3K/BRD4 inhibitor. This agent has completed a phase I clinical trial in humans with good safety profile. Our data support the potential future consideration of a phase II clinical trial of SF1126, a clinically relevant dual "first-in-class" PI3K/BRD4 inhibitor in advanced HCC, and a potential combination with sorafenib. Mol Cancer Ther; 15(11); 2553-62. ©2016 AACR.
Collapse
Affiliation(s)
- Alok R Singh
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Shweta Joshi
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Adam M Burgoyne
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Jason K Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Sadakatsu Ikeda
- Division of Hematology-Oncology, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Yuko Kono
- Division of Hepatology, Department of Medicine, University of California San Diego, La Jolla, California
| | | | | | - Donald L Durden
- Department of Pediatrics, Moores Cancer Center, University of California San Diego, La Jolla, California.
- SignalRx Pharmaceuticals, San Diego, California
- Division of Pediatric Hematology-Oncology, UCSD Rady Children's Hospital, University of California San Diego Health System, La Jolla, California
| |
Collapse
|