1
|
Miao R, Liu Y, Shen S, Wang W, Wang S. Chromatin remodeling in lymphocytic function and fate: the multifaceted roles of SWI/SNF complex. Front Immunol 2025; 16:1575857. [PMID: 40342423 PMCID: PMC12058788 DOI: 10.3389/fimmu.2025.1575857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
The Switch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex comprises 10-15 subunits, which modulate the arrangement, location, or conformation of nucleosomes to upregulate chromatin accessibility. During lymphocytic differentiation and functional development, the SWI/SNF complex exerts its effects by binding to specific transcription factors (TFs) or DNA sequences via its subunits, which are thereafter recruited to the promoter or enhancer regions of target genes, rendering each subunit crucial wherein. The loss of individual subunits during lymphocytic differentiation not only disrupts the targeting of the SWI/SNF complex but also impairs its chromatin remodeling function, ultimately resulting in altered differentiation of immature lymphocytes, dysfunction of mature lymphocytes, and injured immune responses. Therefore, in this paper, we focus on TFs interacting with SWI/SNF complex subunits in lymphocytes, and summarize the effects of the loss of specific subunits of the SWI/SNF complex on lymphocytic differentiation and function, as well as the modification in the expression of key genes. We also summarize the potential clinical treatments and applications targeting the loss of SWI/SNF complex subunits, and focus on the application in Chimeric Antigen Receptor (CAR) technology. In conclusion, the SWI/SNF complex is a key regulatory factor in lymphocytic biology, involved in fundamental cellular processes and closely associated with hematological diseases and immune dysfunction. However, the specific roles of SWI/SNF complex subunits in different lymphocytic subpopulations remain unclear. Future clarification of the specific functions of these subunits in different lymphocytic subsets is expected to promote the development of immunotherapy and personalized therapy.
Collapse
Affiliation(s)
- Renjie Miao
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Liu
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shuo Shen
- Affiliated Third Hospital of Zhenjiang to Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenxin Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| | - Shengjun Wang
- Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang,
Jiangsu, China
| |
Collapse
|
2
|
Guo Y, Lu J, Li X, Yan S, Zhou J, Tian Z, Liu Y, Li N, Zhou Q, Li X, Shi L, Jiang S, Li M, Zhou X, Huang D, Zeng Z, Fan S, Xiong W, Zhou M, Li G, Zhang W. BRD7 Inhibited Immune Escape in Nasopharyngeal Carcinoma via Inhibiting PD-L1 Expression. Int J Biol Sci 2025; 21:1914-1931. [PMID: 40083706 PMCID: PMC11900823 DOI: 10.7150/ijbs.103703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
Therapeutic strategies aimed at harnessing anti-tumor immunity are being intensively investigated as they show promising results in cancer treatment. The PD-1/ PD-L1 pathway is an essential target for restoring functional anti-tumor immune response. BRD7 is a candidate tumor suppressor gene and nuclear transcription factor of nasopharyngeal carcinoma (NPC) which was cloned in our laboratory. In this paper, we reported that the candidate tumor suppressor gene BRD7 was strongly associated with good prognosis of NPC patients and negatively regulated PD-L1 expression. In addition, we found that BRD7 down-regulated PD-L1 expression and enhanced the killing function of CD8+ T lymphocytes in NPC cells through binding to p85α via the 485-651 domain and inhibiting the activity of PI3K, thereby inhibiting the activity of PI3K/AKT/mTOR/STAT3 pathway. In vivo experiments, the results showed that BRD7 could not only inhibit the growth of tumors, but also play a better anti-tumor effect when combined with PD-L1 antibody. These results provided further evidence that BRD7 inhibited immune escape of NPC through down-regulating PD-L1 expression.
Collapse
Affiliation(s)
- Yilin Guo
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Blood Transfusion, Children's Hospital Affiliated to Zhengzhou University,Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiaxue Lu
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoxu Li
- Department of Clinical Laboratory, WuHu Hospital, East China Normal University (The Second People's Hospital, WuHu), Wuhu, Anhui, China
| | - Shiqi Yan
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jieyu Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ziying Tian
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ying Liu
- Department of Clinical Laboratory, Zhengzhou Orthopaedics Hospital, Zhengzhou, China
| | - Nan Li
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Zhou
- Department of Clinical Laboratory, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Shi
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Su Jiang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Xiao Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital/ the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, Hunan, China
- Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Yang N, Jiao M, Zhang Y, Mo S, Wang L, Liang J. Roles and mechanisms of circular RNA in respiratory system cancers. Front Oncol 2024; 14:1430051. [PMID: 39077467 PMCID: PMC11284073 DOI: 10.3389/fonc.2024.1430051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Circular RNAs (circRNAs) constitute a class of endogenous non-coding RNAs (ncRNAs) that lack a 5'-ended cap and 3'-ended poly (A) tail and form a closed ring structure with covalent bonds. Due to its special structure, circRNA is resistant to Exonuclease R (RNaseR), making its distribution in the cytoplasm quite rich. Advanced high-throughput sequencing and bioinformatics methods have revealed that circRNA is highly conserved, stable, and disease- and tissue-specific. Furthermore, increasing research has confirmed that circRNA, as a driver or suppressor, regulates cancer onset and progression by modulating a series of pathophysiological mechanisms. As a result, circRNA has emerged as a clinical biomarker and therapeutic intervention target. This article reviews the biological functions and regulatory mechanisms of circRNA in the context of respiratory cancer onset and progression.
Collapse
Affiliation(s)
- Nan Yang
- School of Basic Medical, Gansu University of Chinese Medicine, Lanzhou, China
| | - Mengwen Jiao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yuewen Zhang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shaokang Mo
- Department of Obstetrics and Gynecology, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| | - Ling Wang
- Department of Obstetrics and Gynecology, The 940th Hospital of Joint Logistics Support Force of Chinese People’s Liberation Army, Lanzhou, China
| | - Jianqing Liang
- School of Basic Medical, Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
4
|
Wei J, Li M, Chen S, Xue C, Zheng L, Duan Y, Deng H, Fan S, Xiong W, Zhou M. CircBRD7 attenuates tumor growth and metastasis in nasopharyngeal carcinoma via epigenetic activation of its host gene. Cancer Sci 2024; 115:139-154. [PMID: 37940358 PMCID: PMC10823269 DOI: 10.1111/cas.15998] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
BRD7 was identified as a tumor suppressor in nasopharyngeal carcinoma (NPC). Circular RNAs (CircRNAs) are involved in the occurrence and development of NPC as oncogenes or tumor suppressors. However, the function and mechanism of the circular RNA forms derived from BRD7 in NPC are not well understood. In this study, we first identified that circBRD7 was a novel circRNA derived from BRD7 that inhibited cell proliferation, migration, invasion of NPC cells, as well as the xenograft tumor growth and metastasis in vivo. Mechanistically, circBRD7 promoted the transcriptional activation and expression of BRD7 by enhancing the enrichment of histone 3 lysine 27 acetylation (H3K27ac) in the promoter region of its host gene BRD7, and BRD7 promoted the formation of circBRD7. Therefore, circBRD7 formed a positive feedback loop with BRD7 to inhibit NPC development and progression. Moreover, restoration of BRD7 expression rescued the inhibitory effect of circBRD7 on the malignant progression of NPC. In addition, circBRD7 demonstrated low expression in NPC tissues, which was positively correlated with BRD7 expression and negatively correlated with the clinical stage of NPC patients. Taken together, circBRD7 attenuates the tumor growth and metastasis of NPC by forming a positive feedback loop with its host gene BRD7, and targeting the circBRD7/BRD7 axis is a promising strategy for the clinical diagnosis and treatment of NPC.
Collapse
Affiliation(s)
- Jianxia Wei
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Shipeng Chen
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Changning Xue
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Lemei Zheng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Yumei Duan
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Hongyu Deng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Songqing Fan
- Department of Pathology, the Second Xiangya HospitalCentral South UniversityChangshaChina
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Cancer Research Institute and School of Basic Medical SciencesCentral South UniversityChangshaChina
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of EducationCentral South UniversityChangshaChina
| |
Collapse
|
5
|
Huang K, Wu Y, Fan W, Zhao Y, Xue M, Liu H, Tang Y, Li J. Identification of BRD7 by whole-exome sequencing as a predictor for intermediate-stage hepatocellular carcinoma in patients undergoing TACE. J Cancer Res Clin Oncol 2023; 149:11247-11261. [PMID: 37365429 DOI: 10.1007/s00432-023-04883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023]
Abstract
OBJECTIVE In the present study, we aimed to identify potential predictors of intermediate-stage hepatocellular carcinoma (HCC) using whole-exome sequencing (WES) in patients undergoing transarterial chemoembolization (TACE). MATERIALS AND METHODS In A total of 51 patients, newly diagnosed with intermediate-stage HCC between January 2013 and December 2020, were enrolled. Prior to treatment, histological samples were collected for western blotting and immunohistochemistry. The predictive roles of clinical indicators and genes in patient prognosis were analyzed using univariate and multivariate analyses. Finally, the correlation between imaging features and gene signatures was examined. RESULTS Using WES, we identified that bromodomain-containing protein 7 (BRD7) was significantly mutated in patients with different TACE responses. No significant difference in BRD7 expression was observed between patients with and without BRD7 mutations. HCC tumors exhibited higher BRD7 than normal liver tissues. Multivariate analysis revealed that alpha-fetoprotein (AFP), BRD7 expression, and BRD7 mutations were independent risk factors for progression-free survival (PFS). In addition, Child-Pugh class, BRD7 expression, and BRD7 mutations were independent risk factors for overall survival (OS). Patients with wild-type BRD7 and high BRD7 expression had worse PFS and OS, whereas those with mutated BRD7 and low BRD7 expression exhibited the best PFS and OS. The Kruskal-Wallis test revealed that wash-in enhancement on computed tomography might be an independent risk factor for high BRD7 expression. CONCLUSION BRD7 expression may be an independent risk factor for prognosis in patients with HCC undergoing TACE. Imaging features such as wash-in enhancement are closely related to BRD7 expression.
Collapse
Affiliation(s)
- Kun Huang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
- Department of Radiology, Guizhou Provincial People's Hospital, No. 83 East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - Yanqin Wu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yue Zhao
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Miao Xue
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Haikuan Liu
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yiyang Tang
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2 Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
6
|
Ordonez-Rubiano SC, Maschinot CA, Wang S, Sood S, Baracaldo-Lancheros LF, Strohmier BP, McQuade AJ, Smith BC, Dykhuizen EC. Rational Design and Development of Selective BRD7 Bromodomain Inhibitors and Their Activity in Prostate Cancer. J Med Chem 2023; 66:11250-11270. [PMID: 37552884 PMCID: PMC10641717 DOI: 10.1021/acs.jmedchem.3c00671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Bromodomain-containing proteins are readers of acetylated lysine and play important roles in cancer. Bromodomain-containing protein 7 (BRD7) is implicated in multiple malignancies; however, there are no selective chemical probes to study its function in disease. Using crystal structures of BRD7 and BRD9 bromodomains (BDs) bound to BRD9-selective ligands, we identified a binding pocket exclusive to BRD7. We synthesized a series of ligands designed to occupy this binding region and identified two inhibitors with increased selectivity toward BRD7, 1-78 and 2-77, which bind with submicromolar affinity to the BRD7 BD. Our binding mode analyses indicate that these ligands occupy a uniquely accessible binding cleft in BRD7 and maintain key interactions with the asparagine and tyrosine residues critical for acetylated lysine binding. Finally, we validated the utility and selectivity of the compounds in cell-based models of prostate cancer.
Collapse
Affiliation(s)
- Sandra C Ordonez-Rubiano
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Chad A Maschinot
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Sijie Wang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Surbhi Sood
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Luisa F Baracaldo-Lancheros
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Brayden P Strohmier
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Alexander J McQuade
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Brian C Smith
- Department of Biochemistry, Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University. Robert Heine Pharmacy Building 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
- Purdue Center for Cancer Research, College of Pharmacy, Purdue University, 201 S University St., West Lafayette, Indiana 47907, United States
| |
Collapse
|
7
|
Zhou C, Xiang Y, Ren Y, Li M, Gou X, Li W. Keratin19 promotes pancreatic cancer progression and poor prognosis via activating the Hedgehog pathway. Int J Oncol 2023; 62:43. [PMID: 36825581 PMCID: PMC9946805 DOI: 10.3892/ijo.2023.5491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/19/2023] [Indexed: 02/25/2023] Open
Abstract
Pancreatic cancer is a serious threat to human health, with strong invasiveness, rapid progression and poor prognosis. Tumors expressing keratin 19 (K19) have stronger invasiveness and a worse prognosis. However, the role and mechanism of K19 in pancreatic cancer have remained largely elusive. In the present study, K19 expression was detected in pancreatic cancer tissues, its effect on proliferation, apoptosis and metastasis of pancreatic cancer at the cellular, in vivo preclinical and clinical levels was evaluated and its effect on the Hedgehog pathway was analyzed. K19 was significantly overexpressed in pancreatic cancer, promoted pancreatic cancer proliferation and metastasis, inhibited tumor cell apoptosis and was associated with poor prognosis. Mechanistically, these effects were mediated through the activation of the Hedgehog pathway. In conclusion, K19 may be a novel target molecule for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Changsheng Zhou
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China,Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Retroperitoneal Tumor Research Center of The Oncology Chapter of The Chinese Medical Association, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Yi Xiang
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yantao Ren
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Retroperitoneal Tumor Research Center of The Oncology Chapter of The Chinese Medical Association, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China
| | - Ming Li
- Xiamen Medicine Research Institute, Xiamen, Fujian 361005, P.R. China
| | - Xin Gou
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China,Correspondence to: Dr Xin Gou, Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, 83 Zhongshandong Road, Guiyang, Guizhou 550002, P.R. China, E-mail:
| | - Wengang Li
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Retroperitoneal Tumor Research Center of The Oncology Chapter of The Chinese Medical Association, School of Medicine, Xiamen University, Xiamen, Fujian 361102, P.R. China,Dr Wengang Li, School of Medicine, Xiamen University, 4221 Xiang'annan Road, Xiamen, Fujian 361102, P.R. China, E-mail:
| |
Collapse
|
8
|
Mollapour Sisakht M, Amirkhani MA, Nilforoushzadeh MA. SWI/SNF complex, promising target in melanoma therapy: Snapshot view. Front Med (Lausanne) 2023; 10:1096615. [PMID: 36844227 PMCID: PMC9947295 DOI: 10.3389/fmed.2023.1096615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
Therapeutic strategies based on epigenetic regulators are rapidly increasing in light of recent advances in discovering the role of epigenetic factors in response and sensitivity to therapy. Although loss-of-function mutations in genes encoding the SWItch/Sucrose NonFermentable (SWI/SNF) subunits play an important role in the occurrence of ~34% of melanomas, the potential of using inhibitors and synthetic lethality interactions between key subunits of the complex that play an important role in melanoma progression must be considered. Here, we discuss the importance of the clinical application of SWI/SNF subunits as a promising potential therapeutic in melanoma.
Collapse
Affiliation(s)
- Mahsa Mollapour Sisakht
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran,Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands,*Correspondence: Mahsa Mollapour Sisakht ✉ ; ✉
| | | | | |
Collapse
|
9
|
Li S, Yang L, Li Y, Yue W, Xin S, Li J, Long S, Zhang W, Cao P, Lu J. Epstein-Barr Virus Synergizes with BRD7 to Conquer c-Myc-Mediated Viral Latency Maintenance via Chromatin Remodeling. Microbiol Spectr 2023; 11:e0123722. [PMID: 36728436 PMCID: PMC10101146 DOI: 10.1128/spectrum.01237-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 01/06/2023] [Indexed: 02/03/2023] Open
Abstract
Epstein-Barr virus (EBV) switches between latent and lytic phases in hosts, which is important in the development of related diseases. However, the underlying mechanism of controlling the viral biphasic life cycle and how EBV mediates this regulation remain largely unknown. This study identified bromodomain-containing protein 7 (BRD7) as a crucial host protein in EBV latent infection. Based on the chromatin immunoprecipitation (ChIP) sequencing of endogenous BRD7 in Burkitt lymphoma cells, we found that EBV drove BRD7 to regulate cellular and viral genomic loci, including the transcriptional activation of c-Myc, a recently reported regulator of EBV latency. Additionally, EBV-mediated BRD7 signals were enriched around the FUSE (far-upstream sequence element) site in chromosome 8 and the enhancer LOC108348026 in the lgH locus, which might activate the c-Myc alleles. Mechanically, EBV-encoded nuclear antigen 1 (EBNA1) bound to BRD7 and colocalized at promoter regions of the related genes, thus serving as cofactors for the maintenance of viral latency. Moreover, the disruption of BRD7 decreased the c-Myc expression, induced the BZLF1 expression, and reactivated the lytic cycle. Our findings reveal the unique role of BRD7 to synergize with EBV in maintaining the viral latency state via chromatin remodeling. This study paves the way for understanding the new molecular mechanism of EBV-induced chromatin remodeling and latent-lytic switch, providing novel therapeutic candidate targets for EBV persistent infection. IMPORTANCE When establishing persistent infection in most human hosts, EBV is usually latent. How the viral latency is maintained in cells remains largely unknown. c-Myc was recently reported to act as a controller of the lytic switch, while whether and how EBV regulates it remain to be explored. Here, we identified that BRD7 is involved in controlling EBV latency. We found that EBV-mediated BRD7 was enriched in both the normal promoter regions and the translocation alleles of c-Myc, and disruption of BRD7 decreased c-Myc expression to reactivate the lytic cycle. We also demonstrated that EBV-encoded EBNA1 bound to and regulated BRD7. Therefore, we reveal a novel mechanism by which EBV can regulate its infection state by coordinating with host BRD7 to target c-Myc. Our findings will help future therapeutic intervention strategies for EBV infection and pathogenesis.
Collapse
Affiliation(s)
- Shen Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Li Yang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Yanling Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Wenxing Yue
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Shuyu Xin
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Jing Li
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Sijing Long
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Wentao Zhang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| | - Pengfei Cao
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianhong Lu
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Hematology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Wu L, Li S, Xu J, Shen C, Qian Q. AGAP2-AS1/BRD7/c-Myc signaling axis promotes skin cutaneous melanoma progression. Am J Transl Res 2023; 15:350-362. [PMID: 36777828 PMCID: PMC9908487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/19/2022] [Indexed: 02/14/2023]
Abstract
OBJECTIVE To examine the effects and mechanisms of AGAP2 Antisense RNA 1 (AGAP2-AS1) in progression of skin cutaneous melanoma (SKCM). METHODS AGAP2-AS1 expression and SKCM survival outcomes were assessed using bioinformatics analysis. In vitro and in vivo assays, including cell proliferation, colony formation, migration, and tumor formation assays, were performed to detect AGAP2-AS1 oncogenic effects in SKCM. RNA pull-down, RNA immunoprecipitation (RIP), and co-immunoprecipitation were used to evaluate the mechanism of AGAP2-AS1 in SKCM progression. RESULTS AGAP2-AS1 was upregulated in human SKCM tissues and cells and predicted a worse prognosis. AGAP2-AS1 silencing in two SKCM cell lines inhibited cell proliferation, as well as colony formation and migration both in vitro and in vivo. The RNA pull-down assay and RIP analysis results indicated that AGAP2-AS1 interacted with bromodomain containing 7 (BRD7). AGAP2-AS1 knockdown attenuated the BRD7 and c-Myc interaction, which reduced c-Myc expression. The altered phenotypes found in AGAP2-AS1- and BRD7-deficient cells were rescued by overexpression of c-Myc. CONCLUSIONS AGAP2-AS1 participated in oncogenesis in SKCM via the BRD7/c-Myc signaling pathway. These results suggest a molecular mechanism for AGAP2-AS1 in the carcinogenesis of SKCM.
Collapse
Affiliation(s)
- Lei Wu
- Department of Dermatology, First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Shenyi Li
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan UniversityWuxi 214062, Jiangsu, China
| | - Jinfu Xu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical UniversityNanjing 211166, Jiangsu, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical UniversityNanjing 211166, Jiangsu, China,State Key Laboratory of Reproductive Medicine, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical UniversitySuzhou 215002, Jiangsu, China
| | - Qihong Qian
- Department of Dermatology, First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| |
Collapse
|
11
|
Czerwinska P, Mackiewicz AA. Bromodomain (BrD) Family Members as Regulators of Cancer Stemness-A Comprehensive Review. Int J Mol Sci 2023; 24:995. [PMID: 36674511 PMCID: PMC9861003 DOI: 10.3390/ijms24020995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Epigenetic mechanisms involving DNA methylation and chromatin modifications have emerged as critical facilitators of cancer heterogeneity, substantially affecting cancer development and progression, modulating cell phenotypes, and enhancing or inhibiting cancer cell malignant properties. Not surprisingly, considering the importance of epigenetic regulators in normal stem cell maintenance, many chromatin-related proteins are essential to maintaining the cancer stem cell (CSC)-like state. With increased tumor-initiating capacities and self-renewal potential, CSCs promote tumor growth, provide therapy resistance, spread tumors, and facilitate tumor relapse after treatment. In this review, we characterized the epigenetic mechanisms that regulate the acquisition and maintenance of cancer stemness concerning selected epigenetic factors belonging to the Bromodomain (BrD) family of proteins. An increasing number of BrD proteins reinforce cancer stemness, supporting the maintenance of the cancer stem cell population in vitro and in vivo via the utilization of distinct mechanisms. As bromodomain possesses high druggable potential, specific BrD proteins might become novel therapeutic targets in cancers exhibiting de-differentiated tumor characteristics.
Collapse
Affiliation(s)
- Patrycja Czerwinska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Andrzej Adam Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
12
|
Kapoor S, Damiani E, Wang S, Dharmanand R, Tripathi C, Tovar Perez JE, Dashwood WM, Rajendran P, Dashwood RH. BRD9 Inhibition by Natural Polyphenols Targets DNA Damage/Repair and Apoptosis in Human Colon Cancer Cells. Nutrients 2022; 14:nu14204317. [PMID: 36297001 PMCID: PMC9610492 DOI: 10.3390/nu14204317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Epigenetic mechanisms play an important role in the etiology of colorectal cancer (CRC) and other malignancies due, in part, to deregulated bromodomain (BRD) functions. Inhibitors of the bromodomain and extraterminal (BET) family have entered into clinical trials as anticancer agents, and interest has grown in other acetyl 'reader' proteins as therapeutic targets, including non-BET member bromodomain-containing protein 9 (BRD9). We report here that overexpression of BRD9 is associated with poor prognosis in CRC patients, and that siRNA-mediated knockdown of BRD9 decreased cell viability and activated apoptosis in human colon cancer cells, coincident with increased DNA damage. Seeking natural compounds as BRD9 antagonists, molecular docking in silico identified several polyphenols such as Epigallocatechin-3-gallate (EGCG), Equol, Quercetin, and Aspalathin, with favorable binding energies, supported by BROMOscan® (DiscoverX) and isothermal titration calorimetry experiments. Polyphenols mimicked BRD9 knockdown and iBRD9 treatment in reducing colon cancer cell viability, inhibiting colony formation, and enhancing DNA damage and apoptosis. Normal colonic epithelial cells were unaffected, signifying cancer-specific effects. These findings suggest that natural polyphenols recognize and target BRD9 for inhibition, and might serve as useful lead compounds for bromodomain therapeutics in the clinical setting.
Collapse
Affiliation(s)
- Sabeeta Kapoor
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | - Elisabetta Damiani
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy
| | - Shan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310030, China
| | - Ravirajan Dharmanand
- Center for Infectious & Inflammatory Diseases, Texas A&M Health, Houston, TX 77030, USA
| | - Chakrapani Tripathi
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | | | - Wan Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.); Tel.: +1-713-677-7803 (P.R.); +1-713-677-7806 (R.H.D.)
| | - Roderick Hugh Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.); Tel.: +1-713-677-7803 (P.R.); +1-713-677-7806 (R.H.D.)
| |
Collapse
|
13
|
The Emerging Roles and Clinical Potential of circSMARCA5 in Cancer. Cells 2022; 11:cells11193074. [PMID: 36231036 PMCID: PMC9562909 DOI: 10.3390/cells11193074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/25/2022] [Accepted: 09/26/2022] [Indexed: 12/24/2022] Open
Abstract
Circular RNAs (circRNAs) are a type of endogenous non-coding RNA and a critical epigenetic regulation way that have a closed-loop structure and are highly stable, conserved, and tissue-specific, and they play an important role in the development of many diseases, including tumors, neurological diseases, and cardiovascular diseases. CircSMARCA5 is a circRNA formed by its parental gene SMARCA5 via back splicing which is dysregulated in expression in a variety of tumors and is involved in tumor development with dual functions as an oncogene or tumor suppressor. It not only serves as a competing endogenous RNA (ceRNA) by binding to various miRNAs, but it also interacts with RNA binding protein (RBP), regulating downstream gene expression; it also aids in DNA damage repair by regulating the transcription and expression of its parental gene. This review systematically summarized the expression and characteristics, dual biological functions, and molecular regulatory mechanisms of circSMARCA5 involved in carcinogenesis and tumor progression as well as the potential applications in early diagnosis and gene targeting therapy in tumors.
Collapse
|
14
|
Garcia-Etxebarria K, Etxart A, Barrero M, Nafria B, Segues Merino NM, Romero-Garmendia I, Franke A, D’Amato M, Bujanda L. Performance of the Use of Genetic Information to Assess the Risk of Colorectal Cancer in the Basque Population. Cancers (Basel) 2022; 14:4193. [PMID: 36077729 PMCID: PMC9454881 DOI: 10.3390/cancers14174193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/18/2022] [Accepted: 08/26/2022] [Indexed: 01/14/2023] Open
Abstract
Although the genetic contribution to colorectal cancer (CRC) has been studied in various populations, studies on the applicability of available genetic information in the Basque population are scarce. In total, 835 CRC cases and 940 controls from the Basque population were genotyped and genome-wide association studies were carried out. Mendelian Randomization analyses were used to discover the effect of modifiable risk factors and microbiota on CRC. In total, 25 polygenic risk score models were evaluated to assess their performance in CRC risk calculation. Moreover, 492 inflammatory bowel disease cases were used to assess whether that genetic information would not confuse both conditions. Five suggestive (p < 5 × 10−6) loci were associated with CRC risk, where genes previously associated with CRC were located (e.g., ABCA12, ATIC or ERBB4). Moreover, the analyses of CRC locations detected additional genes consistent with the biology of CRC. The possible contribution of cholesterol, BMI, Firmicutes and Cyanobacteria to CRC risk was detected by Mendelian Randomization. Finally, although polygenic risk score models showed variable performance, the best model performed correctly regardless of the location and did not misclassify inflammatory bowel disease cases. Our results are consistent with CRC biology and genetic risk models and could be applied to assess CRC risk in the Basque population.
Collapse
Affiliation(s)
- Koldo Garcia-Etxebarria
- Biodonostia, Gastrointestinal Genetics Group, 20014 San Sebastián, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| | - Ane Etxart
- Biodonostia, Gastrointestinal Disease Group, Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
| | - Maialen Barrero
- Biodonostia, Gastrointestinal Disease Group, Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
| | - Beatriz Nafria
- Biodonostia, Gastrointestinal Disease Group, Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
| | - Nerea Miren Segues Merino
- Biodonostia, Gastrointestinal Disease Group, Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
| | - Irati Romero-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), 48940 Leioa, Spain
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE, Basque Research and Technology Alliance, 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Sciences, 48009 Bilbao, Spain
- Department of Medicine and Surgery, LUM University, 70010 Casamassima, Italy
| | - Luis Bujanda
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
- Biodonostia, Gastrointestinal Disease Group, Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain
| |
Collapse
|
15
|
Tan L, Peng D, Cheng Y. Significant position of C-myc in colorectal cancer: a promising therapeutic target. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2295-2304. [PMID: 35972682 DOI: 10.1007/s12094-022-02910-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/23/2022] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a malignant tumor initiating from the mucosa of the colorectum. According to the 2020 statistics from the World Health Organization, there are 10.0% CRC cases among all 19.3 million new cancers, followed by lung and breast cancer, and 9.4% CRC cases among all 9.9 million cancer deaths, ranking second. The population of CRC patients in China is large, and its incidence and mortality continue to increase each year. Despite the continuous development of surgical methods, chemotherapy, radiotherapy, targeted therapy and immunotherapy, the overall survival of CRC patients remains low. Past research has suggested that c-myc plays a pivotal role in the development of CRC. A higher expression level of c-Myc is a negative prognostic marker in CRC. However, there are few drugs targeting c-myc directly. Therefore, we focused on discovering the mechanism of c-myc in CRC to provide a reference for a better therapy choice for patients.
Collapse
Affiliation(s)
- Li Tan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yong Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
16
|
Zheng L, Li M, Wei J, Chen S, Xue C, Zhan Y, Duan Y, Deng H, Xiong W, Li G, Zhou M. The emerging roles of the interaction between m6A modification and c-Myc in driving tumorigenesis and development. J Cell Physiol 2022; 237:2758-2769. [PMID: 35388487 DOI: 10.1002/jcp.30733] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/04/2022] [Accepted: 03/21/2022] [Indexed: 12/24/2022]
Abstract
N6-methyladenosine (m6A) is an extremely common and conservative posttranscriptional modification, that can specifically target and regulate the expression or stability of a series of tumor-related genes, thus playing critical roles in the occurrence and development of tumors. c-Myc is an important tumorigenic transcription factor that promotes tumorigenesis and development by mainly regulating the expression of downstream target genes. Increasing evidence shows that m6A modification, as well as abnormal expression and regulation of c-Myc, is critical molecular mechanisms driving tumorigenesis and development. Although more evidence has been uncovered about the individual roles of m6A modification or c-Myc in tumors, the interaction between m6A modification and c-Myc in tumorigenesis and development has not been systematically summarized. Therefore, this review is focused on the mutual regulation between m6A modification and c-Myc expression and stability as well as its roles in tumorigenesis and development. We also summarized the potential value of the interaction between m6A modification and m6A expression and stability in tumor diagnosis and treatment, which provides a specific reference for revealing the mechanism of tumor occurrence and development as well as clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Lemei Zheng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Mengna Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Jianxia Wei
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Shipeng Chen
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Changning Xue
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Duan
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Hongyu Deng
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, China
| |
Collapse
|
17
|
Dreier MR, de la Serna IL. SWI/SNF Chromatin Remodeling Enzymes in Melanoma. EPIGENOMES 2022; 6:epigenomes6010010. [PMID: 35323214 PMCID: PMC8947417 DOI: 10.3390/epigenomes6010010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Melanoma is an aggressive malignancy that arises from the transformation of melanocytes on the skin, mucosal membranes, and uvea of the eye. SWI/SNF chromatin remodeling enzymes are multi-subunit complexes that play important roles in the development of the melanocyte lineage and in the response to ultraviolet radiation, a key environmental risk factor for developing cutaneous melanoma. Exome sequencing has revealed frequent loss of function mutations in genes encoding SWI/SNF subunits in melanoma. However, some SWI/SNF subunits have also been demonstrated to have pro-tumorigenic roles in melanoma and to affect sensitivity to therapeutics. This review summarizes studies that have implicated SWI/SNF components in melanomagenesis and have evaluated how SWI/SNF subunits modulate the response to current therapeutics.
Collapse
|
18
|
Wang L, Wang Y, Zhao J, Yu Y, Kang N, Yang Z. Theoretical exploration of the binding selectivity of inhibitors to BRD7 and BRD9 with multiple short molecular dynamics simulations. RSC Adv 2022; 12:16663-16676. [PMID: 35754900 PMCID: PMC9169554 DOI: 10.1039/d2ra02637f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/29/2022] [Indexed: 12/18/2022] Open
Abstract
Bromodomain-containing proteins 7 and 9 (BRD7 and BRD9) have been considered as potential targets of clinical drug design toward treatment of human cancers and other diseases. Multiple short molecular dynamics simulations and binding free energy predictions were carried out to decipher the binding selectivity of three inhibitors 4L2, 5U6, and 6KT toward BRD7 and BRD9. The results show that 4L2 has more favorable binding ability to BRD7 over BRD9 compared to 5U6 and 6KT, while 5U6 and 6KT possess more favorable associations with BRD9 than BRD7. Furthermore, estimations of residue-based free energy decompositions further identify that four common residue pairs, including (F155, F44), (V160, V49), (Y168, Y57) and (Y217, Y106) in (BRD7, BRD9) generate obvious binding differences with 4L2, 5U6, and 6KT, which mostly drives the binding selectivity of 4L2, 5U6, and 6KT to BRD7 and BRD9. Dynamic information arising from trajectory analysis also suggests that inhibitor bindings affect structural flexibility and motion modes, which is responsible for the partial selectivity of 4L2, 5U6, and 6KT toward BRD7 and BRD9. As per our expectation, this study theoretically provides useful hints for design of dual inhibitors with high selectivity on BRD7 and BRD9. Bromodomains (BRDs) are structurally conserved epigenetic reader modules observed in numerous chromatin- and transcription-associated proteins that have a capability to identify acetylated lysine residues.![]()
Collapse
Affiliation(s)
- Lifei Wang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Yan Wang
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Juan Zhao
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Yingxia Yu
- School of Science, Shandong Jiaotong University, Jinan 250357, China
| | - Nianqian Kang
- Department of Physics, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zhiyong Yang
- Department of Physics, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
19
|
Mason LD, Chava S, Reddi KK, Gupta R. The BRD9/7 Inhibitor TP-472 Blocks Melanoma Tumor Growth by Suppressing ECM-Mediated Oncogenic Signaling and Inducing Apoptosis. Cancers (Basel) 2021; 13:cancers13215516. [PMID: 34771678 PMCID: PMC8582741 DOI: 10.3390/cancers13215516] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma accounts for the majority of all skin cancer-related deaths and only 1/3rd of melanoma patients with distal metastasis survive beyond five years. However, current therapies including BRAF/MEK targeted therapies or immunotherapies only benefit a subset of melanoma patients due to the emergence of intrinsic or extrinsic resistance mechanisms. Effective treatment of melanoma will thus require new and more effective therapeutic agents. Towards the goal of identifying new therapeutic agents, we conducted an unbiased, druggable epigenetic drug screen using a library of 32 epigenetic inhibitors obtained from the Structural Genome Consortium that targets proteins encoding for epigenetic regulators. This chemical genetic screening identified TP-472, which targets bromodomain-7/9, as the strongest inhibitor of melanoma growth in both short- and long-term survival assays and in mouse models of melanoma tumor growth. Mechanistically, using a transcriptome-wide mRNA sequencing profile we identified TP-472 treatment downregulates genes encoding various extracellular matrix (ECM) proteins, including integrins, collagens, and fibronectins. Reactome-based functional pathway analyses revealed that many of the ECM proteins are involved in extracellular matrix interactions required for cancer cell growth and proliferation. TP-472 treatment also upregulated several pro-apoptotic genes that can inhibit melanoma growth. Collectively, our results identify BRD7/9 inhibitor TP-472 as a potentially useful therapeutic agent for melanoma therapy.
Collapse
Affiliation(s)
- Lawrence David Mason
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35322, USA; (L.D.M.); (S.C.); (K.K.R.)
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35322, USA
- Correspondence: ; Tel.: +1-205-934-6207
| |
Collapse
|