1
|
Sun K, Zhi Y, Ren W, Li S, Zheng J, Gao L, Zhi K. Crosstalk between O-GlcNAcylation and ubiquitination: a novel strategy for overcoming cancer therapeutic resistance. Exp Hematol Oncol 2024; 13:107. [PMID: 39487556 PMCID: PMC11529444 DOI: 10.1186/s40164-024-00569-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/04/2024] [Indexed: 11/04/2024] Open
Abstract
Developing resistance to cancer treatments is a major challenge, often leading to disease recurrence and metastasis. Understanding the underlying mechanisms of therapeutic resistance is critical for developing effective strategies. O-GlcNAcylation, a post-translational modification that adds GlcNAc from the donor UDP-GlcNAc to serine and threonine residues of proteins, plays a crucial role in regulating protein function and cellular signaling, which are frequently dysregulated in cancer. Similarly, ubiquitination, which involves the attachment of ubiquitin to to proteins, is crucial for protein degradation, cell cycle control, and DNA repair. The interplay between O-GlcNAcylation and ubiquitination is associated with cancer progression and resistance to treatment. This review discusses recent discoveries regarding the roles of O-GlcNAcylation and ubiquitination in cancer resistance, their interactions, and potential mechanisms. It also explores how targeting these pathways may provide new opportunities to overcome cancer treatment resistance in cancer, offering fresh insights and directions for research and therapeutic development.
Collapse
Affiliation(s)
- Kai Sun
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
| | - Yuan Zhi
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China
| | - Jingjing Zheng
- School of Stomatology, Qingdao University, Qingdao, 266003, China
- Department of Endodontics, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Ling Gao
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China.
| | - Keqian Zhi
- Department of Oral and Maxillofacial Reconstruction, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China.
- School of Stomatology, Qingdao University, Qingdao, 266003, China.
- Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, 1677 Wutaishan Road, Huangdao Distract, Qingdao, 266003, Shandong, China.
| |
Collapse
|
2
|
Li TT, Hao QG, Teng ZW, Liu Y, Wu JF, Zhang J, Yang LR. SNAI2 as a Prognostic Biomarker Based on Cancer-Associated Fibroblasts in Patients With Lung Adenocarcinoma. Clin Med Insights Oncol 2024; 18:11795549241280506. [PMID: 39314798 PMCID: PMC11418231 DOI: 10.1177/11795549241280506] [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: 04/09/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is a common type of malignant tumor with therapeutic challenges. Cancer-associated fibroblasts (CAFs) promote LUAD growth and metastasis, regulate the tumor immune response, and influence tumor treatment responses and drug resistance. However, the molecular mechanisms through which CAFs control LUAD progression are largely unknown. In this study, we aimed to determine the correlations between CAF-related genes and overall survival (OS) in patients with LUAD. Methods We acquired the gene expression data and clinical information of 522 patients with LUAD patients from The Cancer Genome Atlas (TCGA) and 442 patients with LUAD from the Gene Expression Omnibus (GEO) databases. CAF infiltration levels were assessed using the Microenvironment Cell Population (MCP) counter, the Estimating the Proportions of Immune and Cancer cells (EPIC) algorithm, and Tumor Immune Dysfunction and Exclusion (TIDE) scores. A CAF-related gene network was constructed using the Weighted gene co-expression network analysis (WGCNA). Based on the CAF-related genes, univariate Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression analyses were performed to identify prognostic genes. Gene expression levels within the prognostic model were validated using the Cancer Cell Line Encyclopedia (CCLE) databases and Western blotting. Results Our results demonstrated that high CAF scores were associated with lower survival rates in patients with LUAD. Gene modules that were highly correlated with high CAF scores were closely associated with tissue characteristics and extracellular matrix structures in LUAD. In addition, correlations between CAF scores and responses to immunotherapy and chemotherapy were observed. Finally, we found that SNAI2 expression was higher in lung cancer tissues than in normal tissues. Conclusion Deepening our understanding of the influence of CAFs on tumor progression and treatment response at the molecular level can aid the development of more effective therapeutic strategies. This study provides important insights into the functional mechanisms of action of CAFs in LUAD and highlights their clinical implications.
Collapse
Affiliation(s)
- Tian-Tian Li
- Department of pneumology, The Central Hospital of Wuhan, Wuhan, China
| | - Qing-Gang Hao
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, China
| | - Zhao-Wei Teng
- The Central Laboratory and Department of Orthopedic, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuan Liu
- Department of general surgery, Kunming Medical University, Kunming, China
| | - Jia-Fan Wu
- Department of general surgery, Kunming Medical University, Kunming, China
| | - Jun Zhang
- Department of Oncology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Li-Rong Yang
- Department of Oncology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
3
|
Zhang KC, Chu SY, Ding DC. High-grade serous carcinoma of the fallopian tube in a young woman with chromosomal 4q abnormality: A case report. World J Clin Cases 2024; 12:3539-3547. [PMID: 38983400 PMCID: PMC11229890 DOI: 10.12998/wjcc.v12.i18.3539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Few studies have reported an association between an increased risk of acquiring cancers and survival in patients with 4q deletion syndrome. This study presents a rare association between chromosome 4q abnormalities and fallopian tube high-grade serous carcinoma (HGSC) in a young woman. CASE SUMMARY A 35-year-old woman presented with acute dull abdominal pain and a known chromosomal abnormality involving 4q13.3 duplication and 4q23q24 deletion. Upon arrival at the emergency room, her abdomen appeared ovoid and distended with palpable shifting dullness. Ascites were identified through abdominal ultrasound, and computed tomography revealed an omentum cake and an enlarged bilateral adnexa. Blood tests showed elevated CA-125 levels. Paracentesis was conducted, and immunohistochemistry indicated that the cancer cells favored an ovarian origin, making us suspect ovarian cancer. The patient underwent debulking surgery, which led to a diagnosis of stage IIIC HGSC of the fallopian tube. Subsequently, the patient received adjuvant chemotherapy with carboplatin and paclitaxel, resulting in stable current condition. CONCLUSION This study demonstrates a rare correlation between a chromosome 4q abnormality and HGSC. UBE2D3 may affect crucial cancer-related pathways, including P53, BRCA, cyclin D, and tyrosine kinase receptors, thereby possibly contributing to cancer development. In addition, ADH1 and DDIT4 may be potential influencers of both carcinogenic and therapeutic responses.
Collapse
Affiliation(s)
- Kai-Cheng Zhang
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| | - Shao-Yin Chu
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
4
|
Li J, Li X, Zhang Z, Wang S, Huang X, Min L, Li P. Helicobacter pylori promotes gastric fibroblast proliferation and migration by expulsing exosomal miR-124-3p. Microbes Infect 2024; 26:105236. [PMID: 37813158 DOI: 10.1016/j.micinf.2023.105236] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
Gastric fibroblasts (GFs) are direct targets of Helicobacter pylori (H. pylori). GFs infected with H. pylori exhibit marked changes in their morphology and biological behavior. However, the molecular mechanisms by which H. pylori regulates GFs remain unknown. In this study, we cocultured GFs with H. pylori for 48 h. As a result, GFs exhibited an elongated and spindle-shaped morphology. Further, cancer-associated fibroblast (CAF) biomarkers were increased, and related behaviors were significantly enhanced in H. pylori-activated GFs. The number of extracellular vesicles (EVs) secreted by H. pylori-activated GFs remarkably increased. The miR-124-3p level was increased in secreted EVs but decreased in the cytoplasm of H. pylori-activated GFs. Overexpression of miRNA-124-3p in the original GFs significantly suppressed their proliferation and migration. In addition, the migration-promoting effects of H. pylori-activated GFs were suppressed by miR-124-3p and GW4869, which blocked EV generation. Finally, pull-down and luciferase assays revealed that SNAI2 is a target of miR-124-3p. The migration-inhibitory effects of GFs treated with miR-124-3p were eliminated by the overexpression of SNAI2, and the upregulation of SNAI2 in H. pylori-activated GFs was partially alleviated by miR-124-3p or GW4869. Overall, H. pylori infection promotes the proliferation and migration of GFs by accelerating the expulsion of EVs carrying miRNA-124-3p, a SNAI2 inhibitor.
Collapse
Affiliation(s)
- Jun Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China; Department of Gastroenterology, Chui Yang Liu Hospital Affiliated to Tsinghua University, 100020 Beijing, PR China
| | - Xiangji Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China
| | - Zheng Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China
| | - Shidong Wang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China
| | - Xinyuan Huang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China.
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050 Beijing, PR China.
| |
Collapse
|
5
|
Schuhwerk H, Brabletz T. Mutual regulation of TGFβ-induced oncogenic EMT, cell cycle progression and the DDR. Semin Cancer Biol 2023; 97:86-103. [PMID: 38029866 DOI: 10.1016/j.semcancer.2023.11.009] [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: 06/27/2023] [Revised: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
TGFβ signaling and the DNA damage response (DDR) are two cellular toolboxes with a strong impact on cancer biology. While TGFβ as a pleiotropic cytokine affects essentially all hallmarks of cancer, the multifunctional DDR mostly orchestrates cell cycle progression, DNA repair, chromatin remodeling and cell death. One oncogenic effect of TGFβ is the partial activation of epithelial-to-mesenchymal transition (EMT), conferring invasiveness, cellular plasticity and resistance to various noxae. Several reports show that both individual networks as well as their interface affect chemo-/radiotherapies. However, the underlying mechanisms remain poorly resolved. EMT often correlates with TGFβ-induced slowing of proliferation, yet numerous studies demonstrate that particularly the co-activated EMT transcription factors counteract anti-proliferative signaling in a partially non-redundant manner. Collectively, evidence piled up over decades underscore a multifaceted, reciprocal inter-connection of TGFβ signaling / EMT with the DDR / cell cycle progression, which we will discuss here. Altogether, we conclude that full cell cycle arrest is barely compatible with the propagation of oncogenic EMT traits and further propose that 'EMT-linked DDR plasticity' is a crucial, yet intricate facet of malignancy, decisively affecting metastasis formation and therapy resistance.
Collapse
Affiliation(s)
- Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
6
|
Fang M, Wu HK, Pei Y, Zhang Y, Gao X, He Y, Chen G, Lv F, Jiang P, Li Y, Li W, Jiang P, Wang L, Ji J, Hu X, Xiao RP. E3 ligase MG53 suppresses tumor growth by degrading cyclin D1. Signal Transduct Target Ther 2023; 8:263. [PMID: 37414783 PMCID: PMC10326024 DOI: 10.1038/s41392-023-01458-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 04/09/2023] [Accepted: 04/22/2023] [Indexed: 07/08/2023] Open
Abstract
Due to the essential role of cyclin D1 in regulating transition from G1 to S phase in cell cycle, aberrant cyclin D1 expression is a major oncogenic event in many types of cancers. In particular, the dysregulation of ubiquitination-dependent degradation of cyclin D1 contributes to not only the pathogenesis of malignancies but also the refractory to cancer treatment regiments with CDK4/6 inhibitors. Here we show that in colorectal and gastric cancer patients, MG53 is downregulated in more than 80% of tumors compared to the normal gastrointestinal tissues from the same patient, and the reduced MG53 expression is correlated with increased cyclin D1 abundance and inferior survival. Mechanistically, MG53 catalyzes the K48-linked ubiquitination and subsequent degradation of cyclin D1. Thus, increased expression of MG53 leads to cell cycle arrest at G1, and thereby markedly suppresses cancer cell proliferation in vitro as well as tumor growth in mice with xenograft tumors or AOM/DSS induced-colorectal cancer. Consistently, MG53 deficiency results in accumulation of cyclin D1 protein and accelerates cancer cell growth both in culture and in animal models. These findings define MG53 as a tumor suppressor via facilitating cyclin D1 degradation, highlighting the therapeutic potential of targeting MG53 in treating cancers with dysregulated cyclin D1 turnover.
Collapse
Affiliation(s)
- Meng Fang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Hong-Kun Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 310003, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, 310003, Hangzhou, China
| | - Yumeng Pei
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Xiangyu Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Tumor Center, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Yanyun He
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Gengjia Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
| | - Fengxiang Lv
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China
| | - Peng Jiang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
| | - Yumei Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
| | - Wenwen Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China
| | - Peng Jiang
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Tumor Center, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China.
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China.
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, 100871, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China.
- Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, 100871, Beijing, China.
| |
Collapse
|
7
|
Zhang Y, Wang W, Min J, Liu S, Wang Q, Wang Y, Xiao Y, Li X, Zhou Z, Liu S. ZNF451 favors triple-negative breast cancer progression by enhancing SLUG-mediated CCL5 transcriptional expression. Cell Rep 2023; 42:112654. [PMID: 37342906 DOI: 10.1016/j.celrep.2023.112654] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/01/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited effective therapies because of the absence of definitive targets. Here, we demonstrate that the expression of ZNF451, a poorly characterized vertebrate zinc-finger protein, is upregulated in TNBC and associated with a poor prognosis. Elevated ZNF451 expression facilitates TNBC progression by interacting with and enhancing the activity of the transcriptional activator snail family transcriptional repressor 2 (SLUG). Mechanistically, the ZNF451-SLUG complex preferentially recruits the acetyltransferase p300/CBP-associated factor (PCAF) to the CCL5 promoter, selectively facilitating CCL5 transcription by enhancing the acetylation of SLUG and local chromatin, leading to recruitment and activation of tumor-associated macrophages (TAMs). Disturbing the ZNF451-SLUG interaction using a peptide suppresses TNBC progression by reducing CCL5 expression and counteracting the migration and activation of TAMs. Collectively, our work provides mechanistic insights into the oncogene-like functions of ZNF451 and suggests that ZNF451 is a potential target for development of effective therapies against TNBC.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wanyu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Suosi Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, China.
| |
Collapse
|
8
|
Han D, Wang L, Jiang S, Yang Q. The ubiquitin-proteasome system in breast cancer. Trends Mol Med 2023:S1471-4914(23)00096-5. [PMID: 37328395 DOI: 10.1016/j.molmed.2023.05.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Ubiquitin-proteasome system (UPS) is a selective proteolytic system that is associated with the expression or function of target proteins and participates in various physiological and pathological processes of breast cancer. Inhibitors targeting the 26S proteasome in combination with other drugs have shown promising therapeutic effects in the clinical treatment of breast cancer. Moreover, several inhibitors/stimulators targeting other UPS components are also effective in preclinical studies, but have not yet been applied in the clinical treatment of breast cancer. Therefore, it is vital to comprehensively understand the functions of ubiquitination in breast cancer and to identify potential tumor promoters or tumor suppressors among UPS family members, with the aim of developing more effective and specific inhibitors/stimulators targeting specific components of this system.
Collapse
Affiliation(s)
- Dianwen Han
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shan Jiang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Qifeng Yang
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Research Institute of Breast Cancer, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
9
|
Li S, Qian L, Chen Y, Lu H, Tsai H, Lyu H, Yang R, Chen C. Targeting MYO1B impairs tumorigenesis via inhibiting the SNAI2/cyclin D1 signaling in esophageal squamous cell carcinoma. J Cell Physiol 2022; 237:3671-3686. [DOI: doi.org/10.1002/jcp.30831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/06/2022] [Indexed: 09/01/2023]
Abstract
AbstractMyosin‐related proteins play an important role in cancer progression. However, the clinical significance, biological functions, and mechanisms of myosin 1B (MYO1B), in esophageal squamous cell carcinoma (ESCC) remain unclear. The clinical relevance of MYO1B, SNAI2, and cyclin D1 in ESCC was determined by immunohistochemistry, Oncomine, and GEPIA databases. The oncogenic roles of MYO1B were determined by CCK8, colony formation assays, wound healing, and Transwell assay. MYO1B, SNAI2, and cyclin D1 at mRNA and protein levels in ESCC cells were detected by qPCR and Western blot analysis. In our study, we found that MYO1B expression was increased in ESCC tissue samples and correlated with tumor stage, TNM stage, and poor outcomes. Functional assays indicated that depletion of MYO1B impaired oncogenesis, and enhanced chemosensitivity in ESCC. Bioinformatic analysis and mechanistic studies illustrated that SNAI2 was a key downstream effector of MYO1B. Suppression of MYO1B downregulated expression of SNAI2, thereby inhibiting the SNAI2/cyclin D1 pathway. Furthermore, a selective inhibitor of cyclin D1 activation reversed siMYO1B cells overexpressing SNAI2‐elicited aggressive phenotypes of ESCC cells. MYO1B positively correlated with SNAI2 and cyclin D1 in ESCC samples, and higher SNAI2 expression was also associated with poor prognosis in ESCC patients. Our finding demonstrated that MYO1B activates the SNAI2/cyclin D1 pathway to drive tumorigenesis and cisplatin cytotoxicity in ESCC, indicating that MYO1B is a potential therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Shau‐Hsuan Li
- Department of Hematology‐Oncology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Limei Qian
- Department of Medical Oncology Sun Yat‐sen University Cancer Center Guangzhou China
| | - Yen‐Hao Chen
- Department of Hematology‐Oncology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Hung‐I Lu
- Department of Thoracic & Cardiovascular Surgery Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Hsin‐Ting Tsai
- Institute of Medicine Chung Shan Medical University Taichung Taiwan
| | - Haiwen Lyu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease The Sixth Affiliated Hospital of Sun Yat‐sen University Guangzhou China
- Guangdong Research Institute Of Gastroenterology The Sixth Affiliated Hospital of Sun Yat‐sen University Guangzhou China
| | - Runxiang Yang
- The Second Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University Yunnan Cancer Hospital Kunming Yunnan China
| | - Chang‐Han Chen
- Institute of Medicine Chung Shan Medical University Taichung Taiwan
- Department of Medical Research Chung Shan Medical University Hospital Taichung Taiwan
| |
Collapse
|
10
|
Li SH, Qian L, Chen YH, Lu HI, Tsai HT, Lyu H, Yang R, Chen CH. Targeting MYO1B impairs tumorigenesis via inhibiting the SNAI2/cyclin D1 signaling in esophageal squamous cell carcinoma. J Cell Physiol 2022; 237:3671-3686. [PMID: 35861939 DOI: 10.1002/jcp.30831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/09/2022]
Abstract
Myosin-related proteins play an important role in cancer progression. However, the clinical significance, biological functions, and mechanisms of myosin 1B (MYO1B), in esophageal squamous cell carcinoma (ESCC) remain unclear. The clinical relevance of MYO1B, SNAI2, and cyclin D1 in ESCC was determined by immunohistochemistry, Oncomine, and GEPIA databases. The oncogenic roles of MYO1B were determined by CCK8, colony formation assays, wound healing, and Transwell assay. MYO1B, SNAI2, and cyclin D1 at mRNA and protein levels in ESCC cells were detected by qPCR and Western blot analysis. In our study, we found that MYO1B expression was increased in ESCC tissue samples and correlated with tumor stage, TNM stage, and poor outcomes. Functional assays indicated that depletion of MYO1B impaired oncogenesis, and enhanced chemosensitivity in ESCC. Bioinformatic analysis and mechanistic studies illustrated that SNAI2 was a key downstream effector of MYO1B. Suppression of MYO1B downregulated expression of SNAI2, thereby inhibiting the SNAI2/cyclin D1 pathway. Furthermore, a selective inhibitor of cyclin D1 activation reversed siMYO1B cells overexpressing SNAI2-elicited aggressive phenotypes of ESCC cells. MYO1B positively correlated with SNAI2 and cyclin D1 in ESCC samples, and higher SNAI2 expression was also associated with poor prognosis in ESCC patients. Our finding demonstrated that MYO1B activates the SNAI2/cyclin D1 pathway to drive tumorigenesis and cisplatin cytotoxicity in ESCC, indicating that MYO1B is a potential therapeutic target for patients with ESCC.
Collapse
Affiliation(s)
- Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Limei Qian
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yen-Hao Chen
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-I Lu
- Department of Thoracic & Cardiovascular Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsin-Ting Tsai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Haiwen Lyu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Research Institute Of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Runxiang Yang
- The Second Department of Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Chang-Han Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
11
|
Martínez-López A, García-Casas A, Bragado P, Orimo A, Castañeda-Saucedo E, Castillo-Lluva S. Inhibition of RAC1 activity in cancer associated fibroblasts favours breast tumour development through IL-1β upregulation. Cancer Lett 2021; 521:14-28. [PMID: 34419498 DOI: 10.1016/j.canlet.2021.08.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 11/26/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are highly abundant stromal components in the tumour microenvironment. These cells contribute to tumorigenesis and indeed, they have been proposed as a target for anti-cancer therapies. Similarly, targeting the Rho-GTPase RAC1 has also been suggested as a potential therapeutic target in cancer. Here, we show that targeting RAC1 activity, either pharmacologically or by genetic silencing, increases the pro-tumorigenic activity of CAFs by upregulating IL-1β secretion. Moreover, inhibiting RAC1 activity shifts the CAF subtype to a more aggressive phenotype. Thus, as RAC1 suppresses the secretion of IL-1β by CAFs, reducing RAC1 activity in combination with the depletion of this cytokine should be considered as an interesting therapeutic option for breast cancer in which tumour cells retain intact IL-1β signalling.
Collapse
Affiliation(s)
- Angélica Martínez-López
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Mexico
| | - Ana García-Casas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain
| | - Paloma Bragado
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense, Madrid, 28040, Spain
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Mexico
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, 28040, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain.
| |
Collapse
|
12
|
García-Olivares M, Romero-Córdoba S, Ortiz-Sánchez E, García-Becerra R, Segovia-Mendoza M, Rangel-Escareño C, Halhali A, Larrea F, Barrera D. Regulation of anti-tumorigenic pathways by the combinatory treatment of calcitriol and TGF-β in PC-3 and DU145 cells. J Steroid Biochem Mol Biol 2021; 209:105831. [PMID: 33582304 DOI: 10.1016/j.jsbmb.2021.105831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023]
Abstract
Calcitriol and transforming growth factors beta (TGF-β) are involved in several biological pathways such as cell proliferation, differentiation, migration and invasion. Their cellular effects could be similar or opposite depending on the genetic target, cell type and context. Despite the reported association of calcitriol deficiency and disruption of the TGF-β pathway in prostate cancer and the well-known independent effects of calcitriol and TGF-βs on cancer cells, there is limited information regarding the cellular effects of calcitriol and TGF-β in combination. In this study, we in vitro analyze the combinatory effects of calcitriol and TGF-β on cell growth and apoptosis using PC-3 and DU145 human prostate cancer cell lines. Using high-throughput microarray profiling of PC-3 cells upon independent and combinatory treatments, we identified distinct transcriptional landscapes of each intervention, with a higher effect established by the combinatorial treatment, following by TGF-β1 and later by calcitriol. A set of genes and enriched pathways converge among the treatments, mainly between the combinatory scheme and TGF-β1, but the majority were treatment-specific. Of note, CYP24A1, IGFBP3, CDKN1A, NOX4 and UBE2D3 were significantly up-regulated upon the combinatorial treatment whereas CCNA1, members of the CT45A and APOBEC3 family were down-regulated. By public RNA signatures, we were able to confirm the regulation by the co-treatment over cell proliferation and cell cycle. We finally investigated the possible clinical impact of genes modulated by the combinatorial treatment using benchmark prostate cancer data. This comprehensive analysis reveals that the combinatory treatment impairs cell growth without affecting apoptosis and their combinatory actions might synergize and improved their individual effects to reprogram prostate cancer signaling.
Collapse
Affiliation(s)
- Mitzi García-Olivares
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México, 14080, México
| | - Sandra Romero-Córdoba
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, México; Departamento de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México, 14080, México
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Ciudad de México, México
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Mariana Segovia-Mendoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, 04510, México
| | - Claudia Rangel-Escareño
- Laboratorio de Genómica Computacional y Biología Integrativa, Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Ciudad de México, 14610, México; Departamento de Ingeniería y Ciencias, Tecnológico de Monterrey, Epigmenio González 500, Soriana, 76140 Santiago de Querétaro, Qro. México
| | - Ali Halhali
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México, 14080, México
| | - Fernando Larrea
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México, 14080, México
| | - David Barrera
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México, 14080, México.
| |
Collapse
|
13
|
Walters TS, McIntosh DJ, Ingram SM, Tillery L, Motley ED, Arinze IJ, Misra S. SUMO-Modification of Human Nrf2 at K 110 and K 533 Regulates Its Nucleocytoplasmic Localization, Stability and Transcriptional Activity. Cell Physiol Biochem 2021; 55:141-159. [PMID: 33770425 PMCID: PMC8279473 DOI: 10.33594/000000351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIMS Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that binds to the antioxidant response element(s) (ARE) in target gene promoters, enabling oxidatively stressed cells to respond in order to restore redox homeostasis. Post-translational modifications (PTMs) that mediate activation of Nrf2, in the cytosol and its release from Keap1, have been extensively studied but PTMs that impact its biology after activation are beginning to emerge. In this regard, PTMs like acetylation, phosphorylation, ubiquitination and sumoylation contribute towards the Nrf2 subcellular localization, and its transactivation function. We previously demonstrated that Nrf2 traffics to the promyelocytic leukemia-nuclear bodies (PML-NB), where it is a target for modification by small ubiquitin-like modifier (SUMO) proteins (sumoylation), but the site(s) for SUMO conjugation have not been determined. In this study, we aim to identify SUMO-2 conjugation site(s) and explore the impact, sumoylation of the site(s) have on Nrf2 stability, nuclear localization and transcriptional activation of its target gene expression upon oxidative stress. METHODS The putative SUMO-binding sites in Nrf2 for human isoform1 (NP_006155.2) and mouse homolog (NP_035032.1) were identified using a computer-based SUMO-predictive software (SUMOplot™). Site-directed mutagenesis, immunoblot analysis, and ARE-mediated reporter gene assays were used to assess the impact of sumoylation on these site(s) in vitro. Effect of mutation of these sumoylation sites of Nrf2 on expression of Heme Oxygenase1 (HO-1) was determined in HEK293T cell. RESULTS
Eight putative sumoylation sites were identified by SUMOplot™ analysis. Out of the eight predicted sites only one 532LKDE535 of human (h) and its homologous 524LKDE527 of mouse (m) Nrf2, exactly matches the SUMO-binding consensus motif. The other high probability SUMO-acceptor site identified was residue K110, in the motifs 109PKSD112 and 109PKQD112 of human and mouse Nrf2, respectively. Mutational analysis of putative sumoylation sites (human (h)/mouse (m)
K110, hK533 and mK525) showed that these residues are needed for SUMO-2 conjugation, nuclear localization and ARE driven transcription of reporter genes and the endogenous HO-1 expression by Nrf2. These residues also stabilized Nrf2, as evident from shorter half-lives of the mutant protein compared to wild-type Nrf2. CONCLUSION Our findings indicate that SUMO-2
mediated sumoylation of K110 and K533 in human Nrf2 regulates in part its transcriptional activity by enhancing its stabilization and nuclear localization.
Collapse
Affiliation(s)
- Treniqka S Walters
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Deneshia J McIntosh
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Lakeisha Tillery
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Evangeline D Motley
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Ifeanyi J Arinze
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Smita Misra
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA,
- School of Graduate Studies and Research, Meharry Medical College, Nashville TN, USA
- Center for Women's Health, Meharry Medical College, Nashville TN, USA
| |
Collapse
|
14
|
Du X, Song H, Shen N, Hua R, Yang G. The Molecular Basis of Ubiquitin-Conjugating Enzymes (E2s) as a Potential Target for Cancer Therapy. Int J Mol Sci 2021; 22:ijms22073440. [PMID: 33810518 PMCID: PMC8037234 DOI: 10.3390/ijms22073440] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 01/06/2023] Open
Abstract
Ubiquitin-conjugating enzymes (E2s) are one of the three enzymes required by the ubiquitin-proteasome pathway to connect activated ubiquitin to target proteins via ubiquitin ligases. E2s determine the connection type of the ubiquitin chains, and different types of ubiquitin chains regulate the stability and activity of substrate proteins. Thus, E2s participate in the regulation of a variety of biological processes. In recent years, the importance of E2s in human health and diseases has been particularly emphasized. Studies have shown that E2s are dysregulated in variety of cancers, thus it might be a potential therapeutic target. However, the molecular basis of E2s as a therapeutic target has not been described systematically. We reviewed this issue from the perspective of the special position and role of E2s in the ubiquitin-proteasome pathway, the structure of E2s and biological processes they are involved in. In addition, the inhibitors and microRNAs targeting E2s are also summarized. This article not only provides a direction for the development of effective drugs but also lays a foundation for further study on this enzyme in the future.
Collapse
|
15
|
Yastrebova MA, Khamidullina AI, Tatarskiy VV, Scherbakov AM. Snail-Family Proteins: Role in Carcinogenesis and Prospects for Antitumor Therapy. Acta Naturae 2021; 13:76-90. [PMID: 33959388 PMCID: PMC8084295 DOI: 10.32607/actanaturae.11062] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
The review analyzes Snail family proteins, which are transcription factors involved in the regulation of the epithelial-mesenchymal transition (EMT) of tumor cells. We describe the structure of these proteins, their post-translational modification, and the mechanisms of Snail-dependent regulation of genes. The role of Snail proteins in carcinogenesis, invasion, and metastasis is analyzed. Furthermore, we focus on EMT signaling mechanisms involving Snail proteins. Next, we dissect Snail signaling in hypoxia, a condition that complicates anticancer treatment. Finally, we offer classes of chemical compounds capable of down-regulating the transcriptional activity of Snails. Given the important role of Snail proteins in cancer biology and the potential for pharmacological inhibition, Snail family proteins may be considered promising as therapeutic targets.
Collapse
Affiliation(s)
- M. A. Yastrebova
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - A. I. Khamidullina
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
| | - V. V. Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334 Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| | - A. M. Scherbakov
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| |
Collapse
|
16
|
Blanco-Gómez A, Hontecillas-Prieto L, Corchado-Cobos R, García-Sancha N, Salvador N, Castellanos-Martín A, Sáez-Freire MDM, Mendiburu-Eliçabe M, Alonso-López D, De Las Rivas J, Lorente M, García-Casas A, Del Carmen S, Abad-Hernández MDM, Cruz-Hernández JJ, Rodríguez-Sánchez CA, Claros-Ampuero J, García-Cenador B, García-Criado J, Orimo A, Gridley T, Pérez-Losada J, Castillo-Lluva S. Stromal SNAI2 Is Required for ERBB2 Breast Cancer Progression. Cancer Res 2020; 80:5216-5230. [PMID: 33023950 DOI: 10.1158/0008-5472.can-20-0278] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/07/2020] [Accepted: 10/01/2020] [Indexed: 12/13/2022]
Abstract
SNAI2 overexpression appears to be associated with poor prognosis in breast cancer, yet it remains unclear in which breast cancer subtypes this occurs. Here we show that excess SNAI2 is associated with a poor prognosis of luminal B HER2+/ERBB2+ breast cancers in which SNAI2 expression in the stroma but not the epithelium correlates with tumor proliferation. To determine how stromal SNAI2 might influence HER2+ tumor behavior, Snai2-deficient mice were crossed with a mouse line carrying the ErbB2/Neu protooncogene to generate HER2+/ERBB2+ breast cancer. Tumors generated in this model expressed SNAI2 in the stroma but not the epithelium, allowing for the role of stromal SNAI2 to be studied without interference from the epithelial compartment. The absence of SNAI2 in the stroma of HER2+/ERBB2+ tumors is associated with: (i) lower levels of cyclin D1 (CCND1) and reduced tumor epithelium proliferation; (ii) higher levels of AKT and a lower incidence of metastasis; (iii) lower levels of angiopoietin-2 (ANGPT2), and more necrosis. Together, these results indicate that the loss of SNAI2 in cancer-associated fibroblasts limits the production of some cytokines, which influences AKT/ERK tumor signaling and subsequent proliferative and metastatic capacity of ERBB2+ breast cancer cells. Accordingly, SNAI2 expression in the stroma enhanced the tumorigenicity of luminal B HER2+/ERBB2+ breast cancers. This work emphasizes the importance of stromal SNAI2 in breast cancer progression and patients' prognosis. SIGNIFICANCE: Stromal SNAI2 expression enhances the tumorigenicity of luminal B HER2+ breast cancers and can identify a subset of patients with poor prognosis, making SNAI2 a potential therapeutic target for this disease. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/23/5216/F1.large.jpg.
Collapse
Affiliation(s)
- Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Lourdes Hontecillas-Prieto
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Nélida Salvador
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, Spain.,Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Andrés Castellanos-Martín
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - María Del Mar Sáez-Freire
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Marina Mendiburu-Eliçabe
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Diego Alonso-López
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain
| | - Javier De Las Rivas
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain.,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Mar Lorente
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, Spain.,Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Ana García-Casas
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, Spain.,Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Sofía Del Carmen
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Anatomía Patológica, Universidad de Salamanca, Salamanca, Spain.,Departamento de Anatomía Patológica, Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - María Del Mar Abad-Hernández
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Anatomía Patológica, Universidad de Salamanca, Salamanca, Spain.,Departamento de Anatomía Patológica, Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Juan Jesús Cruz-Hernández
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - César Augusto Rodríguez-Sánchez
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Servicio de Oncología, Hospital Universitario de Salamanca, Salamanca, Spain
| | | | - Begoña García-Cenador
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Cirugía, Universidad de Salamanca, Salamanca, Spain
| | - Javier García-Criado
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain.,Departamento de Cirugía, Universidad de Salamanca, Salamanca, Spain
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Thomas Gridley
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, Salamanca, Spain. .,Instituto de Investigación Biosanitaria de Salamanca (IBSAL), Salamanca, Spain
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular; Facultad de Ciencias Químicas y Biológicas, Universidad Complutense, Madrid, Spain. .,Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
17
|
Fan H, Wang X, Li W, Shen M, Wei Y, Zheng H, Kang Y. ASB13 inhibits breast cancer metastasis through promoting SNAI2 degradation and relieving its transcriptional repression of YAP. Genes Dev 2020; 34:1359-1372. [PMID: 32943576 PMCID: PMC7528707 DOI: 10.1101/gad.339796.120] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022]
Abstract
In this study, Fan et al. studied the post-translational regulation of transcription factor SNAI2, which plays key roles during development and promotes metastasis by inducing invasive phenotype and tumor-initiating activity of cancer cells. They performed a dual-luciferase-based, genome-wide E3 ligase siRNA library screen and identified ASB13 as an E3 ubiquitin ligase that targets SNAI2 for ubiquitination and degradation, thereby establishing ASB13 as a suppressor of breast cancer metastasis. Transcription factor SNAI2 plays key roles during development and has also been known to promote metastasis by inducing invasive phenotype and tumor-initiating activity of cancer cells. However, the post-translational regulation of SNAI2 is less well studied. We performed a dual-luciferase-based, genome-wide E3 ligase siRNA library screen and identified ASB13 as an E3 ubiquitin ligase that targets SNAI2 for ubiquitination and degradation. ASB13 knockout in breast cancer cells promoted cell migration and decreased F-actin polymerization, while overexpression of ASB13 suppressed lung metastasis. Furthermore, ASB13 knockout decreased YAP expression, and such regulation is dependent on an increased protein level of SNAI2, which in turn represses YAP transcription. YAP suppresses tumor progression in breast cancer, as YAP knockout increases tumorsphere formation, anchorage-independent colony formation, cell migration in vitro, and lung metastasis in vivo. Clinical data analysis reveals that ASB13 expression is positively correlated with improved overall survival in breast cancer patients. These findings establish ASB13 as a suppressor of breast cancer metastasis by promoting degradation of SNAI2 and relieving its transcriptional repression of YAP.
Collapse
Affiliation(s)
- Huijuan Fan
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xuxiang Wang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wenyang Li
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Minhong Shen
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Yong Wei
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Hanqiu Zheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA.,Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| |
Collapse
|
18
|
Zhou Y, Chen R, Luo X, Zhang WD, Qin JJ. The E2 ubiquitin-conjugating enzyme UbcH5c: an emerging target in cancer and immune disorders. Drug Discov Today 2020; 25:S1359-6446(20)30369-X. [PMID: 32947046 DOI: 10.1016/j.drudis.2020.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/14/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Ubiquitination is a crucial post-translational modification (PTM) of proteins and regulates their stabilities and activities, thereby modulating multiple signaling pathways. UbcH5c, a member of the UbcH5 ubiquitin-conjugating enzyme (E2) protein family, engages in the ubiquitination of dozens of proteins and regulates nuclear factor kappa-B (NF-κB), p53 tumor suppressor, and several other essential signaling pathways. UbcH5c has been reported to be abnormally expressed in human cancer and immune disorders and is involved in the initiation and progression of these diseases. In this review, we mainly focus on UbcH5c structure, activity, signaling pathways, and its relevance to cancer and immune disorders. We end by integrating all known factors relating to UbcH5c inhibition as a potential cancer therapy method, and discuss associated challenges.
Collapse
Affiliation(s)
- Yuan Zhou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Runzhe Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaofang Luo
- College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China
| | - Wei-Dong Zhang
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; Institute of Cancer and Basic Medicine, Chinese Academy of Sciences; Cancer Hospital of the University of Chinese Academy of Sciences; Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
19
|
Wang T, Wang J, Ren W, Chen S, Cheng YF, Zhang XM. CircRNA-0008717 promotes cell proliferation, migration, and invasion by regulating miR-203/Slug in esophageal cancer cells. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:999. [PMID: 32953799 PMCID: PMC7475474 DOI: 10.21037/atm-20-5205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Esophageal cancer (EC) is one of the deadliest cancers worldwide. Circular RNAs (circRNAs) have been implicated in the regulation of multiple human diseases, including cancer. In particular, the dysregulation of circRNA-0008717 has been linked to multiple types of cancer. However, the clinical significance and the molecular mechanisms of circRNA-0008717 in EC need to be further investigated. Therefore, this study aimed to prove the role of circRNA-0008717 in EC and its underlying molecular mechanism of action. Methods The expression of circRNA-0008717, miR-203, and the Slug was measured in two EC cell lines (EC109 and KYSE-150) by qRT-RCR. EC109 and KYSE-150 cells were first transfected with circRNA-0008717 siRNA (si-circRNA). After that, the proliferation, apoptosis, migration, and invasion of EC109 and KYSE-150 cells were measured. The western blot detected Slug, Vimentin, and E-cadherin protein levels. A dual-luciferase reporter gene assay was used to set up the interactions among circRNA-0008717, miR-203, and Slug. Results circRNA-0008717 expression was significantly upregulated in EC cells, and miR-2031 expression was decreased. Moreover, si-circRNA-0008717 or si-Slug inhibited the proliferation, migration, and invasion of EC cells. We found that circRNA-0008717 functioned as a sponge of miR-203, resulting in increased expression of Slug. We also reversed the effect of circRNA-0008717 knockdown on the EC progression by co-transfecting EC cells with a miR-203 inhibitor or Slug. Conclusions The proliferation, invasion, and migration of EC cells were enhanced by circRNA-0008717 sponging the miR-203 to increase Slug expression.
Collapse
Affiliation(s)
- Tao Wang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Wang
- Department of Ultrasound, Shandong Province Coal Taishan Sanatorium, Taian, China
| | - Wei Ren
- Department of Radiotherapy, the People's Hospital of Lanling County, Linyi, China
| | - Shuai Chen
- Institute of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Feng Cheng
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiao-Mei Zhang
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
20
|
Dai X, Xin Y, Xu W, Tian X, Wei X, Zhang H. CBP-mediated Slug acetylation stabilizes Slug and promotes EMT and migration of breast cancer cells. SCIENCE CHINA-LIFE SCIENCES 2020; 64:563-574. [PMID: 32737855 DOI: 10.1007/s11427-020-1736-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/16/2020] [Indexed: 12/24/2022]
Abstract
Slug, a member of the Snail family of transcriptional repressors, plays a key role in cancer progression, cellular plasticity, and epithelial to mesenchymal transition (EMT). Slug is a fast-turnover protein and its stability is controlled by post-translational modifications. Here, we identified that Slug is acetylated by acetyltransferase CREB-binding protein (CBP) in breast cancer cells. CBP directly interacts with the C-terminal domain of Slug through its catalytic histone acetyltransferase (HAT) domain, leading to acetylation of Slug at lysines 166 and 211. Analysis with acetylation-specific antibodies revealed that Slug is highly acetylated in metastatic breast cancer cells. Notably, Slug acetylation, mediated by CBP at lysines 166 and 211, doubles its half-life and increases its stability. Further, acetylated Slug downregulates the expression of E-cadherin, the epithelial marker, and upregulates the expression of N-cadherin and vimentin, thereby promoting breast cancer cell migration. In conclusion, the present study demonstrates that CBP-mediated Slug acetylation increases its stability, promoting EMT and migration of breast cancer cells.
Collapse
Affiliation(s)
- Xiaoyan Dai
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.,Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yanli Xin
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Weizhi Xu
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xinxia Tian
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Xiaofan Wei
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Hongquan Zhang
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
21
|
Roman-Trufero M, Ito CM, Pedebos C, Magdalou I, Wang YF, Karimi MM, Moyon B, Webster Z, di Gregorio A, Azuara V, Khalid S, Speck C, Rodriguez T, Dillon N. Evolution of an Amniote-Specific Mechanism for Modulating Ubiquitin Signaling via Phosphoregulation of the E2 Enzyme UBE2D3. Mol Biol Evol 2020; 37:1986-2001. [PMID: 32145025 PMCID: PMC7306689 DOI: 10.1093/molbev/msaa060] [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] [Indexed: 11/13/2022] Open
Abstract
Genetic variation in the enzymes that catalyze posttranslational modification of proteins is a potentially important source of phenotypic variation during evolution. Ubiquitination is one such modification that affects turnover of virtually all of the proteins in the cell in addition to roles in signaling and epigenetic regulation. UBE2D3 is a promiscuous E2 enzyme, which acts as an ubiquitin donor for E3 ligases that catalyze ubiquitination of developmentally important proteins. We have used protein sequence comparison of UBE2D3 orthologs to identify a position in the C-terminal α-helical region of UBE2D3 that is occupied by a conserved serine in amniotes and by alanine in anamniote vertebrate and invertebrate lineages. Acquisition of the serine (S138) in the common ancestor to modern amniotes created a phosphorylation site for Aurora B. Phosphorylation of S138 disrupts the structure of UBE2D3 and reduces the level of the protein in mouse embryonic stem cells (ESCs). Substitution of S138 with the anamniote alanine (S138A) increases the level of UBE2D3 in ESCs as well as being a gain of function early embryonic lethal mutation in mice. When mutant S138A ESCs were differentiated into extraembryonic primitive endoderm, levels of the PDGFRα and FGFR1 receptor tyrosine kinases were reduced and primitive endoderm differentiation was compromised. Proximity ligation analysis showed increased interaction between UBE2D3 and the E3 ligase CBL and between CBL and the receptor tyrosine kinases. Our results identify a sequence change that altered the ubiquitination landscape at the base of the amniote lineage with potential effects on amniote biology and evolution.
Collapse
Affiliation(s)
- Monica Roman-Trufero
- Gene Regulation and Chromatin Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Constance M Ito
- Gene Regulation and Chromatin Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Conrado Pedebos
- Department of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Indiana Magdalou
- DNA Replication Group, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Yi-Fang Wang
- Bioinformatics and Computing, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Mohammad M Karimi
- Bioinformatics and Computing, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Benjamin Moyon
- Transgenics and ES Cell Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Zoe Webster
- Transgenics and ES Cell Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Aida di Gregorio
- BHF Centre for Research Excellence, National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Veronique Azuara
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Syma Khalid
- Department of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Christian Speck
- DNA Replication Group, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Tristan Rodriguez
- BHF Centre for Research Excellence, National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Niall Dillon
- Gene Regulation and Chromatin Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| |
Collapse
|
22
|
Abstract
The transcription factor Snai2, encoded by the SNAI2 gene, is an evolutionarily conserved C2H2 zinc finger protein that orchestrates biological processes critical to tissue development and tumorigenesis. Initially characterized as a prototypical epithelial-to-mesenchymal transition (EMT) transcription factor, Snai2 has been shown more recently to participate in a wider variety of biological processes, including tumor metastasis, stem and/or progenitor cell biology, cellular differentiation, vascular remodeling and DNA damage repair. The main role of Snai2 in controlling such processes involves facilitating the epigenetic regulation of transcriptional programs, and, as such, its dysregulation manifests in developmental defects, disruption of tissue homeostasis, and other disease conditions. Here, we discuss our current understanding of the molecular mechanisms regulating Snai2 expression, abundance and activity. In addition, we outline how these mechanisms contribute to disease phenotypes or how they may impact rational therapeutic targeting of Snai2 dysregulation in human disease.
Collapse
Affiliation(s)
- Wenhui Zhou
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kayla M Gross
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Charlotte Kuperwasser
- Department of Developmental, Molecular & Chemical Biology, Sackler School of Graduate Biomedical Sciences, Boston, MA 02111, USA
- Raymond and Beverly Sackler Convergence Laboratory, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
23
|
Zhang F, Cui Y. Dysregulation of DNA methylation patterns may identify patients with breast cancer resistant to endocrine therapy: A predictive classifier based on differentially methylated regions. Oncol Lett 2019; 18:1287-1303. [PMID: 31423189 PMCID: PMC6607238 DOI: 10.3892/ol.2019.10405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
Endocrine therapy (ET) is one of a number of targeted therapies for estrogen receptor-positive breast cancer (BRCA); however, resistance to ET has become the primary issue affecting treatment outcome. In the present study, a predictive classifier was created using a DNA methylation dataset to identify patients susceptible to endocrine resistance. DNA methylation and RNA sequencing data, and the clinicopathological features of BRCA, were obtained from The Cancer Genome Atlas. Stringent criteria were set to select and classify patients into two groups, namely those resistant to ET (n=11) and sensitive to ET (n=21) groups. Bump hunting analysis revealed that 502 out of 135,418 genomic regions were differentially methylated between these two groups; these regions were differentially methylated regions (DMRs). The majority of the CpG sites contained in the DMRs mapped to the promoter region. Functional enrichment analyses indicated that a total of 562 specific genes encompassing these DMRs were primarily associated with 'biological progress of organ morphogenesis and development' and 'cell-cell adhesion' gene ontologies. Logistic regression and Pearson's correlation analysis were conducted to construct a predictive classifier for distinguishing patients resistant or sensitive to ET. The highest areas under the curve and relatively low Akaike information criterion values were associated with a total of 60 DMRs; a risk score retained from this classifier was revealed to be an unfavorable predictor of survival in two additional independent datasets. Furthermore, the majority of genes (55/63) exhibited a statistically significant association between DNA methylation and mRNA expression (P<0.05). The association between the mRNA expression of a number of genes (namely calcium release activated channel regulator 2A, Schlafen family member 12, chromosome 3 open reading frame 18, zinc finger protein 880, dual oxidase 1, major histocompatibility complex, class II, DP β1, C-terminal binding protein 1, ALG13 UDP-N-acetylglucosaminyltransferase subunit and RAS protein activator like 2) and the prognosis of patients with estrogen receptor-positive BRCA and ET resistance was determined using Kaplan-Meier Plotter. In summary, the predictive classifier proposed in the present study may aid the identification of patients sensitive or resistant to ET, and numerous genes maybe potential therapeutic targets to delay the development of resistance to ET.
Collapse
Affiliation(s)
- Fan Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yukun Cui
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
- Correspondence to: Professor Yukun Cui, Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, Guangdong 515041, P.R. China, E-mail:
| |
Collapse
|
24
|
Wang W, Hind T, Lam BWS, Herr DR. Sphingosine 1–phosphate signaling induces SNAI2 expression to promote cell invasion in breast cancer cells. FASEB J 2019; 33:7180-7191. [DOI: 10.1096/fj.201801635r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wei Wang
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Tatsuma Hind
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Department of PharmacologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Brenda Wan Shing Lam
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Deron R. Herr
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore
- Department of BiologySan Diego State UniversitySan DiegoCaliforniaUSA
| |
Collapse
|
25
|
Assani G, Zhou Y. Effect of modulation of epithelial-mesenchymal transition regulators Snail1 and Snail2 on cancer cell radiosensitivity by targeting of the cell cycle, cell apoptosis and cell migration/invasion. Oncol Lett 2018; 17:23-30. [PMID: 30655734 PMCID: PMC6313178 DOI: 10.3892/ol.2018.9636] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of cancer-associated mortality worldwide. Several strategies of treatment, including radiotherapy, have been developed and used to treat this disease. However, post-treatment metastasis and resistance to treatment are two major causes for the limited effectiveness of radiotherapy in cancer patients. Epithelial-mesenchymal transition (EMT) is regulated by SNAIL family transcription factors, including Snail1 and Snail2 (Slug), and serves important roles in progression and cancer resistance to treatment. Snail1 and Slug also have been shown to be implicated in cancer treatment resistance. For resolving the resistance to treatment problems, combining the modulation of gene expression with radiotherapy is a novel strategy to treat patients with cancer. The present review focuses on the effect of Snail1 and Slug on cancer radiosensitivity by targeting cell apoptosis, the cell cycle and cell migration/invasion.
Collapse
Affiliation(s)
- Ganiou Assani
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Hubei Key Laboratory of Tumor Biology Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yunfeng Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China.,Hubei Key Laboratory of Tumor Biology Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
26
|
Srivastava K, Pickard A, Craig SG, Quinn GP, Lambe SM, James JA, McDade SS, McCance DJ. ΔNp63γ/SRC/Slug Signaling Axis Promotes Epithelial-to-Mesenchymal Transition in Squamous Cancers. Clin Cancer Res 2018; 24:3917-3927. [PMID: 29739791 PMCID: PMC6098695 DOI: 10.1158/1078-0432.ccr-17-3775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/28/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
Purpose: To investigate the regulation of epithelial-to-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC) and its importance in tumor invasion.Experimental Design: We use a three-dimensional invasive organotypic raft culture model of human foreskin keratinocytes expressing the E6/E7 genes of the human papilloma virus-16, coupled with bioinformatic and IHC analysis of patient samples to investigate the role played by EMT in invasion and identify effectors and upstream regulatory pathways.Results: We identify SNAI2 (Slug) as a critical effector of EMT-activated downstream of TP63 overexpression in HNSCC. Splice-form-specific depletion and rescue experiments further identify the ΔNp63γ isoform as both necessary and sufficient to activate the SRC signaling axis and SNAI2-mediated EMT and invasion. Moreover, elevated SRC levels are associated with poor outcome in patients with HNSCC in The Cancer Genome Atlas dataset. Importantly, the effects on EMT and invasions and SNAI2 expression can be reversed by genetic or pharmacologic inhibition of SRC.Conclusions: Overexpression of ΔNp63γ modulates cell invasion by inducing targetable SRC-Slug-evoked EMT in HNSCC, which can be reversed by inhibitors of the SRC signaling. Clin Cancer Res; 24(16); 3917-27. ©2018 AACR.
Collapse
Affiliation(s)
- Kirtiman Srivastava
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | - Adam Pickard
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
- The Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Stephanie G Craig
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Gerard P Quinn
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Shauna M Lambe
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Jacqueline A James
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | - Dennis J McCance
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
27
|
Lee SJ, Lee JS, Lee E, Lim TG, Byun S. The ginsenoside metabolite compound K inhibits hormone-independent breast cancer through downregulation of cyclin D1. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.04.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
28
|
Gao P, Wang S, Jing F, Zhan J, Wang Y. microRNA-203 suppresses invasion of gastric cancer cells by targeting ERK1/2/Slug/ E-cadherin signaling. Cancer Biomark 2018; 19:11-20. [PMID: 28269747 DOI: 10.3233/cbm-160167] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Growing evidence suggests that microRNA plays an essential role in the development and metastasis of many tumors, including gastric cancer (GC). Expression of microRNA-203 (miR-203) has been reported to decrease in GC. In addition, overexpression of miR-203 inhibits grow of GC cells in vitro and in vivo. However, whether miR-203 affects cell migration and invasion of GC remains unclear. This study aimed to reveal the role of miR-203 on migration and invasion of GC, and its potential mechanisms. METHODS Synthetic pre-miR-203 (miR-203), anti-miR-203 and scrambled negative control RNAs was transfected into the gastric cancer SGC7901 cells to generate miR-203 or anti-miR-203-transfected stable clones. The roles of miR-203 on cell invasion and motility were then analyzed by Transwell migration assay and Wound healing assay in vitro. Using siRNA to targeting ERK1/2, Slug, and E-cadherin or Slug cDNA transfection (to increase Slug expression) to examine the miR-203 signaling pathway. We also examined the efficacies of miR-203 or anti-miR-203 on peritoneal metastasis of SGC7901 cells in the nude mouse model. RESULTS Overexpression of miR-203 inhibits SGC7901 cell invasion and motility, followed by decreased phospho-ERK1/2 (pERK1/2) and Slug expression, as well as increased E-cadherin expression. Re-expression of Slug in miR-203/SGC7901cells decreased E-cadherin expression and restored the invasive phenotypes. Targeting E-cadherin in miR-203/SGC7901cells also restored the invasive phenotypes. Inhibition of miR-203 promotes SGC7901 cell invasion and motility, followed by increased phospho-ERK1/2 (pERK1/2) and Slug expression, as well as decreased E-cadherin expression. Targeting ERK1/2 or Slug in anti-miR-203/SGC7901cells increased E-cadherin expression and reversed the invasive phenotypes. In addition, targeting ERK1/2 decreased Slug and increased the E-cadherin expression. Significantly, we found that miR-203 could exert marked inhibition of the peritoneal metastasis of SGC7901 in nude mice in vivo. Targeting miR-203 could exert marked promotion of the peritoneal metastasis of SGC7901 in nude mice in vivo. CONCLUSIONS miR-203/ERK1/2/Slug/E-cadherin signaling pathway plays an essential role on SGC7901 cell invasion and motility. miR-203 can be novel modalities to prevent peritoneal metastasis of invasive cancers such as gastric cancer.
Collapse
Affiliation(s)
- Peng Gao
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shijie Wang
- Department of Gastroenterology, People's Hospital of Juxian, Rizhao, Shandong, China
| | - Fuchun Jing
- Department of General Surgery, The Second Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jiang Zhan
- Department of General Surgery, The Second Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yunhui Wang
- Department of General Surgery, The Second Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
29
|
Weidle UH, Dickopf S, Hintermair C, Kollmorgen G, Birzele F, Brinkmann U. The Role of micro RNAs in Breast Cancer Metastasis: Preclinical Validation and Potential Therapeutic Targets. Cancer Genomics Proteomics 2018; 15:17-39. [PMID: 29275360 PMCID: PMC5822183 DOI: 10.21873/cgp.20062] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/18/2022] Open
Abstract
Despite the approval of several molecular therapies in the last years, breast cancer-associated death ranks as the second highest in women. This is due to metastatic disease, which represents a challenge for treatment. A better understanding of the molecular mechanisms of metastasis is, therefore, of paramount importance. In this review we summarize the role of micro RNAs (miRs) involved in metastasis of breast cancer. We present an overview on metastasis-promoting, -suppressing and context-dependent miRs with both activities. We have categorized the corresponding miRs according to their target classes, interaction with stromal cells or exosomes. The pathways affected by individual miRs are outlined in regard to in vitro properties, activity in metastasis-related in vivo models and clinical significance. Current approaches that may be suitable for therapeutic inhibition or restauration of miR activity are outlined. Finally, we discuss the delivery bottlenecks which present as a major challenge in nucleic acid (miR)-based therapies.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Steffen Dickopf
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Gwendlyn Kollmorgen
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
30
|
Shen CJ, Kuo YL, Chen CC, Chen MJ, Cheng YM. MMP1 expression is activated by Slug and enhances multi-drug resistance (MDR) in breast cancer. PLoS One 2017; 12:e0174487. [PMID: 28334049 PMCID: PMC5363985 DOI: 10.1371/journal.pone.0174487] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 03/09/2017] [Indexed: 11/18/2022] Open
Abstract
High matrix metalloproteinase 1 (MMP1) expression is associated with enhanced breast cancer growth and metastasis and also might predict poor prognosis. In this study, we further investigated the functional role of MMP1 and how it is upregulated in multi-drug resistant (MDR) breast cancer cells. By retrieving microarray data in GEO datasets and the survival data in the Kaplan Meier plotter, we observed that MMP1 is significantly upregulated in MCF-7/ADR cells compared to the parental MCF-7 cells, while high MMP1 expression is associated with worse overall survival (OS) and recurrence free survival (RFS) in breast cancer patients after systematic therapy. Functional studies showed that MMP1 overexpression significantly reduced the drug sensitivity in MCF-7 cells, while MMP1 knockdown substantially enhanced the sensitivity in MCF-7/ADR cells. By performing western blotting and immunofluorescent staining, we confirmed that MCF-7/ADR cells had enhanced mesenchymal properties than MCF-7 cells. In MCF-7 cells, enforced Slug expression resulted in significant MMP1 upregulation, while in MCF-7/ADR cells, Slug knockdown led to reduced MMP1 expression. By performing bioinformatic analysis, we observed that the promoter of MMP1 has three putative Slug binding sites. The following dual luciferase assay and ChIP-qPCR verified these three binding sites. Therefore, we infer that Slug enhances MMP1 transcription via directly binding to the promoter region in breast cancer cells, which is a previously unrecognized mechanism in the development of MDR.
Collapse
Affiliation(s)
- Ching-Ju Shen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Ling Kuo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Gynecology and Obstetrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chung Chen
- Department of Plastic and Reconstruction Surgery, E-Da Hospital, Kaohsiung, Taiwan
| | - Ming-Jenn Chen
- Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Sports Management, College of Leisure and Recreation Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Ya-Min Cheng
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
31
|
De Luca P, Dalton GN, Scalise GD, Moiola CP, Porretti J, Massillo C, Kordon E, Gardner K, Zalazar F, Flumian C, Todaro L, Vazquez ES, Meiss R, De Siervi A. CtBP1 associates metabolic syndrome and breast carcinogenesis targeting multiple miRNAs. Oncotarget 2017; 7:18798-811. [PMID: 26933806 PMCID: PMC4951330 DOI: 10.18632/oncotarget.7711] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/11/2016] [Indexed: 12/22/2022] Open
Abstract
Metabolic syndrome (MeS) has been identified as a risk factor for breast cancer. C-terminal binding protein 1 (CtBP1) is a co-repressor of tumor suppressor genes that is activated by low NAD+/NADH ratio. High fat diet (HFD) increases intracellular NADH. We investigated the effect of CtBP1 hyperactivation by HFD intake on mouse breast carcinogenesis. We generated a MeS-like disease in female mice by chronically feeding animals with HFD. MeS increased postnatal mammary gland development and generated prominent duct patterns with markedly increased CtBP1 and Cyclin D1 expression. CtBP1 induced breast cancer cells proliferation. Serum from animals with MeS enriched the stem-like/progenitor cell population from breast cancer cells. CtBP1 increased breast tumor growth in MeS mice modulating multiple genes and miRNA expression implicated in cell proliferation, progenitor cells phenotype, epithelial to mesenchymal transition, mammary development and cell communication in the xenografts. These results define a novel function for CtBP1 in breast carcinogenesis.
Collapse
Affiliation(s)
- Paola De Luca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Guillermo N Dalton
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Georgina D Scalise
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Cristian P Moiola
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Juliana Porretti
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Cintia Massillo
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Edith Kordon
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), and Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET, Buenos Aires, Argentina
| | - Kevin Gardner
- National Cancer Institute and National Institute of Minority Health and Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Florencia Zalazar
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Carolina Flumian
- Área de Investigación del Instituto de Oncología A.H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura Todaro
- Área de Investigación del Instituto de Oncología A.H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elba S Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN - CONICET, Buenos Aires, Argentina
| | - Roberto Meiss
- Departamento de Patología, Instituto de Estudios Oncológicos, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Adriana De Siervi
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
32
|
Chang JTH, Wang F, Chapin W, Huang RS. Identification of MicroRNAs as Breast Cancer Prognosis Markers through the Cancer Genome Atlas. PLoS One 2016; 11:e0168284. [PMID: 27959953 PMCID: PMC5154569 DOI: 10.1371/journal.pone.0168284] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/29/2016] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the second-most common cancer and second-leading cause of cancer mortality in American women. The dysregulation of microRNAs (miRNAs) plays a key role in almost all cancers, including breast cancer. We comprehensively analyzed miRNA expression, global gene expression, and patient survival from the Cancer Genomes Atlas (TCGA) to identify clinically relevant miRNAs and their potential gene targets in breast tumors. In our analysis, we found that increased expression of 12 mature miRNAs-hsa-miR-320a, hsa-miR-361-5p, hsa-miR-103a-3p, hsa-miR-21-5p, hsa-miR-374b-5p, hsa-miR-140-3p, hsa-miR-25-3p, hsa-miR-651-5p, hsa-miR-200c-3p, hsa-miR-30a-5p, hsa-miR-30c-5p, and hsa-let-7i-5p -each predicted improved breast cancer survival. Of the 12 miRNAs, miR-320a, miR-361-5p, miR-21-5p, miR-103a-3p were selected for further analysis. By correlating global gene expression with miRNA expression and then employing miRNA target prediction analysis, we suggest that the four miRNAs may exert protective phenotypes by targeting breast oncogenes that contribute to patient survival. We propose that miR-320a targets the survival-associated genes RAD51, RRP1B, and TDG; miR-361-5p targets ARCN1; and miR-21-5p targets MSH2, RMND5A, STAG2, and UBE2D3. The results of our stringent bioinformatics approach for identifying clinically relevant miRNAs and their targets indicate that miR-320a, miR-361-5p, and miR-21-5p may contribute to breast cancer survival.
Collapse
Affiliation(s)
- Jeremy T-H. Chang
- Biological Sciences Collegiate Division, University of Chicago, Chicago, Illinois, United States of America
| | - Fan Wang
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - William Chapin
- Pritzker School of Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - R. Stephanie Huang
- Department of Medicine, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
33
|
Yang H, Wu L, Ke S, Wang W, Yang L, Gao X, Fang H, Yu H, Zhong Y, Xie C, Zhou F, Zhou Y. Downregulation of Ubiquitin-conjugating Enzyme UBE2D3 Promotes Telomere Maintenance and Radioresistance of Eca-109 Human Esophageal Carcinoma Cells. J Cancer 2016; 7:1152-62. [PMID: 27326259 PMCID: PMC4911883 DOI: 10.7150/jca.14745] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/26/2016] [Indexed: 12/28/2022] Open
Abstract
Ubiquitin-conjugating enzyme UBE2D3 is an important member of the ubiquitin-proteasome pathways. Our previous study showed that the expression of UBE2D3 was negatively related to human telomerase reverse transcriptase (hTERT) and radioresistance in human breast cancer cells. However, in esophageal carcinoma, the exact effects and mechanisms of UBE2D3 in radioresistance remain unclear. This study shows that UBE2D3 knockdown was associated with significant increases in radioresistance to X-rays, telomerase activity, telomere length, and telomere shelterins. UBE2D3 knockdown-mediated radioresistance was related to a decrease in the spontaneous and ionizing radiation-induced apoptosis, resulting from a decrease in the Bax/Bcl-2 ratio. Furthermore, UBE2D3 downregulation was associated with increased G1-S phase transition and prolonged IR-induced G2/M arrest through over expression of cyclin D1, decrease of CDC25A expression and promotion of the ATM/ATR-Chk1-CDC25C pathway. Moreover, UBE2D3 downregulation reduced spontaneous DNA double-strand breaks and accelerated the repair of DNA damage induced by IR. The current data thus demonstrate that UBE2D3 downregulation enhances radioresistance by increased telomere homeostasis and prolonged IR-induced G2/M arrest, but decreases the IR-induced apoptosis and the number of DNA damage foci. These results suggest that UBE2D3 might be a potential molecular target to improve radiotherapy effects in esophageal carcinoma.
Collapse
Affiliation(s)
- Hui Yang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lin Wu
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 3. Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Shaobo Ke
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wenbo Wang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Yang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xiaojia Gao
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Hongyan Fang
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haijun Yu
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yahua Zhong
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Conghua Xie
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- 1. Hubei Key Laboratory of Tumor Biological Behavior, Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, China;; 2. Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Castillo-Lluva S, Hontecillas-Prieto L, Blanco-Gómez A, Del Mar Sáez-Freire M, García-Cenador B, García-Criado J, Pérez-Andrés M, Orfao A, Cañamero M, Mao JH, Gridley T, Castellanos-Martín A, Pérez-Losada J. A new role of SNAI2 in postlactational involution of the mammary gland links it to luminal breast cancer development. Oncogene 2015; 34:4777-4790. [PMID: 26096931 PMCID: PMC4560637 DOI: 10.1038/onc.2015.224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/25/2015] [Accepted: 05/16/2015] [Indexed: 12/30/2022]
Abstract
Breast cancer is a major cause of mortality in women. The transcription factor SNAI2 has been implicated in the pathogenesis of several types of cancer, including breast cancer of basal origin. Here we show that SNAI2 is also important in the development of breast cancer of luminal origin in MMTV-ErbB2 mice. SNAI2 deficiency leads to longer latency and fewer luminal tumors, both of these being characteristics of pretumoral origin. These effects were associated with reduced proliferation and a decreased ability to generate mammospheres in normal mammary glands. However, the capacity to metastasize was not modified. Under conditions of increased ERBB2 oncogenic activity after pregnancy plus SNAI2 deficiency, both pretumoral defects-latency and tumor load-were compensated. However, the incidence of lung metastases was dramatically reduced. Furthermore, SNAI2 was required for proper postlactational involution of the breast. At 3 days post lactational involution, the mammary glands of Snai2-deficient mice exhibited lower levels of pSTAT3 and higher levels of pAKT1, resulting in decreased apoptosis. Abundant noninvoluted ducts were still present at 30 days post lactation, with a greater number of residual ERBB2+ cells. These results suggest that this defect in involution leads to an increase in the number of susceptible target cells for transformation, to the recovery of the capacity to generate mammospheres and to an increase in the number of tumors. Our work demonstrates the participation of SNAI2 in the pathogenesis of luminal breast cancer, and reveals an unexpected connection between the processes of postlactational involution and breast tumorigenesis in Snai2-null mutant mice.
Collapse
Affiliation(s)
- S Castillo-Lluva
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - L Hontecillas-Prieto
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - A Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - M Del Mar Sáez-Freire
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - B García-Cenador
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Cirugía, Universidad de Salamanca, Salamanca, Spain
| | - J García-Criado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Departamento de Cirugía, Universidad de Salamanca, Salamanca, Spain
| | - M Pérez-Andrés
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Unidad de Citometría de flujo, Universidad de Salamanca, IBSAL, Salamanca, Spain
| | - A Orfao
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Unidad de Citometría de flujo, Universidad de Salamanca, IBSAL, Salamanca, Spain
| | - M Cañamero
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - J H Mao
- Life Sciences Division, Lawrence Berkeley National Laboratory (LBNL), University of California, Berkeley, Berkeley, CA, USA
| | - T Gridley
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - A Castellanos-Martín
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| | - J Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Universidad de Salamanca/CSIC, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
| |
Collapse
|
35
|
Lu L, Zeng H, Gu X, Ma W. Circulating tumor cell clusters-associated gene plakoglobin and breast cancer survival. Breast Cancer Res Treat 2015; 151:491-500. [PMID: 25957595 DOI: 10.1007/s10549-015-3416-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/04/2015] [Indexed: 10/23/2022]
Abstract
Breast cancer recurrence is a major cause of the disease-specific death. Circulating tumor cells (CTCs) are negatively associated with breast cancer survival. Plakoglobin, a cell adhesion protein, was recently reported as a determinant of CTCs types, single or clustered ones. Here, we aim to summarize the studies on the roles of plakoglobin and evaluate the association of plakoglobin and breast cancer survival. Plakoglobin as a key component in both cell adhesion and the signaling pathways was briefly reviewed first. Then the double-edge functions of plakoglobin in tumors and its association with CTCs and breast cancer metastasis were introduced. Finally, based on an open-access database, the association between plakoglobin and breast cancer survival was investigated using univariate and multivariate survival analyses. Plakoglobin may be a molecule functioning as a double-edge sword. Loss of plakoglobin expression leads to increased motility of epithelial cells, thereby promoting epithelial-mesenchymal transition and further metastasis of cancer. However, studies also show that plakoglobin can function as an oncogene. High expression of plakoglobin results in clustered tumor cells in circulation with high metastatic potential in breast cancer and shortened patient survival. Plakoglobin may be a potential prognostic biomarker that can be exploited to develop as a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Lingeng Lu
- Department of Chronic Disease Epidemiology, School of Public Health, School of Medicine, Yale Cancer Center, Yale University, 60 College Street, New Haven, CT, 06520-8034, USA,
| | | | | | | |
Collapse
|
36
|
Zhang Z, Yang C, Gao W, Chen T, Qian T, Hu J, Tan Y. FOXA2 attenuates the epithelial to mesenchymal transition by regulating the transcription of E-cadherin and ZEB2 in human breast cancer. Cancer Lett 2015; 361:240-50. [PMID: 25779673 DOI: 10.1016/j.canlet.2015.03.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/09/2015] [Accepted: 03/09/2015] [Indexed: 01/04/2023]
Abstract
The Forkhead Box A2 (FOXA2) transcription factor is required for embryonic development and for normal functions of multiple adult tissues, in which the maintained expression of FOXA2 is usually related to preventing the progression of malignant transformation. In this study, we found that FOXA2 prevented the epithelial to mesenchymal transition (EMT) in human breast cancer. We observed a strong correlation between the expression levels of FOXA2 and the epithelial phenotype. Knockdown of FOXA2 promoted the mesenchymal phenotype, whereas stable overexpression of FOXA2 attenuated EMT in breast cancer cells. FOXA2 was found to endogenously bind to and stimulate the promoter of E-cadherin that is crucial for epithelial phenotype of the tumor cells. Meanwhile, FOXA2 prevented EMT of breast cancer cells by repressing the expression of EMT-related transcription factor ZEB2 through recruiting a transcriptional corepressor TLE3 to the ZEB2 promoter. The stable overexpression of FOXA2 abolished metastasis of breast cancer cells in vivo. This study confirmed that FOXA2 inhibited EMT in breast cancer cells by regulating the transcription of EMT-related genes such as E-cadherin and ZEB2.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | - Chao Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | - Wei Gao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | - Tuanhui Chen
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | - Tingting Qian
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | - Jun Hu
- Department of Pathology, Hunan Provincial Tumor Hospital, Changsha, Hunan 410013, China
| | - Yongjun Tan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
37
|
Guan GG, Wang WB, Lei BX, Wang QL, Wu L, Fu ZM, Zhou FX, Zhou YF. UBE2D3 is a positive prognostic factor and is negatively correlated with hTERT expression in esophageal cancer. Oncol Lett 2015; 9:1567-1574. [PMID: 25789002 PMCID: PMC4356423 DOI: 10.3892/ol.2015.2926] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) is a critical factor in unlimited cell proliferation and immortalization, with numerous studies demonstrating that high expression of hTERT is a poor prognostic factor in various types of cancer. Ubiquitin-conjugating enzyme E2D 3 (UBE2D3) is a member of the E2 family, and participates in the ubiquitin proteasome pathway to regulate basic cellular activities, such as cell cycle control, the DNA damage response, apoptosis, and tumorigenesis. Our previous study initially determined that downregulation of UBE2D3 expression increases hTERT expression and cell proliferation, however, the association between the expression of these two proteins and their functions in cancer tissues remains unknown. Therefore, the protein expression levels of hTERT and UBE2D3 were evaluated in 150 esophageal cancer and 30 adjacent healthy tissue samples by performing immunohistochemical analysis. Concurrently, the clinicopathological data of the enrolled patients were obtained to allow correlation analysis. It was identified that the expression of hTERT in the esophageal cancer tissues was significantly higher compared with that of the adjacent tissues (P=0.015), however, the expression of UBE2D3 was significantly lower in esophageal cancer tissues than the adjacent tissues (P=0.001). Additionally, the study demonstrated that hTERT was significantly upregulated in poorly-differentiated, advanced tumor-node-metastasis (TNM) stage cancer tissues (P<0.05 for all), however, UBE2D3 expression was downregulated in poorly-differentiated, lymph node invaded cancer tissues and recurrent cases. It was also identified that traditional factors, including tumor location, T stage, lymph node status, TNM stage, and molecular factors of hTERT and UBE2D3, were significantly associated with overall survival time (P<0.05 for all). Furthermore, UBE2D3, lymph node status and tumor location were independent prognostic factors for esophageal cancer in multivariate analysis. Most notably, hTERT and UBE2D3 expression were negatively correlated with each other. In conclusion, the findings of the present study indicated that hTERT and UBE2D3 proteins appear to be involved in the development of esophageal cancer, that UBE2D3 may a positive prognostic factor for esophageal cancer, and that UBE2D3 and hTERT expression levels are inversely correlated.
Collapse
Affiliation(s)
- Ge Ge Guan
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wen Bo Wang
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China ; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Bing Xin Lei
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Qiao Li Wang
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Lin Wu
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhen Ming Fu
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China ; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Fu Xiang Zhou
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China ; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yun Feng Zhou
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei 430071, P.R. China ; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
38
|
Shao S, Zhao X, Zhang X, Luo M, Zuo X, Huang S, Wang Y, Gu S, Zhao X. Notch1 signaling regulates the epithelial-mesenchymal transition and invasion of breast cancer in a Slug-dependent manner. Mol Cancer 2015; 14:28. [PMID: 25645291 PMCID: PMC4322803 DOI: 10.1186/s12943-015-0295-3] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 01/13/2015] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of breast cancer. However, how Notch signaling regulates the EMT process and invasion in breast cancer remains largely unknown. METHODS The impact of Notch1 silencing by specific shRNAs on the EMT and invasion of human breast cancer MCF-7 and MDA-MB-231 cells as well as xenografts was tested by western blot, real-time polymerase chain reaction (RT-PCR), immunofluorescence, transwell, and immunohistochemistry assays. The effect of Slug silencing or upregulation on the EMT and invasion of breast cancer cells was analyzed, and the effect of Notch1 signaling on Slug expression was determined by the luciferase reporter assay. RESULTS The Notch1 intracellular domain (N1ICD) and Jagged1 were expressed in breast cancer cells. Notch1 silencing reversed the spontaneous EMT process and inhibited the migration and invasion of breast cancer cells and the growth of xenograft breast cancers. The expression of N1ICD was upregulated significantly by Jagged1-mediated Notch signaling activation. Moreover, Jagged1-mediated Notch signaling promoted the EMT process, migration, and invasion of breast cancer cells, which were abrogated by Notch silencing. Furthermore, the N1ICD positively regulated the Slug expression by inducing Slug promoter activation. Importantly, the knockdown of Slug weakened the invasion ability of breast cancer cells and reversed the Jagged1-induced EMT process with significantly decreased expression of vimentin and increased expression of E-cadherin. In addition, Slug overexpression restored the Notch1 knockdown-suppressed EMT process. CONCLUSIONS Our novel data indicate that Notch signaling positively regulates the EMT, invasion, and growth of breast cancer cells by inducing Slug expression. The Notch1-Slug signaling axis may represent a potential therapeutic target for breast cancer therapy.
Collapse
Affiliation(s)
- Shan Shao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoai Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaojin Zhang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Minna Luo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xiaoxiao Zuo
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shangke Huang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Ying Wang
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Shanzhi Gu
- The Department of Forensic Medicine, Medical School, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| | - Xinhan Zhao
- The Department of Oncology, the First Hospital Affiliated to the School of Medicine, Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
39
|
Kim S, Yao J, Suyama K, Qian X, Qian BZ, Bandyopadhyay S, Loudig O, De Leon-Rodriguez C, Zhou ZN, Segall J, Macian F, Norton L, Hazan RB. Slug promotes survival during metastasis through suppression of Puma-mediated apoptosis. Cancer Res 2014; 74:3695-706. [PMID: 24830722 DOI: 10.1158/0008-5472.can-13-2591] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor cells must overcome apoptosis to survive throughout metastatic dissemination and distal organ colonization. Here, we show in the Polyoma Middle T mammary tumor model that N-cadherin (Cdh2) expression causes Slug (Snai2) upregulation, which in turn promotes carcinoma cell survival. Slug was dramatically upregulated in metastases relative to primary tumors. Consistent with a role in metastasis, Slug knockdown in carcinoma cells suppressed lung colonization by decreasing cell survival at metastatic sites, but had no effect on tumor cell invasion or extravasation. In support of this idea, Slug inhibition by shRNA sensitized tumor cells to apoptosis by DNA damage, resulting in caspase-3 and PARP cleavage. The prosurvival effect of Slug was found to be caused by direct repression of the proapoptotic gene, Puma (Bbc3), by Slug. Consistent with a pivotal role for a Slug-Puma axis in metastasis, inhibition of Puma by RNA interference in Slug-knockdown cells rescued lung colonization, whereas Puma overexpression in control tumor cells suppressed lung metastasis. The survival function of the Slug-Puma axis was confirmed in human breast cancer cells, where Slug knockdown increased Puma expression and inhibited lung colonization. This study demonstrates a pivotal role for Slug in carcinoma cell survival, implying that disruption of the Slug-Puma axis may impinge on the survival of metastatic cells.
Collapse
Affiliation(s)
- Seaho Kim
- Authors' Affiliations: Departments of Pathology
| | - Jiahong Yao
- Authors' Affiliations: Departments of Pathology
| | | | - Xia Qian
- Authors' Affiliations: Departments of Pathology
| | | | | | | | | | - Zhen Ni Zhou
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx; and
| | - Jeffrey Segall
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx; and
| | | | - Larry Norton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | |
Collapse
|
40
|
Casimiro MC, Velasco-Velázquez M, Aguirre-Alvarado C, Pestell RG. Overview of cyclins D1 function in cancer and the CDK inhibitor landscape: past and present. Expert Opin Investig Drugs 2014; 23:295-304. [PMID: 24387133 DOI: 10.1517/13543784.2014.867017] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Intensive efforts, over the last decade, have been made to inhibit the kinase activity of cyclins that act as mediators during cell-cycle progression. Activation of the cyclin D1 oncogene, often by amplification or rearrangement, is a major driver of multiple types of human tumors including breast and squamous cell cancers, B-cell lymphoma, myeloma and parathyroid adenoma. AREAS COVERED In this review, the authors summarize the activity of cyclins and cyclin-dependent kinases in cell-cycle progression and transcription. They focus on cyclin D1/CDK4/CDK6, a central mediator in the transition from G1 to S phase. Furthermore, the authors discuss the first generation of pan-cyclin-dependent kinase inhibitors that failed to meet expectation and discuss, in detail, the second generation of highly specific cyclin D1/CDK4/CDK6 inhibitors that are proving to be more efficacious. EXPERT OPINION The mechanism by which cyclin D1 drives tumorigenesis may be dependent on kinase and kinase-independent functions. Further evidence is necessary to delineate the roles of cyclin D1 in early pre-neoplastic lesions where its overexpression may promote genomic instability in a kinase-independent manner.
Collapse
Affiliation(s)
- Mathew C Casimiro
- Thomas Jefferson University & Hospital, Department of Cancer Biology , 233 South 10th Street, Philadelphia, PA 19107 , USA
| | | | | | | |
Collapse
|
41
|
Yang C, Chen H, Tan G, Gao W, Cheng L, Jiang X, Yu L, Tan Y. FOXM1 promotes the epithelial to mesenchymal transition by stimulating the transcription of Slug in human breast cancer. Cancer Lett 2013; 340:104-12. [PMID: 23856032 DOI: 10.1016/j.canlet.2013.07.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 06/05/2013] [Accepted: 07/08/2013] [Indexed: 01/11/2023]
Abstract
The Forkhead Box M1 (FOXM1) transcription factor is involved in tumorigenesis and tumor progression in multiple human carcinomas. In this study, we found that FOXM1 promoted the epithelial to mesenchymal transition (EMT) in human breast cancer. We observed a strong correlation between the expression levels of FOXM1 and the mesenchymal phenotype. Knockdown of FOXM1 inhibited the mesenchymal phenotype, whereas stable overexpression of FOXM1 induced EMT in breast cancer cells. FOXM1 was found to endogenously bind to and stimulate the promoter of Slug that is crucial for EMT progression. The knockdown of Slug abolished the EMT-inducing function of FOXM1. The stable overexpression of FOXM1 promoted metastasis of breast cancer cells in vivo. This study confirmed that FOXM1 promoted EMT in breast cancer cells by stimulating the transcription of EMT-related genes such as Slug.
Collapse
Affiliation(s)
- Chao Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, Hunan 410082, China
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang W, Yang L, Hu L, Li F, Ren L, Yu H, Liu Y, Xia L, Lei H, Liao Z, Zhou F, Xie C, Zhou Y. Inhibition of UBE2D3 expression attenuates radiosensitivity of MCF-7 human breast cancer cells by increasing hTERT expression and activity. PLoS One 2013; 8:e64660. [PMID: 23741361 PMCID: PMC3669415 DOI: 10.1371/journal.pone.0064660] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/17/2013] [Indexed: 12/25/2022] Open
Abstract
The known functions of telomerase in tumor cells include replenishing telomeric DNA and maintaining cell immortality. We have previously shown the existence of a negative correlation between human telomerase reverse transcriptase (hTERT) and radiosensitivity in tumor cells. Here we set out to elucidate the molecular mechanisms underlying regulation by telomerase of radiosensitivity in MCF-7 cells. Toward this aim, yeast two-hybrid (Y2H) screening of a human laryngeal squamous cell carcinoma radioresistant (Hep2R) cDNA library was first performed to search for potential hTERT interacting proteins. We identified ubiquitin-conjugating enzyme E2D3 (UBE2D3) as a principle hTERT-interacting protein and validated this association biochemically. ShRNA-mediated inhibition of UBE2D3 expression attenuated MCF-7 radiosensitivity, and induced the accumulation of hTERT and cyclin D1 in these cells. Moreover, down-regulation of UBE2D3 increased hTERT activity and cell proliferation, accelerating G1 to S phase transition in MCF-7 cells. Collectively these findings suggest that UBE2D3 participates in the process of hTERT-mediated radiosensitivity in human breast cancer MCF-7 cells by regulating hTERT and cyclin D1.
Collapse
Affiliation(s)
- Wenbo Wang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Lei Yang
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Liu Hu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fen Li
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Li Ren
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Haijun Yu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yu Liu
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Ling Xia
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Han Lei
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Zhengkai Liao
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Fuxiang Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Conghua Xie
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yunfeng Zhou
- Hubei Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, China
- Department of Radiation Oncology & Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
43
|
Tamura D, Arao T, Nagai T, Kaneda H, Aomatsu K, Fujita Y, Matsumoto K, De Velasco MA, Kato H, Hayashi H, Yoshida S, Kimura H, Maniwa Y, Nishio W, Sakai Y, Ohbayashi C, Kotani Y, Nishimura Y, Nishio K. Slug increases sensitivity to tubulin-binding agents via the downregulation of βIII and βIVa-tubulin in lung cancer cells. Cancer Med 2013; 2:144-54. [PMID: 23634282 PMCID: PMC3639653 DOI: 10.1002/cam4.68] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/12/2013] [Accepted: 01/22/2013] [Indexed: 12/11/2022] Open
Abstract
Transcription factor Slug/SNAI2 (snail homolog 2) plays a key role in the induction of the epithelial mesenchymal transition in cancer cells; however, whether the overexpression of Slug mediates the malignant phenotype and alters drug sensitivity in lung cancer cells remains largely unclear. We investigated Slug focusing on its biological function and involvement in drug sensitivity in lung cancer cells. Stable Slug transfectants showed typical morphological changes compared with control cells. Slug overexpression did not change the cellular proliferations; however, migration activity and anchorage-independent growth activity with an antiapoptotic effect were increased. Interestingly, stable Slug overexpression increased drug sensitivity to tubulin-binding agents including vinorelbine, vincristine, and paclitaxel (5.8- to 8.9-fold increase) in several lung cancer cell lines but did not increase sensitivity to agents other than tubulin-binding agents. Real-time RT-PCR (polymerase chain reaction) and western blotting revealed that Slug overexpression downregulated the expression of βIII and βIVa-tubulin, which is considered to be a major factor determining sensitivity to tubulin-binding agents. A luciferase reporter assay confirmed that Slug suppressed the promoter activity of βIVa-tubulin at a transcriptional level. Slug overexpression enhanced tumor growth, whereas Slug overexpression increased drug sensitivity to vinorelbine with the downregulation of βIII and βIV-tubulin in vivo. Immunohistochemistry of Slug with clinical lung cancer samples showed that Slug overexpression tended to be involved in response to tubulin-binding agents. In conclusion, our data indicate that Slug mediates an aggressive phenotype including enhanced migration activity, anoikis suppression, and tumor growth, but increases sensitivity to tubulin-binding agents via the downregulation of βIII and βIVa-tubulin in lung cancer cells.
Collapse
Affiliation(s)
- Daisuke Tamura
- Department of Genome Biology, Kinki University Faculty of Medicine Osaka, Japan; Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Liang YJ, Wang QY, Zhou CX, Yin QQ, He M, Yu XT, Cao DX, Chen GQ, He JR, Zhao Q. MiR-124 targets Slug to regulate epithelial-mesenchymal transition and metastasis of breast cancer. Carcinogenesis 2012; 34:713-22. [PMID: 23250910 PMCID: PMC3581604 DOI: 10.1093/carcin/bgs383] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs (miRNAs or miR) have been integrated into tumorigenic programs as either oncogenes or tumor suppressor genes. The miR-124 was reported to be attenuated in several tumors, such as glioma, medulloblastoma and hepatocellular carcinoma. However, its role in cancer remains greatly elusive. In this study, we show that the miR-124 expression is significantly suppressed in human breast cancer specimens, which is reversely correlated to histological grade of the cancer. More intriguingly, ectopic expression of miR-124 in aggressive breast cancer cell lines MDA-MB-231 and BT-549 strongly inhibits cell motility and invasive capacity, as well as the epithelial–mesenchymal transition process. Also, lentivirus-delivered miR-124 endows MDA-MB-231 cells with the ability to suppress cell colony formation in vitro and pulmonary metastasis in vivo. Further studies have identified the E-cadherin transcription repressor Slug as a direct target gene of miR-124; its downregulation by miR-124 increases the expression of E-cadherin, a hallmark of epithelial cells and a repressor of cell invasion and metastasis. Moreover, knockdown of Slug notably impairs the motility of MDA-MB-231 cells, whereas re-expression of Slug abrogates the reduction of motility and invasion ability induced by miR-124 in MDA-MB-231 cells. These findings highlight an important role for miR-124 in the regulation of invasive and metastatic potential of breast cancer and suggest a potential application of miR-124 in cancer treatment.
Collapse
Affiliation(s)
- Yong-Jun Liang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Breast cancer is the most common malignancy in women and a significant cause of morbidity and mortality. Sub-types of breast cancer defined by the expression of steroid hormones and Her2/Neu oncogene have distinct prognosis and undergo different therapies. Besides differing in their phenotype, sub-types of breast cancer display various molecular lesions that participate in their pathogenesis. BRCA1 is one of the common hereditary cancer predisposition genes and encodes for an ubiquitin ligase. Ubiquitin ligases or E3 enzymes participate together with ubiquitin activating enzyme and ubiquitin conjugating enzymes in the attachment of ubiquitin (ubiquitination) in target proteins. Ubiquitination is a post-translational modification regulating multiple cell functions. It also plays important roles in carcinogenesis in general and in breast carcinogenesis in particular. Ubiquitin conjugating enzymes are a central component of the ubiquitination machinery and are often perturbed in breast cancer. This paper will discuss ubiquitin and ubiquitin-like proteins conjugating enzymes participating in breast cancer pathogenesis, their relationships with other proteins of the ubiquitination machinery and their role in phenotype of breast cancer sub-types.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Centre Pluridisciplinaire d'Oncologie, BH06, University Hospital of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
46
|
Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A. EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci 2012; 69:3429-56. [PMID: 22945800 PMCID: PMC11115078 DOI: 10.1007/s00018-012-1122-2] [Citation(s) in RCA: 401] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 12/13/2022]
Abstract
Cancer is a complex multistep process involving genetic and epigenetic changes that eventually result in the activation of oncogenic pathways and/or inactivation of tumor suppressor signals. During cancer progression, cancer cells acquire a number of hallmarks that promote tumor growth and invasion. A crucial mechanism by which carcinoma cells enhance their invasive capacity is the dissolution of intercellular adhesions and the acquisition of a more motile mesenchymal phenotype as part of an epithelial-to-mesenchymal transition (EMT). Although many transcription factors can trigger it, the full molecular reprogramming occurring during an EMT is mainly orchestrated by three major groups of transcription factors: the ZEB, Snail and Twist families. Upregulated expression of these EMT-activating transcription factors (EMT-ATFs) promotes tumor invasiveness in cell lines and xenograft mice models and has been associated with poor clinical prognosis in human cancers. Evidence accumulated in the last few years indicates that EMT-ATFs also regulate an expanding set of cancer cell capabilities beyond tumor invasion. Thus, EMT-ATFs have been shown to cooperate in oncogenic transformation, regulate cancer cell stemness, override safeguard programs against cancer like apoptosis and senescence, determine resistance to chemotherapy and promote tumor angiogenesis. This article reviews the expanding portfolio of functions played by EMT-ATFs in cancer progression.
Collapse
Affiliation(s)
- Ester Sánchez-Tilló
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
| | - Yongqing Liu
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Oriol de Barrios
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
| | - Lucia Fanlo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- Master Program in Biomedical Research, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Miriam Cuatrecasas
- Department of Pathology, Hospital Clinic and IDIBAPS’ Tumor Bank, 08036 Barcelona, Spain
| | - Douglas S. Darling
- Department of Oral Health and Rehabilitation, Center for Genetics and Molecular Medicine, University of Louisville, Louisville, KY 40202 USA
| | - Douglas C. Dean
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- Department of Ophthalmology and Birth Defects Center, Louisville Health Science Center, Louisville, KY 40202 USA
| | - Antoni Castells
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- Institute of Digestive and Metabolic Diseases, Hospital Clinic, 08036 Barcelona, Spain
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036 Barcelona, Spain
- CIBERehd (Gastrointestinal and Pancreatic Oncology), IDIBAPS, 08036 Barcelona, Spain
- James Graham Brown Cancer Center, Louisville Health Science Center, Louisville, KY 40202 USA
- ICREA, 08010 Barcelona, Spain
| |
Collapse
|
47
|
Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc Natl Acad Sci U S A 2012; 109:16654-9. [PMID: 23011797 DOI: 10.1073/pnas.1205822109] [Citation(s) in RCA: 241] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Slug (Snail2) plays critical roles in regulating the epithelial-mesenchymal transition (EMT) programs operative during development and disease. However, the means by which Slug activity is controlled remain unclear. Herein we identify an unrecognized canonical Wnt/GSK3β/β-Trcp1 axis that controls Slug activity. In the absence of Wnt signaling, Slug is phosphorylated by GSK3β and subsequently undergoes β-Trcp1-dependent ubiquitination and proteosomal degradation. Alternatively, in the presence of canonical Wnt ligands, GSK3β kinase activity is inhibited, nuclear Slug levels increase, and EMT programs are initiated. Consistent with recent studies describing correlative associations in basal-like breast cancers between Wnt signaling, increased Slug levels, and reduced expression of the tumor suppressor Breast Cancer 1, Early Onset (BRCA1), further studies demonstrate that Slug-as well as Snail-directly represses BRCA1 expression by recruiting the chromatin-demethylase, LSD1, and binding to a series of E-boxes located within the BRCA1 promoter. Consonant with these findings, nuclear Slug and Snail expression are increased in association with BRCA1 repression in a cohort of triple-negative breast cancer patients. Together, these findings establish unique functional links between canonical Wnt signaling, Slug expression, EMT, and BRCA1 regulation.
Collapse
|
48
|
Liu Z, Li S, Cai Y, Wang A, He Q, Zheng C, Zhao T, Ding X, Zhou X. Manganese superoxide dismutase induces migration and invasion of tongue squamous cell carcinoma via H2O2-dependent Snail signaling. Free Radic Biol Med 2012; 53:44-50. [PMID: 22580338 PMCID: PMC3377784 DOI: 10.1016/j.freeradbiomed.2012.04.031] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 04/18/2012] [Accepted: 04/21/2012] [Indexed: 01/06/2023]
Abstract
Our previous studies had revealed that the dysregulation of manganese superoxide dismutase (SOD2) expression was a frequent event in tongue squamous cell carcinoma (TSCC) and may be associated with enhanced metastatic potential. To further evaluate the mechanism of SOD2-mediated metastasis in TSCC, TSCC cell lines with different metastatic potentials (i.e., the highly metastatic UM1 line and the UM2 line, which displays fewer metastases) were used. Compared to UM2 cells, UM1 cells exhibited significantly higher SOD2 activity and intracellular H(2)O(2); higher protein levels of Snail, MMP1, and pERK1/2; lower protein levels of E-cadherin; and no difference in catalase activity. Upon knockdown of SOD2 by RNA interference, UM1 cells displayed significantly reduced migration and invasion abilities; reduced activities of SOD2; lower intracellular H(2)O(2); decreased protein levels of Snail, MMP1, and pERK1/2; and increased protein levels of E-cadherin. The migration and invasion abilities of UM2 and SOD2 shRNA-transfected UM1 cells were enhanced by H(2)O(2) treatment and accompanied by increased protein levels of Snail, MMP1, and pERK1/2 and decreased protein levels of E-cadherin. Moreover the migration and invasion abilities of UM1 cells were decreased after catalase treatment. Thus, we conclude that the SOD2-dependent production of H(2)O(2) contributes to both the migration and the invasion of TSCC via the Snail signaling pathway, through increased Snail, MMP1, and pERK1/2 protein levels and the repression of the E-cadherin protein.
Collapse
Affiliation(s)
- Zhonghua Liu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Su Li
- Department of Medicine, Tumor Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuchen Cai
- Department of Medicine, Tumor Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Corresponding author: Anxun Wang, Department of Oral and Maxillofacial Surgery, First Affiliated Hospital, Sun-Yat-Sen University. 58 Zhongshan Road II, Guangzhou, 510080, P.R.China; Phone: +86-0-13724896216;
| | - Qianting He
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoxu Zheng
- Department of General Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tingting Zhao
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xueqiang Ding
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaofeng Zhou
- Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, Chicago, IL
- Graduate College, UIC Cancer Center, University of Illinois at Chicago, Chicago, IL
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
49
|
EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012; 22:194-207. [DOI: 10.1016/j.semcancer.2012.02.013] [Citation(s) in RCA: 354] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/24/2022]
|
50
|
Molina-Ortiz P, Villarejo A, MacPherson M, Santos V, Montes A, Souchelnytskyi S, Portillo F, Cano A. Characterization of the SNAG and SLUG domains of Snail2 in the repression of E-cadherin and EMT induction: modulation by serine 4 phosphorylation. PLoS One 2012; 7:e36132. [PMID: 22567133 PMCID: PMC3342263 DOI: 10.1371/journal.pone.0036132] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 03/26/2012] [Indexed: 11/18/2022] Open
Abstract
Snail1 and Snail2, two highly related members of the Snail superfamily, are direct transcriptional repressors of E-cadherin and EMT inducers. Previous comparative gene profiling analyses have revealed important differences in the gene expression pattern regulated by Snail1 and Snail2, indicating functional differences between both factors. The molecular mechanism of Snail1-mediated repression has been elucidated to some extent, but very little is presently known on the repression mediated by Snail2. In the present work, we report on the characterization of Snail2 repression of E-cadherin and its regulation by phosphorylation. Both the N-terminal SNAG and the central SLUG domains of Snail2 are required for efficient repression of the E-cadherin promoter. The co-repressor NCoR interacts with Snail2 through the SNAG domain, while CtBP1 is recruited through the SLUG domain. Interestingly, the SNAG domain is absolutely required for EMT induction while the SLUG domain plays a negative modulation of Snail2 mediated EMT. Additionally, we identify here novel in vivo phosphorylation sites at serine 4 and serine 88 of Snail2 and demonstrate the functional implication of serine 4 in the regulation of Snail2-mediated repressor activity of E-cadherin and in Snail2 induction of EMT.
Collapse
Affiliation(s)
- Patricia Molina-Ortiz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Ana Villarejo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Matthew MacPherson
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Vanesa Santos
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Amalia Montes
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Serhiy Souchelnytskyi
- Karolinska Biomics Center, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Francisco Portillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
| | - Amparo Cano
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Madrid (UAM), Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, IdiPAZ, Madrid, Spain
- * E-mail:
| |
Collapse
|