1
|
Zhou Y, Dong C, Shen X, Wang P, Chen T, Li W, Sun X, Li P, Xu C, Duan K, Li D, Zhou J. Targeting PTBP3-Mediated Alternative Splicing of COX11 Induces Cuproptosis for Inhibiting Gastric Cancer Peritoneal Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415983. [PMID: 40270362 DOI: 10.1002/advs.202415983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/29/2025] [Indexed: 04/25/2025]
Abstract
Numerous aberrant splicing events are implicated in tumor progression, yet comprehensive reports on splicing factors and events associated with peritoneal metastasis in gastric cancer (GCPM) are lacking. In this study, PTBP3 is found to be significantly overexpressed in peritoneal metastatic tissues of gastric cancer compared to primary tumor tissues, and higher PTBP3 expression correlates with poorer prognosis. Using gastric cancer cells and patient-derived organoids (PDO), the role of PTBP3 in promoting tumor invasion and proliferation is investigated. Mechanistically, through full-length transcriptome sequencing, PTBP3 mediates exon 4 skipping in its target gene COX11, leading to shorter transcripts that impair COX11 protein function, reducing mitochondrial copper content and enabling tumor cells to evade cuproptosis. Antisense oligonucleotide (ASO) drugs targeting the short COX11 transcripts effectively degrade mRNA, disrupting copper homeostasis. In PDO-based xenograft models, exogenous copper ionophores combined with ASO drugs induce excessive copper accumulation in mitochondria, triggering proteotoxic stress and cuproptosis. Overall, PTBP3-mediated exon 4 skipping in COX11 pre-mRNA is critical for tumor cell survival and progression in GCPM, offering potential therapeutic strategies targeting copper metabolism.
Collapse
Affiliation(s)
- Yajing Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Chao Dong
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Xiaochun Shen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Pengbo Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Tao Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Weikang Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Xiaotong Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Peiyuan Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Chengxiang Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Kaipeng Duan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Dongbao Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu Province, 215031, China
| |
Collapse
|
2
|
Cao ND, Zhu XH, Ma FQ, Xu Y, Dong JH, Qin MM, Liu TS, Zhu CC, Guo WJ, Ding HH, Guo YB, Liu LK, Song JJ, Wu JP, Cheng YL, Zeng L, Zhao AG. Chinese Medicine Prolongs Overall Survival of Chinese Patients with Advanced Gastric Cancer: Treatment Pattern and Survival Analysis of a 20-Year Real-World Study. Chin J Integr Med 2024; 30:489-498. [PMID: 38801641 DOI: 10.1007/s11655-024-4107-8] [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] [Accepted: 12/25/2023] [Indexed: 05/29/2024]
Abstract
OBJECTIVE To describe the treatment patterns and survival status of advanced gastric cancer (AGC) in China in the past two decades, and objectively evaluate the impact of standardized Chinese medicine (CM) treatment on the survival of AGC patients. METHODS This multicenter registry designed and propensity score analysis study described the diagnosis characteristics, treatment-pattern development and survival status of AGC from 10 hospitals in China between January 1, 2000 and July 31, 2021. Overall survival (OS) was evaluated between non-CM cohort (standard medical treatment) and CM cohort (integrated standard CM treatment ≥3 months). Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were performed to adjust any difference in average outcomes for bias. RESULTS A total of 2,001 patients histologically confirmed locally advanced and/or metastasis stomach and gastroesophageal junction adenocarcinoma were enrolled. Among them, 1,607 received systemic chemotherapy, 215 (10.74%) accepted molecular targeted therapy, 44 (2.2%) received checkpoint inhibitor therapy, and 769 (38.43%) received CM. Two-drug regimen was the main choice for first-line treatment, with fluoropyrimidine plus platinum as the most common regimen (530 cases, 60.09%). While 45.71% (16 cases) of patients with HER2 amplification received trastuzumab in first-line. The application of apatinib increased (33.33%) in third-line. The application of checkpoint inhibitors has increased since 2020. COX analysis showed that Lauren mixed type (P=0.017), cycles of first-line treatment >6 (P=0.000), CM (P=0.000), palliative gastrectomy (P=0.000), trastuzumab (P=0.011), and apatinib (P=0.008) were independent prognostic factors for the OS of AGC. After PSM and IPTW, the median OS of CM cohort and non-CM cohort was 18.17 and 12.45 months, respectively (P<0.001). CONCLUSIONS In real-world practice for AGC in China, therapy choices consisted with guidelines. Two-drug regimen was the main first-line choice. Standardized CM treatment was an independent prognostic factor and could prolong the OS of Chinese patients with AGC. (Registration No. NCT02781285).
Collapse
Affiliation(s)
- Ni-da Cao
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xiao-Hong Zhu
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fang-Qi Ma
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yan Xu
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jia-Huan Dong
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Meng-Meng Qin
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Tian-Shu Liu
- Oncology Department, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chun-Chao Zhu
- Gastrointestinal Surgery Department, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Wei-Jian Guo
- Oncology Department, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Hong-Hua Ding
- Oncology Department, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Yuan-Biao Guo
- Traditional Chinese Medicine Department, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Li-Kun Liu
- Oncology Department, Shanxi Traditional Chinese Medicine Hospital, Taiyuan, 030012, China
| | - Jin-Jie Song
- Oncology Department, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, 102445, China
| | - Ji-Ping Wu
- Oncology Department, Yunnan Province Hospital of Traditional Chinese Medicine, Kunming, 650021, China
| | - Yue-Lei Cheng
- Oncology Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lin Zeng
- Shanghai NewCore Biotechnology Co., Ltd., Shanghai, 200240, China
| | - Ai-Guang Zhao
- Oncology Department I, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
3
|
Shi G, Li H, Chen Y, Chen Z, Lin X. CircSEPT9 promotes breast cancer progression by regulating PTBP3 expression via sponging miR-625-5p. Thorac Cancer 2024; 15:808-819. [PMID: 38409914 PMCID: PMC10995703 DOI: 10.1111/1759-7714.15252] [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: 12/27/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is a common malignancy which threatens the health of women. Circular RNAs (circRNAs) are critical factors in multiple cancers, including BC. The aim of this experiment was to investigate the molecular mechanisms of circRNA Septin 9 (circSEPT9) in the progression of BC. METHODS CircSEPT9, microRNA-625-5p (miR-625-5p) and polypyrimidine tract-binding protein 3 (PTBP3) levels were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was performed to detect the protein levels of PTBP3, E-cadherin and vimentin. Cell counting kit-8 assay (CCK8) and thymidine analog 5-ethynyl-2'-deoxyuridine (EDU) was utilized for proliferation examination. Flow cytometry was conducted to measure apoptosis. Transwell assay and wound healing assay to investigate the migration of BC cells. Glucose uptake and lactate production were determined by specific kits. Additionally, dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were utilized to verify the interaction. A murine xenograft model was established to investigate the function of circSEPT9 in BC in vivo. RESULTS Overexpression of circSEPT9 was found in BC tissues and cells. Silencing circSEPT9 impeded BC cell proliferation, migration, epithelial-mesenchymal transition (EMT) and glycolytic metabolism but facilitated cell apoptosis in vitro. Meanwhile, circSEPT9 knockdown constrained tumor growth in vivo. MiR-625-5p was targeted by circSEPT9. The influence of silencing circSEPT9 on BC cell function was regained by miR-625-5p inhibitor. Furthermore, miR-625-5p regulated BC cell malignant phenotypes via downregulating PTBP3. CONCLUSION circSEPT9 contributed to the malignant progression of BC by up-regulating PTBP3 via sponging miR-625-5p.
Collapse
Affiliation(s)
- Guangtao Shi
- Zhejiang Society for Mathematical MedicineHangzhouChina
| | - Hongbo Li
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou CityChina
| | - Ying Chen
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou CityChina
| | - Zhi Chen
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou CityChina
| | - Xiaoji Lin
- Oncology Discipline Group, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou CityChina
| |
Collapse
|
4
|
Lu N, Min J, Peng L, Huang S, Chai X, Wang S, Wang J. MiR-297 inhibits tumour progression of liver cancer by targeting PTBP3. Cell Death Dis 2023; 14:564. [PMID: 37633911 PMCID: PMC10460384 DOI: 10.1038/s41419-023-06097-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
Whereas increasing evidences demonstrate that miR-297 contributes to the tumour development and progression, the role of miR-297 and its underlying molecular mechanisms in hepatocellular carcinoma (HCC) was still unclear. Here, we reported that the expression of miR-297 increased significantly in hepG2 cells after the treatment of the conditioned medium of human amniotic epithelial cells(hAECs) which can inhibit the proliferation and migration of hepG2. And the overexpression of miR-297 inhibits the cell proliferation, migration and invasion of HCC cell lines in vitro and suppressed the tumorigenesis of HCC in vivo. Polypyrimidine tract-binding protein 3 (PTBP3) was identified as a direct target gene of miR-297 in HCC cell lines, and mediated the function of miR-297 in HCC cells. In clinical samples, miR-297 levels have a tendency to decrease, but there are no statistically significant differences. Furthermore, in vitro cell experiments confirmed that overexpression of miR-297 could inhibit the PI3K/AKT signaling pathway by down-regulating PTBP3 expression, thereby inhibiting the proliferation, migration and invasion of HCC cells. In conclusion, our results revealed that miR-297 could down-regulate the expression of PTBP3 and inhibit the activation of PI3K/AKT signaling pathway, thereby preventing HCC growth, migration and invasion.
Collapse
Affiliation(s)
- Na Lu
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Jiali Min
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lin Peng
- Changsha Hospital for Maternal and Child Health Care of Hunan Normal University, Changsha, China
| | - Shengjian Huang
- Hunan Guangxiu Hi-tech Life Technology Co., Ltd., Changsha, China
| | - Xiahua Chai
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Susu Wang
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jian Wang
- The Institute of Reproduction and Stem Cell Engineering, School of Basic Medical Sciences, Central South University, Changsha, China.
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China.
- National Engineering and Research Center of Human Stem Cells, Changsha, China.
| |
Collapse
|
5
|
Ogiya D, Chyra Z, Verselis SJ, O'Keefe M, Cobb J, Abiatari I, Talluri S, Sithara AA, Hideshima T, Chu MP, Hájek R, Dorfman DM, Pilarski LM, Anderson KC, Adamia S. Identification of disease-related aberrantly spliced transcripts in myeloma and strategies to target these alterations by RNA-based therapeutics. Blood Cancer J 2023; 13:23. [PMID: 36737429 PMCID: PMC9898564 DOI: 10.1038/s41408-023-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 12/17/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
Novel drug discoveries have shifted the treatment paradigms of most hematological malignancies, including multiple myeloma (MM). However, this plasma cell malignancy remains incurable, and novel therapies are therefore urgently needed. Whole-genome transcriptome analyses in a large cohort of MM patients demonstrated that alterations in pre-mRNA splicing (AS) are frequent in MM. This manuscript describes approaches to identify disease-specific alterations in MM and proposes RNA-based therapeutic strategies to eradicate such alterations. As a "proof of concept", we examined the causes of aberrant HMMR (Hyaluronan-mediated motility receptor) splicing in MM. We identified clusters of single nucleotide variations (SNVs) in the HMMR transcript where the altered splicing took place. Using bioinformatics tools, we predicted SNVs and splicing factors that potentially contribute to aberrant HMMR splicing. Based on bioinformatic analyses and validation studies, we provided the rationale for RNA-based therapeutic strategies to selectively inhibit altered HMMR splicing in MM. Since splicing is a hallmark of many cancers, strategies described herein for target identification and the design of RNA-based therapeutics that inhibit gene splicing can be applied not only to other genes in MM but also more broadly to other hematological malignancies and solid tumors as well.
Collapse
Affiliation(s)
- Daisuke Ogiya
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Zuzana Chyra
- Department of Hemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic
| | - Sigitas J Verselis
- Molecular Diagnostic Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Morgan O'Keefe
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jacquelyn Cobb
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ivane Abiatari
- Institute of Medical and Public Health Research, School of Medicine, Ilia State University, Tbilisi, Georgia
| | - Srikanth Talluri
- Molecular Diagnostic Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Veterans Administration Boston Healthcare System, West Roxbury, MA, USA
| | - Anjana Anilkumar Sithara
- Department of Hemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michael P Chu
- Department of Medicine, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Roman Hájek
- Department of Hemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic
- Department of Hemato-oncology, University of Ostrava, Ostrava, Czech Republic
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Linda M Pilarski
- Department of Medicine, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Sophia Adamia
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Institute of Medical and Public Health Research, School of Medicine, Ilia State University, Tbilisi, Georgia.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Lin GR, Chen WR, Zheng PH, Chen WS, Cai GY. Circular RNA circ_0006089 promotes the progression of gastric cancer by regulating the miR-143-3p/PTBP3 axis and PI3K/AKT signaling pathway. J Dig Dis 2022; 23:376-387. [PMID: 35844201 DOI: 10.1111/1751-2980.13116] [Citation(s) in RCA: 4] [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: 12/30/2020] [Revised: 07/04/2022] [Accepted: 07/16/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Circular RNAs (circRNAs) play pivotal roles in malignancies including gastric cancer (GC). We aimed to investigate the biological function and regulatory mechanism of circ_0006089 in GC. METHODS Circ_0006089, microRNA (miR)-143-3p, and polypyrimidine tract-binding protein 3 (PTBP3) expressions were measured via quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in GC cell lines. Cell proliferative capacity was determined by colony formation and CCK-8 assays. Flow cytometry was employed for measuring cell apoptosis. Cell invasion and migration were measured via transwell and wound-healing assays. Western blot analysis was utilized for detecting protein expressions of E-cadherin, N-cadherin, vimentin, PTBP3, PI3K, p-PI3K, AKT, and p-AKT. Dual-reporter luciferase analysis was conducted to confirm the association between miR-143-3p and circ_0006089 or PTBP3. The role of circ_0006089 in vivo was detected via establishing a mice xenograft model. RESULTS Circ_0006089 expression was increased in GC. Circ_0006089 downregulation suppressed the proliferation and metastasis and induced apoptosis of GC cells, which was counteracted by miR-143-3p inhibition or PTBP3 overexpression. In addition, circ_0006089 overexpression could promote GC progression. MiR-143-3p specially bound to circ_0006089 and PTBP3 was targeted by miR-143-3p. Moreover, circ_0006089 could regulate PTBP3 expression and the PI3K/AKT pathway by sponging miR-143-3p. Circ_0006089 knockdown also suppressed tumor growth. CONCLUSION Circ_0006089 regulated miR-143-3p/PTBP3/PI3K/AKT pathway to facilitate GC progression.
Collapse
Affiliation(s)
- Guang Rong Lin
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Wei Rong Chen
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Pei Hong Zheng
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Wei Shan Chen
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Gao Yang Cai
- Department of General Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| |
Collapse
|
7
|
Fang Z, Li P, Li H, Chong W, Li L, Shang L, Li F. New Insights Into PTBP3 in Human Cancers: Immune Cell Infiltration, TMB, MSI, PDCD1 and m6A Markers. Front Pharmacol 2022; 13:811338. [PMID: 35359851 PMCID: PMC8960631 DOI: 10.3389/fphar.2022.811338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Polypyrimidine tract binding protein 3 (PTBP3) plays a critical role in post-transcriptional regulation. The role of PTBP3 in various human tumours was explored and analysed in this study based on the Cancer Genome Atlas and Gene Expression Omnibus datasets. PTBP3 was highly expressed in most tumours, such as breast invasive carcinoma, colon adenocarcinoma and hepatocellular carcinoma. PTBP3 overexpression generally predicts poor overall survival and disease-free survival in patients with adrenocortical carcinoma, lung squamous cell carcinoma, and pancreatic adenocarcinoma. However, low PTBP3 expression predicts poor prognosis in kidney renal clear cell carcinoma. We also explored PTBP3 genetic alterations in different tumour tissues. The result found that the frequency of PTBP3 alteration (>4%) was the highest in uterine tumours with “mutation” as the primary type. Furthermore, we found a significant correlation between PTBP3 expression and tumour mutational burden and microsatellite instability in various human tumours, and found that PTBP3 expression was positively correlated with TMB in ACC, STAD, PAAD, LUAD, and SARC. Two enhanced phosphorylation levels of S30 and S426 in colon cancer, ovarian cancer, and uterine corpus endometrial carcinoma were found. Further analysis indicated that PTBP3 expression was positively correlated with the cancer-associated fibroblasts for most tumour types. This study also found a relationship between immune checkpoints and N6-methyladenosine-related markers and PTBP3 expression. Moreover, the “mRNA surveillance pathway” and “RNA degradation” were involved in the functional mechanisms of PTBP3. These results provide new insights for molecular studies, and integrative analysis provided a framework for determining the predictive, prognostic, and therapeutic relevance of PTBP3 in cancer patients.
Collapse
Affiliation(s)
- Zhen Fang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Peijuan Li
- Emergency Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Han Li
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Liang Shang, ; Fei Li,
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Liang Shang, ; Fei Li,
| |
Collapse
|
8
|
Chen B, Chen W, Mu X, Yang L, Gu X, Zhao A, Liang X, Liu J. PTBP3 Induced Inhibition of Differentiation of Gastric Cancer Cells Through Alternative Splicing of Id1. Front Oncol 2020; 10:1477. [PMID: 32974175 PMCID: PMC7461954 DOI: 10.3389/fonc.2020.01477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 07/10/2020] [Indexed: 11/13/2022] Open
Abstract
Overexpression of PTBP3, a factor involved in alternative splicing, may inhibit the differentiation of leukemia cells. However, its role in gastric cancer differentiation and the specific pathways involved are unclear. In this study, we found that PTBP3 was upregulated in the poorly differentiated gastric cancer tissues. Patients with high levels of PTBP3 expression had significantly shorter survival than those with low PTBP3 expression. In gastric cancer cells, the regulatory effect of PTBP3 on alternative splicing of the Id1 gene was investigated. Following sodium butyrate-induced differentiation of MKN45 cells, the expression of Id1a decreased, but the expression of Id1b increased. RNA interference and overexpression experiments showed that PTBP3 upregulated Id1a expression and downregulated Id1b expression. RNA immunoprecipitation (RIP) assays indicated PTBP3 could interact with Id1. UV cross-linking assays indicated that PTBP3 interacted with the CU rich region of the Id1 gene. Two-hybrid experiments and a gel mobility shift assays found that Id1b had a more potent affinity for Hes1 than Id1a. Chromatin immunoprecipitation (ChIP) assays verified the association of Hes1 and the promoter of PTBP3 gene. Luciferase assays revealed that Hes1 bound the N-box sequence in the PTBP3 promoter. After silencing or overexpression of Hes1, PTBP3 protein expression remained unchanged. Thus, the loss of feedback regulation among PTBP3, Id1, and Hes1 in gastric cancer cells may be one of the causes of inhibited differentiation and malignant proliferation of these cells.
Collapse
Affiliation(s)
- Bin Chen
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weixia Chen
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyan Mu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyan Yang
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiangyu Gu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
9
|
Zhou W, She G, Yang K, Zhang B, Liu J, Yu B. MiR-384 inhibits proliferation and migration of trophoblast cells via targeting PTBP3. Pregnancy Hypertens 2020; 21:132-138. [PMID: 32512528 DOI: 10.1016/j.preghy.2020.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Preeclampsia (PE) is one type of hypertension during pregnancy that seriously threatens maternal and infant health. Trophoblast dysfunction, such as decreased proliferation and migration, is closely related to the occurrence and development of PE. MicroRNAs (miRNAs) have been proven to play an important role in many diseases, including PE. miR-384 was reported to play a regulatory role in promoting cell apoptosis and inhibiting proliferation, migration and invasion in a variety of tumors. Previously, we found that miR-384 is upregulated in the placenta and plasma in the context of PE. In this study, we elucidated the function of miR-384 in the trophoblast cell line HTR-8/SVneo and the trophoblastic tumor cell line JEG-3. Cell proliferation and migration were inhibited by miR-384 overexpression but promoted by miR-384 downregulation. Subsequently, polypyrimidine tract-binding protein 3(PTBP3) was found to be a direct target gene of miR-384. PTBP3 was downregulated in placental tissues from PE patients, and a negative correlation was found between PTBP3 and miR-384. Our results suggest that the miR-384/PTBP3 axis plays an important role in regulating trophoblast function during the progression of PE, and these data provide novel insight into the molecular pathogenesis of this disorder.
Collapse
Affiliation(s)
- Wenbo Zhou
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Guangtong She
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Kaiyan Yang
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Bin Zhang
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Jingbing Liu
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Bin Yu
- Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, Jiangsu, China.
| |
Collapse
|
10
|
Liang Y, Song X, Li Y, Chen B, Zhao W, Wang L, Zhang H, Liu Y, Han D, Zhang N, Ma T, Wang Y, Ye F, Luo D, Li X, Yang Q. LncRNA BCRT1 promotes breast cancer progression by targeting miR-1303/PTBP3 axis. Mol Cancer 2020; 19:85. [PMID: 32384893 PMCID: PMC7206728 DOI: 10.1186/s12943-020-01206-5] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/23/2020] [Indexed: 12/28/2022] Open
Abstract
Background Long noncoding RNAs (lncRNAs) play crucial roles in tumor progression and are aberrantly expressed in various cancers. However, the functional roles of lncRNAs in breast cancer remain largely unknown. Methods Based on public databases and integrating bioinformatics analyses, the overexpression of lncRNA BCRT1 in breast cancer tissues was detected and further validated in a cohort of breast cancer tissues. The effects of lncRNA BCRT1 on proliferation, migration, invasion and macrophage polarization were determined by in vitro and in vivo experiments. Luciferase reporter assay and RNA immunoprecipitation (RIP) were carried out to reveal the interaction between lncRNA BCRT1, miR-1303, and PTBP3. Chromatin immunoprecipitation (ChIP) and RT-PCR were used to evaluate the regulatory effect of hypoxia-inducible factor-1α (HIF-1α) on lncRNA BCRT1. Results LncRNA BCRT1 was significantly upregulated in breast cancer tissues, which was correlated with poor prognosis in breast cancer patients. LncRNA BCRT1 knockdown remarkably suppressed tumor growth and metastasis in vitro and in vivo. Mechanistically, lncRNA BCRT1 could competitively bind with miR-1303 to prevent the degradation of its target gene PTBP3, which acts as a tumor-promoter in breast cancer. LncRNA BCRT1 overexpression could promote M2 polarization of macrophages, mediated by exosomes, which further accelerated breast cancer progression. Furthermore, lncRNA BCRT1 was upregulated in response to hypoxia, which was attributed to the binding of HIF-1α to HREs in the lncRNA BCRT1 promoter. Conclusions Collectively, these results reveal a novel HIF-1α/lncRNA BCRT1/miR-1303/PTBP3 pathway for breast cancer progression and suggest that lncRNA BCRT1 might be a potential biomarker and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Yiran Liang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaojin Song
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yaming Li
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Bing Chen
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Wenjing Zhao
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Lijuan Wang
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Hanwen Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Ying Liu
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Dianwen Han
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Ning Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Tingting Ma
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Yajie Wang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Fangzhou Ye
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Dan Luo
- Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Xiaoyan Li
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China
| | - Qifeng Yang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China. .,Pathology Tissue Bank, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People's Republic of China.
| |
Collapse
|
11
|
Gao M, Dang F, Deng C. β-Cryptoxanthin induced anti-proliferation and apoptosis by G0/G1 arrest and AMPK signal inactivation in gastric cancer. Eur J Pharmacol 2019; 859:172528. [PMID: 31288004 DOI: 10.1016/j.ejphar.2019.172528] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 01/08/2023]
Abstract
β-Cryptoxanthin has been associated with reduced-risk of some cancers. However, the mechanisms of β-cryptoxanthin still remain unclearly understood in gastric cancer (GC). In this study, we examined the effect of β-cryptoxanthin on AMPK signal in human gastric cancer cells. AGS and SGC-7901 cells were treated with β-cryptoxanthin (0-40 μM) and AGS cells were injected in BALB/c (nu/nu) mice to analyze the effect of β-cryptoxanthin on GC. We found that β-cryptoxanthin induced inhibitory effect on the cell viability in a time- and concentration-dependent manner. The number of migrated cells and protein levels of matrix metalloproteinase (MMP) -2 and MMP-9 were obviously decreased. β-Cryptoxanthin treatment induced G0/G1 arrest, and reduced the expression of Cyclin E, Cyclin D1, cyclin-dependent kinases (CDK) of CDK4 and CDK6, and increased the expression of p53 and p21 in the two GC cells. Additionally, β-cryptoxanthin induced apoptosis and increased the expression of cleaved caspase-3, -8, -9 as well as cytochrome C (cyt C). β-Cryptoxanthin induced AMP-activated protein kinase (AMPK) signal inactivation by the down-regulation of protein kinase A (PKA), p-AMPK, eukaryotic elongation factor 2 kinase (eEF2k). Furthermore, β-cryptoxanthin inhibited tumor growth through suppressing the tumor volume and weight, inducing apoptotic cells. Besides, β-cryptoxanthin induced significant reductions of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9). In conclusion, our data provide the novel evidence to understand the mechanism of anti-pcancer of β-cryptoxanthin and indicate that β-cryptoxanthin can serve as a promising chemopreventive agent against gastric cancer.
Collapse
Affiliation(s)
- Meili Gao
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Fan Dang
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Chun Deng
- Department of Biological Science and Engineering, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
12
|
Monteiro LF, Forti FL. Network analysis of DUSP12 partners in the nucleus under genotoxic stress. J Proteomics 2019; 197:42-52. [PMID: 30779967 DOI: 10.1016/j.jprot.2019.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/23/2019] [Accepted: 02/12/2019] [Indexed: 01/01/2023]
Abstract
Dual Specificity Phosphatase 12 is a member of the Atypical DUSP Protein Tyrosine Phosphatase family, meaning that it does not contain typical MAP kinase targeting motifs, while being able to dephosphorylate tyrosine and serine/threonine residues. DUSP12 contains, apart from its catalytic domain, a zinc finger domain, making it one of the largest DUSPs, which displays strong nuclear expression in several tissues. In this work we identified nuclear targets of DUSP12 in two different cancer cell lines (A549 and MCF-7), challenging them with genotoxic stimuli to observe the effect on the networks and to link existing information about DUSP12 functions to the data obtained though mass spectrometry. We found network connections to the cytoskeleton (e.g. IQGAP1), to the chromatin (e.g. HP1BP3), to the splicing machinery and to the previously known pathway of ribosome maturation (e.g. TCOF1), which draw insight into many of the functions of this phosphatase, much likely connecting it to distinct, previously unknown genomic stability mechanisms.
Collapse
Affiliation(s)
- Lucas Falcão Monteiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Fábio Luís Forti
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
13
|
Huang J, Qian Z, Gong Y, Wang Y, Guan Y, Han Y, Yi X, Huang W, Ji L, Xu J, Su M, Yuan Q, Cui S, Zhang J, Bao C, Liu W, Chen X, Zhang M, Gao X, Wu R, Zhang Y, Xu H, Zhu S, Zhu H, Yang L, Xu X, Zhou P, Liang Z. Comprehensive genomic variation profiling of cervical intraepithelial neoplasia and cervical cancer identifies potential targets for cervical cancer early warning. J Med Genet 2018; 56:186-194. [PMID: 30567904 PMCID: PMC6581088 DOI: 10.1136/jmedgenet-2018-105745] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 12/31/2022]
Abstract
Background To better understand the pathogenesis of cervical cancer (CC), we systematically analysed the genomic variation and human papillomavirus (HPV) integration profiles of cervical intraepithelial neoplasia (CIN) and CC. Methods We performed whole-genome sequencing or whole-exome sequencing of 102 tumour-normal pairs and human papillomavirus probe capture sequencing of 45 CCs, 44 CIN samples and 25 normal cervical samples, and constructed strict integrated workflow of genomic analysis. Results Mutational analysis identified eight significantly mutated genes in CC including four genes (FAT1, MLL3, MLL2 and FADD), which have not previously been reported in CC. Targetable alterations were identified in 55.9% of patients. In addition, HPV integration breakpoints occurred in 97.8% of the CC samples, 70.5% of the CIN samples and 42.8% of the normal cervical samples with HPV infection. Integrations of high-risk HPV strains in CCs, including HPV16, 18, 33 and 58, also occurred in the CIN samples. Moreover, gene mutations were detected in 52% of the CIN specimens, and 54.8% of these mutations occurred in genes that also mutated in CCs. Conclusion Our results lay the foundation for a deep understanding of the molecular mechanisms and finding new diagnostic and therapeutic targets of CC.
Collapse
Affiliation(s)
- Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome at Shanghai, Shanghai, China
| | - Zhaoyang Qian
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Yanzhou Wang
- Department of Obstetrics and Gynecology, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | | | - Yingxin Han
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yi
- Geneplus-Beijing, Beijing, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liyan Ji
- Geneplus-Beijing, Beijing, China
| | - Jiajia Xu
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Mengyuan Su
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Qing Yuan
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shujian Cui
- Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome at Shanghai, Shanghai, China
| | - Jinling Zhang
- Shenzhen People’s Hospital, Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Chaohui Bao
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weilong Liu
- Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Third People’s Hospital, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Xi Chen
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Ming Zhang
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Xiaohuan Gao
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Renhua Wu
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Yinxin Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huicheng Xu
- Department of Obstetrics and Gynecology, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Shida Zhu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Hongmei Zhu
- Binhai Genomics Institute, BGI-Tianjin, Tianjin, China
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | | | - Xun Xu
- BGI-Shenzhen, Shenzhen, Guangdong, China
| | - Pingyu Zhou
- STD Institute, Shanghai Skin Disease Hospital, Tong Ji University, Shanghai, China
| | - Zhiqing Liang
- Department of Obstetrics and Gynecology, Southwestern Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
14
|
Zhou Y, Zou H, Wu E, Huang L, Yin R, Mei Y, Zhu X. Overexpression of ROD1 inhibits invasion of breast cancer cells by suppressing the translocation of β-catenin into the nucleus. Oncol Lett 2018; 16:2645-2653. [PMID: 30013660 DOI: 10.3892/ol.2018.8917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 04/16/2018] [Indexed: 12/14/2022] Open
Abstract
The incidence of breast cancer is increasing throughout the world. Although significant progress has been made in diagnostic techniques and targeted therapies, the prognosis of breast cancer remains poor. Regulator of differentiation 1 (ROD1) may inhibit the development of several types of cancer. However, the role of ROD1 in breast cancer cells remains unknown. In the present study, western blot analysis and reverse transcription-quantitative polymerase chain reaction revealed that expression of ROD1 was significantly reduced in breast cancer cells. Overexpression of ROD1 reduced the proliferation rate, demonstrated using a Cell Counting Kit-8 assay. Additionally, the overexpression of ROD1 decreased the invasiveness of breast cancer cells, indicating that ROD1 may serve as a tumor suppressor. Additionally, the data suggested that ROD1 significantly suppressed the activity of Wnt luciferase reporter (TOP Flash) in MDA-MB-231 cells. Furthermore, it was demonstrated that ROD1 may interact with β-catenin by using co-immunoprecipitation, resulting in suppression of β-catenin migration into the nucleus. Notably, ROD1 demonstrated its anticancer effect by decreasing β-catenin (Y333) phosphorylation in a nude mouse xenograft model. Overexpression of ROD1 may downregulate Ki67 protein levels, as determined by immunohistochemistry. These results indicated that ROD1 may be used as a therapeutic target in patients with breast cancer.
Collapse
Affiliation(s)
- Ya Zhou
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Hanqing Zou
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Enhao Wu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Lei Huang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Rui Yin
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Yuxin Mei
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xun Zhu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
15
|
PTBP3 contributes to the metastasis of gastric cancer by mediating CAV1 alternative splicing. Cell Death Dis 2018; 9:569. [PMID: 29752441 PMCID: PMC5948206 DOI: 10.1038/s41419-018-0608-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/23/2022]
Abstract
Polypyrimidine tract-binding protein 3 (PTBP3) is an essential RNA-binding protein with roles in RNA splicing, 3' end processing and translation. Although increasing evidence implicates PTBP3 in several cancers, its role in gastric cancer metastasis remains poorly explored. In this study, we found that PTBP3 was upregulated in the gastric cancer tissues of patients with lymph node metastasis. Patients with high PTBP3 expression levels had significantly shorter survival than those with low PTBP3 expression. Overexpression/knockdown of PTBP3 expression had no effect on proliferation, whereas it regulated migration and invasion in vitro. In addition, when a mouse xenotransplant model of MKN45 was established, knockdown of PTBP3 in MKN45 cells caused the formation of tumours that were smaller in size than their counterparts, with suppression of tumour lymphangiogenesis and metastasis to regional lymph nodes. Furthermore, we identified caveolin 1 (CAV1) as a downstream target of PTBP3. RNA immunoprecipitation (RIP) assays and dual-luciferase reporter gene assays indicated that PTBP3 interacted with the CU-rich region of the CAV1 gene to downregulate CAV1α expression. Knockdown of CAV1α abrogated the reduction of FAK and Src induced by PTBP3 knockdown. In summary, our findings provide experimental evidence that PTBP3 may function as a metastatic gene in gastric cancer by regulating CAV1 through alternative splicing.
Collapse
|
16
|
Hou P, Li L, Chen F, Chen Y, Liu H, Li J, Bai J, Zheng J. PTBP3-Mediated Regulation of ZEB1 mRNA Stability Promotes Epithelial-Mesenchymal Transition in Breast Cancer. Cancer Res 2017; 78:387-398. [PMID: 29187406 DOI: 10.1158/0008-5472.can-17-0883] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 07/28/2017] [Accepted: 11/20/2017] [Indexed: 11/16/2022]
Abstract
The RNA polypyrimidine tract-binding protein PTBP3 is a little studied paralog of PTBP1, which has oncogenic properties. In this study, we demonstrate that PTBP3 induces epithelial-mesenchymal transition (EMT) in breast tumor cells and promotes their invasive growth and metastasis. Elevated expression of PTBP3 associated significantly with lymph node metastasis, advanced histology grade, TNM stage, and poor 5-year overall survival of patients. In human mammary epithelial cells, PTBP3 overexpression was sufficient to induce EMT and to enhance cell migration, invasion, and cancer stem-like cell properties. PTBP3 regulated expression of the EMT regulatory transcription factor ZEB1 by binding the 3'UTR of its mRNA, thereby preventing its degradation. Conversely, ZEB1 ablation blocked the ability of PTBP3 to induce EMT. Overall, our findings define PTBP3 as a regulator of EMT that acts by governing expression of ZEB1, and they establish an oncogenic function of PTBP3, suggesting its candidacy as a theranostic target.Significance: These findings define PTBP3 as a regulator of EMT that acts by governing expression of ZEB1, and they establish an oncogenic function of PTBP3, suggesting its candidacy as a theranostic target. Cancer Res; 78(2); 387-98. ©2017 AACR.
Collapse
Affiliation(s)
- Pingfu Hou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lin Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fang Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yansu Chen
- School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui Liu
- School of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingjing Li
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
17
|
Xia Q, Zhao Y, Wang J, Qiao W, Zhang D, Yin H, Xu D, Chen F. Proteomic analysis of cell cycle arrest and differentiation induction caused by ATPR, a derivative of all-trans retinoic acid, in human gastric cancer SGC-7901 cells. Proteomics Clin Appl 2017; 11. [PMID: 28164444 DOI: 10.1002/prca.201600099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/31/2016] [Accepted: 02/02/2017] [Indexed: 12/16/2022]
Abstract
PURPOSE 4-amino-2-trifluoromethyl-phenyl retinate (ATPR) was reported to potentially inhibit proliferation and induce differentiation activity in some tumor cells. In this study, a proteomics approach was used to investigate the possible mechanism by screening the differentially expressed protein profiles of SGC-7901 cells before and after ATPR-treatment in vitro. EXPERIMENTAL DESIGN Peptides digested from the total cellular proteins were analyzed by reverse phase LC-MS/MS followed by a label-free quantification analysis. The SEQUEST search engine was used to identify proteins and bioinformatics resources were used to investigate the involved pathways for the differentially expressed proteins. RESULTS Thirteen down-regulated proteins were identified in the ATPR-treated group. Bioinformatics analysis showed that the effects of ATPR on 14-3-3ε might potentially involve the PI3K-AKT-FOXO pathway and P27Kip1 expression. Western blot and RT-PCR analysis showed that ATPR could inhibit AKT phosphorylation, up-regulate the expression of FOXO1A and P27Kip1 at both the protein and mRNA levels, and down-regulate the cytoplasmic expression of cyclin E and CDK2. ATPR-induced G0/G1 phase arrest and differentiation can be ablated if the P27kip1 gene is silenced with sequence-specific siRNA or in 14-3-3ε overexpression of SGC-7901 cells. CONCLUSION AND CLINICAL RELEVANCE ATPR might cause cell cycle arrest and differentiation in SGC-7901 cells by simultaneously inhibiting the phosphorylation of AKT and down-regulating 14-3-3ε. This change would then enhance the inhibition of cyclin E/CDK2 by up-regulating FOXO1A and P27Kip1. Our findings could be of value for finding new drug targets and for developing more effective differentiation inducer.
Collapse
Affiliation(s)
- Quan Xia
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yingli Zhao
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiali Wang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wenhao Qiao
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Dongling Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Hao Yin
- Chromatography and Mass Spectrometry Lab Hefei National Laboratory for Physical Science at Microscale, University of Science and Technology of China, Hefei, China
| | - Dujuan Xu
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Feihu Chen
- School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
18
|
Liang X, Shi H, Yang L, Qiu C, Lin S, Qi Y, Li J, Zhao A, Liu J. Inhibition of polypyrimidine tract-binding protein 3 induces apoptosis and cell cycle arrest, and enhances the cytotoxicity of 5- fluorouracil in gastric cancer cells. Br J Cancer 2017; 116:903-911. [PMID: 28222070 PMCID: PMC5379144 DOI: 10.1038/bjc.2017.32] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Human polypyrimidine tract binding protein 3 (PTBP3) was first discovered in 1999 and has been well characterised as a differentiation regulator. However, its role in human cancer has rarely been reported. Our previous study revealed increased PTBP3 protein level in gastric cancer tissues. Downregulation of PTBP3 suppressed the proliferation and differentiation of gastric cancer cells in vivo. Methods: PTBP3 mRNA levels in human gastric cancer and adjuvant non-tumour tissues were detected. Apoptosis and 5-FU effect were determined in PTBP3-silenced gastric cancer cells. Underlying molecular mechanisms were investigated. Results: MRNA expression of PTBP3 was upregulated in gastric cancer tissues, especially in those at an advanced stage. PTBP3 silencing led to apoptosis, under which modulation of PTB and thereby switch of Bcl-x pre-mRNA splicing pattern might be an important mechanism. Further research found that inhibition of PTBP3 expression enhanced the chemosensitivity of gastric cancer cells towards 5-FU treatment. This was mediated by reduced expression of histone deacetylase 6 (HDAC6), which further inhibited the phosphorylation of Akt and the expression of thymidylate synthase (TYMS), the critical determinant of 5-FU cytotoxicity. Conclusions: PTBP3 might serve as a biomarker of gastric cancer or potential target for anti-cancer therapy.
Collapse
Affiliation(s)
- Xin Liang
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Haiyang Shi
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Liyan Yang
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Cen Qiu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Shengchao Lin
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Yingxue Qi
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| | - Jiyu Li
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Yanchang Road, Shanghai 200072, China
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, PO Box 268, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|