1
|
Chen X, Gong R, Wang J, Ma B, Lei K, Ren H, Wang J, Zhao C, Wang L, Yu Q. Identification of HnRNP Family as Prognostic Biomarkers in Five Major Types of Gastrointestinal Cancer. Curr Gene Ther 2022; 22:449-461. [PMID: 35794744 PMCID: PMC9906633 DOI: 10.2174/1566523222666220613113647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Heterogeneous nuclear ribonucleoproteins (hnRNPs), a large family of RNAbinding proteins, have been implicated in tumor progression in multiple cancer types. However, the expression pattern and prognostic value of hnRNPs in five gastrointestinal (GI) cancers, including gastric, colorectal, esophageal, liver, and pancreatic cancer, remain to be investigated. OBJECTIVE The current research aimed to identify prognostic biomarkers of the hnRNP family in five major types of gastrointestinal cancer. METHODS Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA), and Kaplan-Meier Plotter were used to explore the hnRNPs expression levels concerning clinicopathological parameters and prognostic values. The protein level of hnRNPU was validated by immunohistochemistry (IHC) in human tissue specimens. Genetic alterations of hnRNPs were analyzed using cBioportal, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to illustrate the biological functions of co-expressed genes of hnRNPs. RESULTS The vast majority of hnRNPs were highly expressed in five types of GI cancer tissues compared to their adjacent normal tissues, and mRNA levels of hnRNPA2B1, D, Q, R, and U were significantly different in various GI cancer types at different stages. In addition, Kaplan-Meier analysis revealed that the increased hnRNPs expression levels were correlated with better prognosis in gastric and rectal cancer patients (log-rank p < 0.05). In contrast, patients with high levels of hnRNPs exhibited a worse prognosis in esophageal and liver cancer (log-rank p < 0.05). Using immunohistochemistry, we further confirmed that hnRNPU was overexpressed in gastric, rectal, and liver cancers. In addition, hnRNPs genes were altered in patients with GI cancers, and RNA-related processing was correlated with hnRNPs alterations. CONCLUSION We identified differentially expressed genes of hnRNPs in tumor tissues versus adjacent normal tissues, which might contribute to predicting tumor types, early diagnosis, and targeted therapies in five major types of GI cancer.
Collapse
Affiliation(s)
- Xianghan Chen
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao 266071, China;,Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China;,These authors contribute to this work equally.
| | - Ruining Gong
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China;,Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China;,These authors contribute to this work equally.
| | - Jia Wang
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Boyi Ma
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China;,Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Ke Lei
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - He Ren
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China;,Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chenyang Zhao
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lili Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China,Address correspondence to these authors at the Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266000, China; Tel/Fax: 86-532-82917308; E-mail: and Department of Pathology, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266003, China; Tel/Fax: 86-532- 82919350; E-mail:
| | - Qian Yu
- Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, Qingdao 266000, China;,Address correspondence to these authors at the Center of Tumor Immunology and Cytotherapy, Medical Research Center of the Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266000, China; Tel/Fax: 86-532-82917308; E-mail: and Department of Pathology, The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266003, China; Tel/Fax: 86-532- 82919350; E-mail:
| |
Collapse
|
2
|
GPX2 stabilized by PCBP2 induces autophagy to protect Het-1A esophageal cells from apoptosis and inflammation. Cell Signal 2022; 97:110397. [DOI: 10.1016/j.cellsig.2022.110397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022]
|
3
|
Kattuah W, Rogelj B, King A, Shaw CE, Hortobágyi T, Troakes C. Heterogeneous Nuclear Ribonucleoprotein E2 (hnRNP E2) Is a Component of TDP-43 Aggregates Specifically in the A and C Pathological Subtypes of Frontotemporal Lobar Degeneration. Front Neurosci 2019; 13:551. [PMID: 31213972 PMCID: PMC6558155 DOI: 10.3389/fnins.2019.00551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is the major component of the ubiquitin-positive protein aggregates seen in the majority of frontotemporal lobar degeneration and amyotrophic lateral sclerosis cases. TDP-43 belongs to the heterogeneous nuclear ribonucleoprotein (hnRNP) family that is involved in the regulation of RNA transcription, splicing, transport and translation. There are a great many hnRNPs, which often have overlapping functions and act cooperatively in RNA processing. Here we demonstrate that another hnRNP family member, hnRNP E2, shows a striking accumulation within dystrophic neurites and cytoplasmic inclusions in the frontal cortex and hippocampus of a subset of FTLD-TDP cases belonging to pathological subtypes A and C, where hnRNP E2 was found to co-localize with 87% of TDP-43 immunopositive inclusions. hnRNP E2-positive inclusions were not seen in FTLD-TDP cases with the C9orf72 expansion or in any other neurodegenerative disorders examined. This interaction with TDP-43 in specific FTLD subtypes suggests different underlying neurodegenerative pathways.
Collapse
Affiliation(s)
- Wejdan Kattuah
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Boris Rogelj
- Department of Biotechnology, Jozef Stefan Institute, Ljubljana, Slovenia.,Biomedical Research Institute BRIS, Ljubljana, Slovenia.,Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Andrew King
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Christopher E Shaw
- UK Dementia Research Institute, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Pathology, University of Szeged, Szeged, Hungary.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Claire Troakes
- London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
4
|
Chen C, Lei J, Zheng Q, Tan S, Ding K, Yu C. Poly(rC) binding protein 2 (PCBP2) promotes the viability of human gastric cancer cells by regulating CDK2. FEBS Open Bio 2018; 8:764-773. [PMID: 29744291 PMCID: PMC5929926 DOI: 10.1002/2211-5463.12408] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/09/2018] [Accepted: 02/14/2018] [Indexed: 12/12/2022] Open
Abstract
Survival rates for patients with gastric cancer, especially the advanced form, remain poor and the development of targeted treatments is hampered by a lack of efficient biological targets. Poly(rC) binding protein 2 (PCBP2) is an RNA-binding protein that contributes to mRNA stabilization, translational silencing and enhancement and it has been implicated as a promoter of gastric cancer growth. In the present study, we demonstrated that the expression level of PCBP2 was higher in human gastric cancer tissues compared to adjacent normal gastric tissues. A high level of PCBP2 was correlated with worse postoperative relapse-free survival and overall survival rates of gastric cancer patients. Small hairpin RNA-mediated depletion of PCBP2 dramatically decreased the viability of gastric cancer cells. Cyclin-dependent kinase 2 (CDK2) was positively regulated by PCBP2 via a direct 3' UTR binding pathway as determined using a ribonucleoprotein immunoprecipitation assay and a biotin pulldown assay. CDK2 mediated the promoting role of PCBP2. These results suggest that PCBP2 acts as an oncogene in human gastric cancer cells and that functionally depleting PCBP2 could be considered as a potential target for gastric cancer therapy.
Collapse
Affiliation(s)
- Changyu Chen
- Department of General Surgery (Gastrointestinal Surgery) The First Affiliated Hospital of Anhui Medicial University Hefei China
| | - Jun Lei
- Department of General Surgery (Gastrointestinal Surgery) The First Affiliated Hospital of Anhui Medicial University Hefei China
| | - Qiang Zheng
- Department of General Surgery (Gastrointestinal Surgery) The First Affiliated Hospital of Anhui Medicial University Hefei China
| | - Sheng Tan
- Laboratory of Molecular Tumor Pathology School of Life Science University of Science and Technology of China Hefei China
| | - Keshuo Ding
- Department of Pathology Anhui Medical University Hefei China
| | - Changjun Yu
- Department of General Surgery (Gastrointestinal Surgery) The First Affiliated Hospital of Anhui Medicial University Hefei China
| |
Collapse
|
5
|
High expression of PCBP2 is associated with progression and poor prognosis in patients with glioblastoma. Biomed Pharmacother 2017; 94:659-665. [PMID: 28787701 DOI: 10.1016/j.biopha.2017.07.103] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Poly(C)-binding protein 2 (PCBP2) has been found to have ambiguous functions in a variety of cancers. However, the specific biological function of PCBP2 and its mechanism in glioblastoma remain unclear. We investigated the expression of PCBP2 in 143 glioblastoma specimens to explore the linkage between PCBP2 expression and clinicopathological parameters as well as clinical significance. Furthermore, the underlying mechanisms of PCBP2 on glioblastoma progression were discussed in vitro. METHODS The transcriptional and translational levels of PCBP2 in 143 glioblastoma patients were detected by quantitative Real-time PCR (qRT-PCR) and western blot. The association of prognostic outcomes and PCBP2 expression was evaluated using Kaplan-Meier analysis. RESULTS PCBP2 expression was markedly increased in higher stages of glioblastoma compared with those in lower stages (P<0.001). High expression of PCBP2 was associated with higher clinical stage and histological grade (P<0.001). Further research suggested that PCBP2 upregulation was connected with poorer prognosis in patients with glioblastoma (P<0.001). Moreover, PCBP2 knockdown could significantly decreased the colony formation and invasion capability of glioblastoma cells (P<0.01). Conversely, PCBP2 overexpression could increase the colony formation and invasion capability (P<0.01). CONCLUSION These findings indicated that PCBP2 might be a novel prognostic biomarker and a potential therapeutic target of glioblastoma.
Collapse
|
6
|
Ye J, Zhou G, Zhang Z, Sun L, He X, Zhou J. Poly (C)-binding protein 2 (PCBP2) promotes the progression of esophageal squamous cell carcinoma (ESCC) through regulating cellular proliferation and apoptosis. Pathol Res Pract 2016; 212:717-25. [DOI: 10.1016/j.prp.2016.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 04/21/2016] [Accepted: 05/25/2016] [Indexed: 12/14/2022]
|
7
|
PCBP2 Modulates Neural Apoptosis and Astrocyte Proliferation After Spinal Cord Injury. Neurochem Res 2016; 41:2401-14. [DOI: 10.1007/s11064-016-1953-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 04/05/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022]
|
8
|
Ren C, Zhang J, Yan W, Zhang Y, Chen X. RNA-binding Protein PCBP2 Regulates p73 Expression and p73-dependent Antioxidant Defense. J Biol Chem 2016; 291:9629-37. [PMID: 26907686 DOI: 10.1074/jbc.m115.712125] [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: 12/22/2015] [Indexed: 11/06/2022] Open
Abstract
TAp73, a member of the p53 family tumor suppressors, plays a critical rule in tumor suppression and neuronal development. However, how p73 activity is controlled at the posttranscriptional level is not well understood. Here, we showed that TAp73 activity is regulated by RNA-binding protein PCBP2. Specifically, we found that knockdown or knock-out of PCBP2 reduces, whereas ectopic expression of PCBP2 increases, TAp73 expression. We also showed that PCBP2 is necessary for p73 mRNA stability via the CU-rich elements in p73 3'-UTR. To uncover the biological relevance of PCBP2-regulated TAp73 expression, we showed that ectopic expression of PCBP2 inhibits, whereas knockdown or knock-out of PCBP2 increases, the production of reactive oxygen species (ROS) in a TAp73-dependent manner. Additionally, we found that glutaminase 2 (GLS2), a modulator of p73-dependent antioxidant defense, is also involved in PCBP2-regulated ROS production. Moreover, we generated PCBP2-deficient mice and primary mouse embryonic fibroblasts (MEFs) and showed that loss of PCBP2 leads to decreased p73 expression and, subsequently, increased ROS production and accelerated cellular senescence. Together, our data suggest that PCBP2 regulates p73 expression via mRNA stability and p73-dependent biological function in ROS production and cellular senescence.
Collapse
Affiliation(s)
- Cong Ren
- From the Comparative Oncology Laboratory, Schools of Medicine and Veterinary Medicine, University of California, Davis, California 95616
| | - Jin Zhang
- From the Comparative Oncology Laboratory, Schools of Medicine and Veterinary Medicine, University of California, Davis, California 95616
| | - Wensheng Yan
- From the Comparative Oncology Laboratory, Schools of Medicine and Veterinary Medicine, University of California, Davis, California 95616
| | - Yanhong Zhang
- From the Comparative Oncology Laboratory, Schools of Medicine and Veterinary Medicine, University of California, Davis, California 95616
| | - Xinbin Chen
- From the Comparative Oncology Laboratory, Schools of Medicine and Veterinary Medicine, University of California, Davis, California 95616
| |
Collapse
|
9
|
Wan C, Gong C, Zhang H, Hua L, Li X, Chen X, Chen Y, Ding X, He S, Cao W, Wang Y, Fan S, Xiao Y, Zhou G, Shen A. β2-adrenergic receptor signaling promotes pancreatic ductal adenocarcinoma (PDAC) progression through facilitating PCBP2-dependent c-myc expression. Cancer Lett 2016; 373:67-76. [PMID: 26803058 DOI: 10.1016/j.canlet.2016.01.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 01/14/2016] [Accepted: 01/15/2016] [Indexed: 02/06/2023]
Abstract
The β2-adrenergic receptor (β2-AR) plays a crucial role in pancreatic ductal adenocarcinoma (PDAC) progression. In this report, we identified poly(rC)-binding protein 2 (PCBP2) as a novel binding partner for β2-AR using immunoprecipitation-mass spectrometry (IP-MS) approach. The association between β2-AR and PCBP2 was verified using reciprocal immunoprecipitation. Importantly, we found significant interaction and co-localization of the two proteins in the presence of β2-AR agonist in Panc-1 and Bxpc3 PDAC cells. β2-AR-induced recruitment of PCBP2 led to augmented protein level of c-myc in PDAC cells, likely as a result of enhanced internal ribosome entry segment (IRES)-mediated translation of c-myc. The activation of β2-AR accelerated cell proliferation and colony formation, while knockdown of PCBP2 or c-myc restrained the effect. Furthermore, overexpression of PCBP2 was observed in human PDAC cell lines and tissue specimens compared to the normal pancreatic ductal epithelial cells and the non-cancerous tissues respectively. Overexpression of β2-AR and PCBP2 was associated with advanced tumor stage and significantly worsened prognosis in patients with PDAC. Our results elucidate a new molecular mechanism by which β2-AR signaling facilitates PDAC progression through triggering PCBP2-dependent c-myc expression.
Collapse
Affiliation(s)
- Chunhua Wan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China; Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Chen Gong
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Haifeng Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Lu Hua
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xiaohong Li
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xudong Chen
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu Province 226001, China
| | - Yinji Chen
- National Engineering Lab of Food Storage and Transportation, Nanjing University of Finance and Economics, Nanjing, Jiangsu Province 210023, China
| | - Xiaoling Ding
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Song He
- Department of Pathology, Nantong University Cancer Hospital, Nantong, Jiangsu Province 226001, China
| | - Wei Cao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yingying Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Shaoqing Fan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ying Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Aiguo Shen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
10
|
Regulation of Stem Cell Self-Renewal and Oncogenesis by RNA-Binding Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 907:153-88. [PMID: 27256386 DOI: 10.1007/978-3-319-29073-7_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Throughout their life span, multicellular organisms rely on stem cell systems. During development pluripotent embryonic stem cells give rise to all cell types that make up the organism. After birth, tissue stem cells maintain properly functioning tissues and organs under homeostasis as well as promote regeneration after tissue damage or injury. Stem cells are capable of self-renewal, which is the ability to divide indefinitely while retaining the potential of differentiation into multiple cell types. The ability to self-renew, however, is a double-edged sword; the molecular mechanisms of self-renewal can be a target of malignant transformation driving tumor development and progression. Growing lines of evidence have shown that RNA-binding proteins (RBPs) play pivotal roles in the regulation of self-renewal by modulating metabolism of coding and non-coding RNAs both in normal tissues and in cancers. In this review, we discuss our current understanding of tissue stem cell systems and how RBPs regulate stem cell fates as well as how the regulatory functions of RBPs contribute to oncogenesis.
Collapse
|
11
|
Ito Y, Narita N, Nomi N, Sugimoto C, Takabayashi T, Yamada T, Karaya K, Matsumoto H, Fujieda S. Suppression of Poly(rC)-Binding Protein 4 (PCBP4) reduced cisplatin resistance in human maxillary cancer cells. Sci Rep 2015. [PMID: 26196957 PMCID: PMC4508830 DOI: 10.1038/srep12360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cisplatin plays an important role in the therapy for human head and neck cancers. However, cancer cells develop cisplatin resistance, leading to difficulty in treatment and poor prognosis. To analyze cisplatin-resistant mechanisms, a cisplatin-resistant cell line, IMC-3CR, was established from the IMC-3 human maxillary cancer cell line. Flow cytometry revealed that, compared with IMC-3 cells, cisplatin more dominantly induced cell cycle G2/M arrest rather than apoptosis in IMC-3CR cells. That fact suggests that IMC-3CR cells avoid cisplatin-induced apoptosis through induction of G2/M arrest, which allows cancer cells to repair damaged DNA and survive. In the present study, we specifically examined Poly(rC)-Binding Protein 4 (PCBP4), which reportedly induces G2/M arrest. Results showed that suppression of PCBP4 by RNAi reduced cisplatin-induced G2/M arrest and enhanced apoptosis in IMC-3CR cells, resulting in the reduction of cisplatin resistance. In contrast, overexpression of PCBP4 in IMC-3 cells induced G2/M arrest after cisplatin treatment and enhanced cisplatin resistance. We revealed that PCBP4 combined with Cdc25A and suppressed the expression of Cdc25A, resulting in G2/M arrest. PCBP4 plays important roles in the induction of cisplatin resistance in human maxillary cancers. PCBP4 is a novel molecular target for the therapy of head and neck cancers, especially cisplatin-resistant cancers.
Collapse
Affiliation(s)
- Yumi Ito
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Norihiko Narita
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Nozomi Nomi
- Department of Otorhinolaryngology, Faculty of Medical Sciences, University of Oita
| | - Chizuru Sugimoto
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Tetsuji Takabayashi
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Takechiyo Yamada
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Kazuhiro Karaya
- Division of Bioresearch, Life Science Research Laboratory, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Hideki Matsumoto
- Division of Oncology, Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medical Sciences, University of Fukui, 23 Shimoaizuki, Matsuoka, Eiheiji, Fukui, 910-1193, Japan
| |
Collapse
|
12
|
Zhu G, He Y, Yang S, Chen B, Zhou M, Xu XJ. Identification of Gene and MicroRNA Signatures for Oral Cancer Developed from Oral Leukoplakia. BIOMED RESEARCH INTERNATIONAL 2015; 2015:841956. [PMID: 26064958 PMCID: PMC4433644 DOI: 10.1155/2015/841956] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 10/14/2014] [Indexed: 11/17/2022]
Abstract
In clinic, oral leukoplakia (OLK) may develop into oral cancer. However, the mechanism underlying this transformation is still unclear. In this work, we present a new pipeline to identify oral cancer related genes and microRNAs (miRNAs) by integrating both gene and miRNA expression profiles. In particular, we find some network modules as well as their miRNA regulators that play important roles in the development of OLK to oral cancer. Among these network modules, 91.67% of genes and 37.5% of miRNAs have been previously reported to be related to oral cancer in literature. The promising results demonstrate the effectiveness and efficiency of our proposed approach.
Collapse
Affiliation(s)
- Guanghui Zhu
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| | - Yuan He
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Middle Yanchang Road 399, Shanghai 200072, China
| | - Shaofang Yang
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Middle Yanchang Road 399, Shanghai 200072, China
| | - Beimin Chen
- Shanghai Tenth People's Hospital, Middle Yanchang Road 30, Shanghai 200072, China
| | - Min Zhou
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Middle Yanchang Road 399, Shanghai 200072, China
| | - Xin-Jian Xu
- Department of Mathematics, Shanghai University, Shanghai 200444, China
| |
Collapse
|
13
|
Hu CE, Liu YC, Zhang HD, Huang GJ. The RNA-binding protein PCBP2 facilitates gastric carcinoma growth by targeting miR-34a. Biochem Biophys Res Commun 2014; 448:437-42. [PMID: 24796666 DOI: 10.1016/j.bbrc.2014.04.124] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 04/24/2014] [Indexed: 12/31/2022]
Abstract
Gastric carcinoma is the fourth most common cancer worldwide, with a high rate of death and low 5-year survival rate. However, the mechanism underling gastric cancer is still not fully understood. Here in the present study, we identify the RNA-binding protein PCBP2 as an oncogenic protein in human gastric carcinoma. Our results show that PCBP2 is up-regulated in human gastric cancer tissues compared to adjacent normal tissues, and that high level of PCBP2 predicts poor overall and disease-free survival. Knockdown of PCBP2 in gastric cancer cells inhibits cell proliferation and colony formation in vitro, whereas opposing results are obtained when PCBP2 is overexpressed. Our in vivo subcutaneous xenograft results also show that PCBP2 can critically regulate gastric cancer cell growth. In addition, we find that PCBP2-depletion induces apoptosis in gastric cancer cells via up-regulating expression of pro-apoptotic proteins and down-regulating anti-apoptotic proteins. Mechanically, we identify that miR-34a as a target of PCBP2, and that miR-34a is critically essential for the function of PCBP2. In summary, PCBP2 promotes gastric carcinoma development by regulating the level of miR-34a.
Collapse
Affiliation(s)
- Cheng-En Hu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong-Chao Liu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hui-Dong Zhang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai, China
| | - Guang-Jian Huang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Kooistra SM, Nørgaard LCR, Lees MJ, Steinhauer C, Johansen JV, Helin K. A screen identifies the oncogenic micro-RNA miR-378a-5p as a negative regulator of oncogene-induced senescence. PLoS One 2014; 9:e91034. [PMID: 24651706 PMCID: PMC3961217 DOI: 10.1371/journal.pone.0091034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/06/2014] [Indexed: 12/15/2022] Open
Abstract
Oncogene-induced senescence (OIS) can occur in response to hyperactive oncogenic signals and is believed to be a fail-safe mechanism protecting against tumorigenesis. To identify new factors involved in OIS, we performed a screen for microRNAs that can overcome or inhibit OIS in human diploid fibroblasts. This screen led to the identification of miR-378a-5p and in addition several other miRNAs that have previously been shown to play a role in senescence. We show that ectopic expression of miR-378a-5p reduces the expression of several senescence markers, including p16INK4A and senescence-associated β-galactosidase. Moreover, cells with ectopic expression of miR-378a-5p retain proliferative capacity even in the presence of an activated Braf oncogene. Finally, we identified several miR-378a-5p targets in diploid fibroblasts that might explain the mechanism by which the microRNA can delay OIS. We speculate that miR-378a-5p might positively influence tumor formation by delaying OIS, which is consistent with a known pro-oncogenic function of this microRNA.
Collapse
Affiliation(s)
- Susanne Marije Kooistra
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Lise Christine Rudkjær Nørgaard
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Michael James Lees
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Cornelia Steinhauer
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Jens Vilstrup Johansen
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; The Bioinformatics Centre, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Helin
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark; Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark; The Danish Stem Cell Center (DanStem), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Han W, Xin Z, Zhao Z, Bao W, Lin X, Yin B, Zhao J, Yuan J, Qiang B, Peng X. RNA-binding protein PCBP2 modulates glioma growth by regulating FHL3. J Clin Invest 2013; 123:2103-18. [PMID: 23585479 DOI: 10.1172/jci61820] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/07/2013] [Indexed: 02/01/2023] Open
Abstract
PCBP2 is a member of the poly(C)-binding protein (PCBP) family, which plays an important role in posttranscriptional and translational regulation by interacting with single-stranded poly(C) motifs in target mRNAs. Several PCBP family members have been reported to be involved in human malignancies. Here, we show that PCBP2 is upregulated in human glioma tissues and cell lines. Knockdown of PCBP2 inhibited glioma growth in vitro and in vivo through inhibition of cell-cycle progression and induction of caspase-3-mediated apoptosis. Thirty-five mRNAs were identified as putative PCBP2 targets/interactors using RIP-ChIP protein-RNA interaction arrays in a human glioma cell line, T98G. Four-and-a-half LIM domain 3 (FHL3) mRNA was downregulated in human gliomas and was identified as a PCBP2 target. Knockdown of PCBP2 enhanced the expression of FHL3 by stabilizing its mRNA. Overexpression of FHL3 attenuated cell growth and induced apoptosis. This study establishes a link between PCBP2 and FHL3 proteins and identifies a new pathway for regulating glioma progression.
Collapse
Affiliation(s)
- Wei Han
- State Key Laboratory of Medical Molecular Biology, Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ruggero D. Translational control in cancer etiology. Cold Spring Harb Perspect Biol 2013; 5:cshperspect.a012336. [PMID: 22767671 DOI: 10.1101/cshperspect.a012336] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The link between perturbations in translational control and cancer etiology is becoming a primary focus in cancer research. It has now been established that genetic alterations in several components of the translational apparatus underlie spontaneous cancers as well as an entire class of inherited syndromes known as "ribosomopathies" associated with increased cancer susceptibility. These discoveries have illuminated the importance of deregulations in translational control to very specific cellular processes that contribute to cancer etiology. In addition, a growing body of evidence supports the view that deregulation of translational control is a common mechanism by which diverse oncogenic pathways promote cellular transformation and tumor development. Indeed, activation of these key oncogenic pathways induces rapid and dramatic translational reprogramming both by increasing overall protein synthesis and by modulating specific mRNA networks. These translational changes promote cellular transformation, impacting almost every phase of tumor development. This paradigm represents a new frontier in the multihit model of cancer formation and offers significant promise for innovative cancer therapies. Current research, in conjunction with cutting edge technologies, will further enable us to explore novel mechanisms of translational control, functionally identify translationally controlled mRNA groups, and unravel their impact on cellular transformation and tumorigenesis.
Collapse
Affiliation(s)
- Davide Ruggero
- Helen Diller Cancer Center, School of Medicine, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
17
|
Shimizu Y, Mullins N, Blanchard Z, Elshamy WM. BRCA1/p220 loss triggers BRCA1-IRIS overexpression via mRNA stabilization in breast cancer cells. Oncotarget 2012; 3:299-313. [PMID: 22431556 PMCID: PMC3359886 DOI: 10.18632/oncotarget.462] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BRCA1/p220-assocaited and triple negative/basal-like (TN/BL) tumors are aggressive and incurable breast cancer diseases that share among other features the no/low BRCA1/p220 expression. Here we show that BRCA1/p220 silencing in normal human mammary epithelial (HME) cells reduces expression of two RNA-destabilizing proteins, namely AUF1 and pCBP2, both proteins bind and destabilize BRCA1-IRIS mRNA. BRCA1-IRIS overexpression in HME cells triggers expression of several TN/BL markers, e.g., cytokeratins 5 and 17, p-cadherin, EGFR and cyclin E as well as expression and activation of the pro-survival proteins; AKT and survivin. BRCA1-IRIS silencing in the TN/BL cell line, SUM149 or restoration of BRCA1/p220 expression in the mutant cell line, HCC1937 reduced expression of TN/BL markers, AKT and survivin and induced cell death. Collectively, we propose that BRCA1/p220 loss of expression or function triggers BRCA1-IRIS overexpression through a post-transcriptional mechanism, which in turn promotes formation of aggressive and invasive breast tumors by inducing expression of TN/BL and survival proteins.
Collapse
Affiliation(s)
- Yoshiko Shimizu
- Cancer Institute and Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | |
Collapse
|
18
|
Expression and mutation analysis of heterogeneous nuclear ribonucleoprotein G in human oral cancer. Oral Oncol 2011; 47:1011-6. [PMID: 21840245 DOI: 10.1016/j.oraloncology.2011.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/27/2011] [Accepted: 07/05/2011] [Indexed: 11/24/2022]
Abstract
We previously reported that wild type (wt) hnRNP G exhibited tumor suppressive activity in human oral squamous cell carcinoma (HOSCC) cell lines lacking hnRNP G. Wt hnRNP G markedly inhibited the proliferation capacity, anchorage independency and in vivo tumorigenicity of HOSCC cells and notably enhanced the DNA repair capabilities of these cells. In the present study, we studied the genetic and expression states of hnRNP G in normal, premalignant and malignant human oral tissues to further understand the relationship between the hnRNP G alterations and the development of human oral cancer. To correlate the cancer development and the level of hnRNP G expression, we performed an immunohistochemistry staining of hnRNP G in normal, premalignant and malignant human oral tissues. Moreover, we examined the entire coding regions of hnRNP G from selected samples to understand the cause of the alterations of the gene expression. The expression of hnRNP G was notably decreased or completely abolished in 80% of premalignant-dysplastic and malignant oral epithelial tissues, whereas 100% of normal and 90% of hyperplastic non-dysplastic epithelium showed high level of hnRNP G in the nucleus of the basal cell layers. Approximately 80% of HOSCC lacking the expression of hnRNP G showed genetic alteration in hnRNP G, i.e., point mutation and exonic deletion. This study suggest that genetic alterations and aberrant expression of hnRNP G occurring during oral carcinogenesis might be useful markers for the early detection of human oral cancer.
Collapse
|
19
|
Stumpf CR, Ruggero D. The cancerous translation apparatus. Curr Opin Genet Dev 2011; 21:474-83. [PMID: 21543223 PMCID: PMC3481834 DOI: 10.1016/j.gde.2011.03.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/25/2011] [Indexed: 01/04/2023]
Abstract
Deregulations in translational control are critical features of cancer initiation and progression. Activation of key oncogenic pathways promotes rapid and dramatic translational reprogramming, not simply by increasing overall protein synthesis, but also by modulating specific mRNA networks that promote cellular transformation. Additionally, ribosomopathies caused by mutations in ribosome components alter translational regulation leading to specific pathological features, including cancer susceptibility. Exciting advances in our understanding of translational control in cancer have illuminated a striking specificity innate to the translational apparatus. Characterizing this specificity will provide novel insights into how cells normally utilize translational control to modulate gene expression, how it is deregulated in cancer, and how these processes can be targeted to develop new cancer therapies.
Collapse
Affiliation(s)
- Craig R. Stumpf
- School of Medicine and Department of Urology, Helen Diller Family, Comprehensive Cancer Center, University of California, San Francisco, Helen, Diller Family Cancer Research Building Room 386, 1450 3rd Street, San Francisco, CA 94158-3110
| | - Davide Ruggero
- School of Medicine and Department of Urology, Helen Diller Family, Comprehensive Cancer Center, University of California, San Francisco, Helen, Diller Family Cancer Research Building Room 386, 1450 3rd Street, San Francisco, CA 94158-3110
| |
Collapse
|
20
|
In vitro and in vivo reduction of sodium arsenite induced toxicity by aqueous garlic extract. Food Chem Toxicol 2007; 46:740-51. [PMID: 17983699 DOI: 10.1016/j.fct.2007.09.108] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 08/30/2007] [Accepted: 09/25/2007] [Indexed: 01/12/2023]
Abstract
BACKGROUND Arsenic is ubiquitous in the environment, and chronic or acute exposure through food and water as well as occupational sources can contribute to a well-defined spectrum of disease. Despite arsenic being a health hazard and a well-documented human carcinogen, a safe, effective and specific preventive or therapeutic measure for treating arsenic induced toxicity still eludes us. OBJECTIVE This study was undertaken to evaluate the therapeutic efficacy of aqueous garlic (Allium sativum L.) extract (AGE) in terms of normalization of altered biochemical parameters particularly indicative of oxidative stress following sodium arsenite (NaAsO(2)) exposure and depletion of inorganic arsenic burden, in vitro and in vivo. RESULTS AGE (2mg/ml) co-administered with 10 microM NaAsO(2) attenuated arsenite induced cytotoxicity, reduced intracellular reactive oxygen species (ROS) level in human malignant melanoma cells (A375), human keratinocyte cells (HaCaT) and in cultured human normal dermal fibroblast cells. Moreover, AGE application in NaAsO(2) intoxicated Sprague-Dawley rats resulted in a marked inhibition of tissue lipid peroxide generation; enhanced level of total tissue sulfhydryl groups and glutathione; and also increased the activities of antioxidant enzymes, superoxide dismutase and catalase to near normal. An increase in blood ROS level and myeloperoxidase activity in arsenic-intoxicated rats was effectively prevented by AGE administration. AGE was also able to counter arsenic mediated incongruity in blood hematological variables and glucose level. CONCLUSIONS The restorative property of AGE was attributed to its antioxidant activity, chelating efficacy, and/or oxidizing capability of trivalent arsenic to its less toxic pentavalent form. Taken together, evidences indicate that AGE can be a potential protective regimen for arsenic mediated toxicity.
Collapse
|