1
|
Bassot A, Dragic H, Haddad SA, Moindrot L, Odouard S, Corlazzoli F, Marinari E, Bomane A, Brassens A, Marteyn A, Hibaoui Y, Petty TJ, Chalabi-Dchar M, Larrouquere L, Zdobnov EM, Legrand N, Tamburini J, Lincet H, Castets M, Yebra M, Migliorini D, Dutoit V, Walker PR, Preynat-Seauve O, Dietrich PY, Cosset É. Identification of a miRNA multi-targeting therapeutic strategy in glioblastoma. Cell Death Dis 2023; 14:630. [PMID: 37749143 PMCID: PMC10519979 DOI: 10.1038/s41419-023-06117-z] [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: 09/12/2022] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
Glioblastoma (GBM) is a deadly and the most common primary brain tumor in adults. Due to their regulation of a high number of mRNA transcripts, microRNAs (miRNAs) are key molecules in the control of biological processes and are thereby promising therapeutic targets for GBM patients. In this regard, we recently reported miRNAs as strong modulators of GBM aggressiveness. Here, using an integrative and comprehensive analysis of the TCGA database and the transcriptome of GBM biopsies, we identified three critical and clinically relevant miRNAs for GBM, miR-17-3p, miR-222, and miR-340. In addition, we showed that the combinatorial modulation of three of these miRNAs efficiently inhibited several biological processes in patient-derived GBM cells of all these three GBM subtypes (Mesenchymal, Proneural, Classical), induced cell death, and delayed tumor growth in a mouse tumor model. Finally, in a doxycycline-inducible model, we observed a significant inhibition of GBM stem cell viability and a significant delay of orthotopic tumor growth. Collectively, our results reveal, for the first time, the potential of miR-17-3p, miR-222 and miR-340 multi-targeting as a promising therapeutic strategy for GBM patients.
Collapse
Affiliation(s)
- Arthur Bassot
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Helena Dragic
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Sarah Al Haddad
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Laurine Moindrot
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Soline Odouard
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Francesca Corlazzoli
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Eliana Marinari
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Alexandra Bomane
- Department of CITI, Team Cell Death and Chilhood Cancers, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Augustin Brassens
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Antoine Marteyn
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Youssef Hibaoui
- Service de Gynécologie Obstétrique, HFR Fribourg - Hôpital Cantonal, Fribourg, Switzerland
| | - Tom J Petty
- Swiss Institute of Bioinformatics, Geneva, Switzerland
- SOPHiA GENETICS, Rolle, Switzerland
| | - Mounira Chalabi-Dchar
- Department of CITI, Team Ribosome, Translation & Cancer, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Louis Larrouquere
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Evgeny M Zdobnov
- Department of Genetic Medicine and Development, University of Geneva, and Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Noémie Legrand
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| | - Jérôme Tamburini
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Hubert Lincet
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Marie Castets
- Department of CITI, Team Cell Death and Chilhood Cancers, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France
| | - Mayra Yebra
- Department of Surgery, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - Denis Migliorini
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Valérie Dutoit
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Paul R Walker
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
- Laboratory of Immunobiology of Brain Tumors, Center for Translational Research in OncoHematology, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Olivier Preynat-Seauve
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Leman (SCCL), Agora Cancer Research Center, Geneva and Lausanne, Switzerland
| | - Érika Cosset
- Department of CITI, Team GLIMMER Of lIght, Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France.
- Laboratory of Tumor Immunology, Department of Oncology, Geneva University Hospitals, Geneva, Switzerland.
- Center for Translational Research in Onco-Hematology, Department of Oncology, University Hospital of Geneva, University of Geneva, Geneva, Switzerland.
- Team: GLIMMER Of lIght "GLIoblastoma MetabolisM, HetERogeneity, and OrganoIds"; Cancer Research Centre of Lyon - CRCL, INSERM U1052, CNRS UMR 5286, Lyon, France.
| |
Collapse
|
2
|
Rodríguez-Gómez G, Paredes-Villa A, Cervantes-Badillo MG, Gómez-Sonora JP, Jorge-Pérez JH, Cervantes-Roldán R, León-Del-Río A. Tristetraprolin: A cytosolic regulator of mRNA turnover moonlighting as transcriptional corepressor of gene expression. Mol Genet Metab 2021; 133:137-147. [PMID: 33795191 DOI: 10.1016/j.ymgme.2021.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/12/2023]
Abstract
Tristetraprolin (TTP) is a nucleocytoplasmic 326 amino acid protein whose sequence is characterized by possessing two CCCH-type zinc finger domains. In the cytoplasm TTP function is to promote the degradation of mRNAs that contain adenylate/uridylate-rich elements (AREs). Mechanistically, TTP promotes the recruitment of poly(A)-specific deadenylases and exoribonucleases. By reducing the half-life of about 10% of all the transcripts in the cell TTP has been shown to participate in multiple cell processes that include regulation of gene expression, cell proliferation, metabolic homeostasis and control of inflammation and immune responses. However, beyond its role in mRNA decay, in the cell nucleus TTP acts as a transcriptional coregulator by interacting with chromatin modifying enzymes. TTP has been shown to repress the transactivation of NF-κB and estrogen receptor suggesting the possibility that it participates in the transcriptional regulation of hundreds of genes in human cells and its possible involvement in breast cancer progression. In this review, we discuss the cytoplasmic and nuclear functions of TTP and the effect of the dysregulation of its protein levels in the development of human diseases. We suggest that TTP be classified as a moonlighting tumor supressor protein that regulates gene expression through two different mechanims; the decay of ARE-mRNAs and a transcriptional coregulatory function.
Collapse
Affiliation(s)
- Gabriel Rodríguez-Gómez
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Alejandro Paredes-Villa
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Mayte Guadalupe Cervantes-Badillo
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Jessica Paola Gómez-Sonora
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Jesús H Jorge-Pérez
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Rafael Cervantes-Roldán
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Alfonso León-Del-Río
- Programa de Investigación en Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| |
Collapse
|
3
|
Zhang D, Zhou Z, Yang R, Zhang S, Zhang B, Tan Y, Chen L, Li T, Tu J. Tristetraprolin, a Potential Safeguard Against Carcinoma: Role in the Tumor Microenvironment. Front Oncol 2021; 11:632189. [PMID: 34026612 PMCID: PMC8138596 DOI: 10.3389/fonc.2021.632189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Tristetraprolin (TTP), a well-known RNA-binding protein, primarily affects the expression of inflammation-related proteins by binding to the targeted AU-rich element in the 3' untranslated region after transcription and subsequently mediates messenger RNA decay. Recent studies have focused on the role of TTP in tumors and their related microenvironments, most of which have referred to TTP as a potential tumor suppressor involved in regulating cell proliferation, apoptosis, and metastasis of various cancers, as well as tumor immunity, inflammation, and metabolism of the microenvironment. Elevated TTP expression levels could aid the diagnosis and treatment of different cancers, improving the prognosis of patients. The aim of this review is to describe the role of TTP as a potential safeguard against carcinoma.
Collapse
Affiliation(s)
- Diwen Zhang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Zhigang Zhou
- The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ruixia Yang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Sujun Zhang
- Department of Experimental Animals, University of South China, Hengyang, China
| | - Bin Zhang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Yanxuan Tan
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Lingyao Chen
- Pharmacy School of Guilin Medical University, Guilin, China
| | - Tao Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agriculture Science, Shanghai, China
| | - Jian Tu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,Pharmacy School of Guilin Medical University, Guilin, China
| |
Collapse
|
4
|
Chemoprevention and therapeutic role of essential oils and phenolic compounds: Modeling tumor microenvironment in glioblastoma. Pharmacol Res 2021; 169:105638. [PMID: 33933637 DOI: 10.1016/j.phrs.2021.105638] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/28/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is the most common primary tumor of the central nervous system. Current treatments available for GBM entails surgical resection followed by temozolomide chemotherapy and/or radiotherapy, which are associated with multidrug resistance and severe side effects. While this treatment could yield good results, in almost all cases, patients suffer from relapse, which leads to reduced survival rates. Thus, therapeutic approaches with improved efficiency and reduced off-target risks are needed to overcome these problems. Regarding this, natural products appear as a safe and attractive strategy as chemotherapeutic agents or adjuvants in the treatment of GBM. Besides the increasing role of natural compounds for chemoprevention of GBM, it has been proposed to prevent carcinogenesis and metastasis of GBM. Numerous investigations showed that natural products are able to inhibit proliferation and angiogenesis, to induce apoptosis, and to target GBM stem cells, which are associated with tumor development and recurrence. This review gives a timely and comprehensive overview of the current literature regarding chemoprevention and therapy of GBM by natural products with a focus on essential oils and phenolic compounds and their molecular mechanisms.
Collapse
|
5
|
Sun X, Zhang H, Xie L, Qian C, Ye Y, Mao H, Wang B, Zhang H, Zhang Y, He X, Zhang S. Tristetraprolin destabilizes NOX2 mRNA and protects dopaminergic neurons from oxidative damage in Parkinson's disease. FASEB J 2020; 34:15047-15061. [PMID: 32954540 DOI: 10.1096/fj.201902967r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 08/04/2020] [Accepted: 08/31/2020] [Indexed: 01/11/2023]
Abstract
Tristetraprolin (TTP), an RNA-binding protein encoded by the ZFP36 gene, is vital for neural differentiation; however, its involvement in neurodegenerative diseases such as Parkinson's disease (PD) remains unclear. To explore the role of TTP in PD, an in vitro 1-methyl-4-phenylpyridinium (MPP+ ) cell model and an in vivo 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) of PD were used. Transfection of small interfering (si)-TTP RNA upregulated pro-oxidative NOX2 expression and ROS formation, downregulated anti-oxidative GSH and SOD activity;si-TTP upregulated pro-apoptotic cleaved-caspase-3 expression, and downregulated antiapoptotic Bcl-2 expression; while overexpression (OE)-TTP lentivirus caused opposite effects. Through database prediction, luciferase experiment, RNA immunoprecipitation (RIP), and mRNA stability analysis, we evaluated the potential binding sites of TTP to 3'-untranslated regions (3'-UTR) of NOX2 mRNA. TTP affected the NOX2 luciferase activity by binding to two sites in the NOX2 3'-UTR. RIP-qPCR confirmed TTP binding to both sites, with a higher affinity for site-2. In addition, TTP reduced the NOX2 mRNA stability. si-NOX2 and antioxidant N-acetyl cysteine (NAC) reversed si-TTP-induced cell apoptosis. In MPTP-treated mice, TTP expression increased and was co-located with dopaminergic neurons. TTP also inhibited NOX2 and decreased the oxidative stress in vivo. In conclusion, TTP protects against dopaminergic oxidative injury by promoting NOX2 mRNA degradation in the MPP+ /MPTP model of PD, suggesting that TTP could be a potential therapeutic target for regulating the oxidative stress in PD.
Collapse
Affiliation(s)
- Xiang Sun
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbo Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linghai Xie
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Qian
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yongyi Ye
- Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hengxu Mao
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Baoyan Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yizhou Zhang
- Tarbut V'Torah Community Day School, Irvine, CA, USA
| | - Xiaozheng He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shizhong Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
The Tristetraprolin Family of RNA-Binding Proteins in Cancer: Progress and Future Prospects. Cancers (Basel) 2020; 12:cancers12061539. [PMID: 32545247 PMCID: PMC7352335 DOI: 10.3390/cancers12061539] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Post-transcriptional regulation of gene expression plays a key role in cellular proliferation, differentiation, migration, and apoptosis. Increasing evidence suggests dysregulated post-transcriptional gene expression as an important mechanism in the pathogenesis of cancer. The tristetraprolin family of RNA-binding proteins (RBPs), which include Zinc Finger Protein 36 (ZFP36; commonly referred to as tristetraprolin (TTP)), Zinc Finger Protein 36 like 1 (ZFP36L1), and Zinc Finger Protein 36 like 2 (ZFP36L2), play key roles in the post-transcriptional regulation of gene expression. Mechanistically, these proteins function by binding to the AU-rich elements within the 3′-untranslated regions of their target mRNAs and, in turn, increasing mRNA turnover. The TTP family RBPs are emerging as key regulators of multiple biological processes relevant to cancer and are aberrantly expressed in numerous human cancers. The TTP family RBPs have tumor-suppressive properties and are also associated with cancer prognosis, metastasis, and resistance to chemotherapy. Herein, we summarize the various hallmark molecular traits of cancers that are reported to be regulated by the TTP family RBPs. We emphasize the role of the TTP family RBPs in the regulation of trait-associated mRNA targets in relevant cancer types/cell lines. Finally, we highlight the potential of the TTP family RBPs as prognostic indicators and discuss the possibility of targeting these TTP family RBPs for therapeutic benefits.
Collapse
|
7
|
Enrichment of Aldolase C Correlates with Low Non-Mutated IDH1 Expression and Predicts a Favorable Prognosis in Glioblastomas. Cancers (Basel) 2019; 11:cancers11091238. [PMID: 31450822 PMCID: PMC6770576 DOI: 10.3390/cancers11091238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022] Open
Abstract
The aldolases family is one of the main enzymes involved in the process of glycolysis. Aldolase C (ALDOC), which belongs to the aldolase family, is found in normal brain tissue and is responsible for the repair of injured tissue. However, the role of ALDOC in glioblastoma remains unclear. In this study, we data-mined in silico databases to evaluate aldolase family members’ mRNA expression in glioblastoma patient cohorts for determining its prognostic values. After that, we also performed immunohistochemical stain (IHC) analysis to evaluate protein expression levels of ALDOC in glioblastoma tissues. From The Cancer Genome Atlas (TCGA) database analyses, higher mRNA expression levels in normal brain tissue compared to glioblastoma was observed. In addition, compared to low-grade glioma, ALDOC expression was significantly downregulated in high-grade glioblastoma. Besides, the expression level of ALDOC was associated with molecular subtypes of glioblastomas and recurrent status in several data sets. In contrast, aldolase A (ALDOA) and aldolase B (ALDOB) revealed no significant prognostic impacts in the glioblastoma cohorts. Furthermore, we also proved that ALDOC mRNA and protein expression inversely correlated with non-mutated IDH1 expressions in glioblastoma patient cohorts. Additionally, the concordance of low ALDOC and high non-mutated IDH1 expressions predicted a stronger poor prognosis in glioblastoma patients compared to each of above tests presented alone. The plausible ALDOC and IDH1 regulatory mechanism was further elucidated. Our results support high ALDOC expression in glioblastomas that might imply the mutated status of IDH1, less possibility of mesenchymal subtype, and predict a favorable prognosis.
Collapse
|
8
|
Lin JC, Tsai JT, Chao TY, Ma HI, Liu WH. Musashi-1 Enhances Glioblastoma Migration by Promoting ICAM1 Translation. Neoplasia 2019; 21:459-468. [PMID: 30959276 PMCID: PMC6453839 DOI: 10.1016/j.neo.2019.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a lethal brain tumor with a mean survival time of 1 year. One major reason for therapeutic failure is that GBM cells have an extraordinary capacity to invade normal brain tissue beyond the surgical margin, accounting for the lack of treatment efficacy. GBM cells that can infiltrate into the healthy brain possess tumor properties of stemness and invasion, and previous studies demonstrate that Musashi-1 (MSI1), a neural stem cell marker, plays an important role in the maintenance of stem cell status, cellular differentiation, and tumorigenesis in cancers. By analyzing neuronal progenitor cell markers and stemness genes, we predicted that MSI1 might be an important factor in GBM pathogenesis. Because inflammation aids in the proliferation and survival of malignant cells, the inflammatory microenvironment also promotes GBM invasion, and intercellular adhesion molecule-1 (ICAM1), a member of the immunoglobulin superfamily, is involved in inflammation. Our results indicate that the above phenomena are likely due to MSI1 upregulation, which occurred simultaneously with higher expression of ICAM1 in GBM cells. Indeed, MSI1 knockdown effectively suppressed ICAM1 expression and blocked GBM cell motility and invasion, whereas overexpressing ICAM1 reversed these effects. According to RNA immunoprecipitation assays, MSI1-mediated mRNA interactions promote ICAM1 translation. Finally, immunohistochemical analysis showed MSI1 and ICAM-1 to be coexpressed at high levels in GBM tissues. Thus, the MSI1/ICAM1 pathway plays an important role in oncogenic resistance, including increased tumor invasion, and MSI1/ICAM1 may be a target for GBM treatment.
Collapse
Affiliation(s)
- Jang-Chun Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jo-Ting Tsai
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsu-Yi Chao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Division of Hematology/Oncology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, No.325, Sec. 2, Cheng-Kung Road, Taipei 11490, Taiwan; Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Hsiu Liu
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, No.325, Sec. 2, Cheng-Kung Road, Taipei 11490, Taiwan; Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
9
|
Roles of Tristetraprolin in Tumorigenesis. Int J Mol Sci 2018; 19:ijms19113384. [PMID: 30380668 PMCID: PMC6274954 DOI: 10.3390/ijms19113384] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 10/25/2018] [Accepted: 10/26/2018] [Indexed: 12/13/2022] Open
Abstract
Genetic loss or mutations in tumor suppressor genes promote tumorigenesis. The prospective tumor suppressor tristetraprolin (TTP) has been shown to negatively regulate tumorigenesis through destabilizing the messenger RNAs of critical genes implicated in both tumor onset and tumor progression. Regulation of TTP has therefore emerged as an important issue in tumorigenesis. Similar to other tumor suppressors, TTP expression is frequently downregualted in various human cancers, and its low expression is correlated with poor prognosis. Additionally, disruption in the regulation of TTP by various mechanisms results in the inactivation of TTP protein or altered TTP expression. A recent study showing alleviation of Myc-driven lymphomagenesis by the forced expression of TTP has shed light on new therapeutic avenues for cancer prevention and treatment through the restoration of TTP expression. In this review, we summarize key oncogenes subjected to the TTP-mediated mRNA degradation, and discuss how dysregulation of TTP can contribute to tumorigenesis. In addition, the control mechanism underlying TTP expression at the posttranscriptional and posttranslational levels will be discussed.
Collapse
|
10
|
Ren C, Yang T, Qiao P, Wang L, Han X, Lv S, Sun Y, Liu Z, Du Y, Yu Z. PIM2 interacts with tristetraprolin and promotes breast cancer tumorigenesis. Mol Oncol 2018; 12:690-704. [PMID: 29570932 PMCID: PMC5928357 DOI: 10.1002/1878-0261.12192] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 01/03/2023] Open
Abstract
Tristetraprolin (TTP) is an AU‐rich element‐binding protein that regulates mRNA stability and plays important roles in cancer. The mechanisms by which TTP is regulated in breast cancer are poorly understood. Using multiple biochemical approaches, we found that proviral insertion in murine lymphomas 2 (PIM2) is a novel binding partner of TTP. Interestingly, PIM2 decreased TTP protein levels independent of its kinase activity. PIM2 instead targeted TTP protein for degradation via the ubiquitin‐proteasome pathway. Furthermore, immunohistochemical staining showed that PIM2 and TTP protein levels were negatively correlated in human breast cancer samples. Indeed, PIM2 overexpression de‐repressed TTP‐mediated inhibition of breast cancer cell proliferation and migration in vitro and promoted breast tumor xenograft growth in vivo. These findings demonstrate an important role for the PIM2‐TTP complex in breast cancer tumorigenesis, suggesting that PIM2 may represent a potential therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Chune Ren
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Tingting Yang
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Pengyun Qiao
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Li Wang
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Xue Han
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Shijun Lv
- Department of PathologyAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Yonghong Sun
- Department of PathologyAffiliated Hospital of Weifang Medical UniversityShandongChina
| | - Zhijun Liu
- Department of Medical BiologyWeifang Medical UniversityShandongChina
| | - Yu Du
- Department of Medical BiologyWeifang Medical UniversityShandongChina
| | - Zhenhai Yu
- Department of Reproductive MedicineAffiliated Hospital of Weifang Medical UniversityShandongChina
| |
Collapse
|
11
|
Chen HY, Lin LT, Wang ML, Tsai KL, Huang PI, Yang YP, Lee YY, Chen YW, Lo WL, Lan YT, Chiou SH, Lin CM, Ma HI, Chen MT. Musashi-1 promotes chemoresistant granule formation by PKR/eIF2α signalling cascade in refractory glioblastoma. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1850-1861. [PMID: 29486283 DOI: 10.1016/j.bbadis.2018.02.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/25/2018] [Accepted: 02/21/2018] [Indexed: 01/08/2023]
Abstract
Musashi-1 (MSI1), one of the RNA-binding proteins, is abundantly found not only in neural stem cells but also in several cancer tissues and has been reported to act as a positive regulator of cancer progression. Growing evidence indicates that PKR and eIF2α play pivotal roles in the stimulation of stress granule formation as well as in the subsequent translation modulation in response to stressful conditions; however, little is known about whether MSI1 is involved in this PKR/eIF2α cancer stem cell-enhancing machinery. In this study, we demonstrated that MSI1 promotes human glioblastoma multiforme (GBM) stem cells and enhances chemoresistance when exposed to sublethal stress. The overexpression of MSI1 leads to a protective effect in mitigating drug-induced cell death, thus facilitating the formation of chemoresistant stress granules (SGs) in response to arsenic trioxide (ATO) treatment. SG components, such as PKR and eIF2α, were dominantly activated and assembled, while ATO was engaged. The activated PKR and eIF2α contribute to the downstream enhancement of stem cell genes, thereby promoting the progression of GBM. The silencing of MSI1 or PKR both obviously withdrew the phenomena. Taken together, our findings indicate that MSI1 plays a leading role in stress granule formation that grants cancer stem cell properties and chemoresistant stress granules in GBM, in response to stressful conditions via the PKR/eIF2α signalling cascade.
Collapse
Affiliation(s)
- Hsiao-Yun Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Liang-Ting Lin
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Mong-Lien Wang
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pin-I Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ping Yang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Yen Lee
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Wei Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Cancer Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Liang Lo
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Oral and Maxillofacial Surgery, Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuan-Tzu Lan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chien-Min Lin
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Ming-Teh Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
12
|
Montorsi L, Guizzetti F, Alecci C, Caporali A, Martello A, Atene CG, Parenti S, Pizzini S, Zanovello P, Bortoluzzi S, Ferrari S, Grande A, Zanocco-Marani T. Loss of ZFP36 expression in colorectal cancer correlates to wnt/ ß-catenin activity and enhances epithelial-to-mesenchymal transition through upregulation of ZEB1, SOX9 and MACC1. Oncotarget 2018; 7:59144-59157. [PMID: 27463018 PMCID: PMC5312301 DOI: 10.18632/oncotarget.10828] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/09/2016] [Indexed: 01/20/2023] Open
Abstract
The mRNA-destabilizing protein ZFP36 has been previously described as a tumor suppressor whose expression is lost during colorectal cancer development. In order to evaluate its role in this disease, we restored ZFP36 expression in different cell contexts, showing that the presence of this protein impairs the epithelial-to-mesenchymal transition (EMT) and induces a higher susceptibility to anoikis. Consistently, we found that ZFP36 inhibits the expression of three key transcription factors involved in EMT: ZEB1, MACC1 and SOX9. Finally, we observed for the first time that its expression negatively correlates with the activity of Wnt/β-catenin pathway, which is constitutively activated in colorectal cancer. This evidence provides a clue on the mechanism leading to the loss of ZFP36 in CRC.
Collapse
Affiliation(s)
- Lucia Montorsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Filippo Guizzetti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudia Alecci
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Caporali
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Andrea Martello
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | | | - Sandra Parenti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Pizzini
- Centre for Integrative Biology (CIBIO), University of Trento, Povo (Trento), Italy
| | - Paola Zanovello
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | | | - Sergio Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alexis Grande
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | |
Collapse
|
13
|
Santio NM, Koskinen PJ. PIM kinases: From survival factors to regulators of cell motility. Int J Biochem Cell Biol 2017; 93:74-85. [DOI: 10.1016/j.biocel.2017.10.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 01/01/2023]
|
14
|
Liu F, Zhang S, Zhao Z, Mao X, Huang J, Wu Z, Zheng L, Wang Q. MicroRNA-27b up-regulated by human papillomavirus 16 E7 promotes proliferation and suppresses apoptosis by targeting polo-like kinase2 in cervical cancer. Oncotarget 2017; 7:19666-79. [PMID: 26910911 PMCID: PMC4991410 DOI: 10.18632/oncotarget.7531] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/30/2015] [Indexed: 12/13/2022] Open
Abstract
The infection with high-risk human papillomavirus is linked to cervical cancer, nevertheless, the role of miRNAs regulated by HPV oncogenes in cancer progression remain largely unknown. Here, we knocked down endogenous E6/E7 in HPV16-positive CaSki cell lines, screened differences in miRNA expression profile with control using miRNA array. 38 miRNAs were down-regulated and 6 miRNAs were up-regulated in the E6/E7 silenced CaSki cells (>2-fold changes with P <0.05). The levels of miR-27b, miR-20a, miR-24, miR-93, and miR-106b were verified by qPCR in E6/E7 silenced CaSki and SiHa cells. MiR-27b, up-regulated by E7, promoted CaSki and SiHa cell proliferation and invasion, inhibit paclitaxel-induced apoptosis. Dual-luciferase experiment confirmed miR-27b down-regulated its target gene PLK2 through the “seed regions”. The tumor suppressor PLK2 inhibited SiHa cell proliferation, reduced cell viability, and promoted paclitaxel/cisplatin -induced apoptosis. Furthermore, DGCR8 was found to mediate the up-regulation of miR-27b by HPV16 E7. Our study demonstrated that HPV16 E7 could increase DGCR8 to promote the generation of miR-27b, which accelerated cell proliferation and inhibited paclitaxel-induced cell apoptosis through down-regulating PLK2. These findings provide an insight into the interaction network of viral oncogene, miR-27b and PLK2, and support the potential strategies using antisense nucleic acid of miR-27b for therapy of cervical cancer in the future.
Collapse
Affiliation(s)
- Fei Liu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Shimeng Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Central Laboratory, Shenzhen Shekou People's Hospital, Shenzhen 518000, China
| | - Zhen Zhao
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD 20892, USA
| | - Xinru Mao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jinlan Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zixian Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Wei ZR, Liang C, Feng D, Cheng YJ, Wang WM, Yang DJ, Wang YX, Cai QP. Low tristetraprolin expression promotes cell proliferation and predicts poor patients outcome in pancreatic cancer. Oncotarget 2017; 7:17737-50. [PMID: 26894969 PMCID: PMC4951246 DOI: 10.18632/oncotarget.7397] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
Tristetraprolin (also known as TTP, TIS11, ZFP36, and Nup475) is a well-characterized tumor suppressor that is down-regulated in several tumor types. In the current study, we found that TTP expression was markedly reduced in pancreatic cancer samples as compared to matched normal tissues. Low TTP level was associated with age (P=0.037), tumor size (P=0.008), tumor differentiation (P=0.004), postoperative T stage (pT stage, P<0.001), postoperative N stage (pN stage, P=0.008) and TNM stage (P<0.001). Moreover, low TTP expression predicted reduced survival rates and poor patient outcome. We also found that TTP impairs pancreatic cancer cell proliferation both in vivo and in vitro. Fluorescence Activated Cell Sorting (FACS) assay showed that TTP over-expression both increases apoptosis and decreases proliferation in pancreatic cancer cells. RNA-sequencing analysis showed that TTP over-expression downregulates several tumor-related factors, including Pim-1 and IL-6. Our findings indicate that TTP could serve as a potential prognostic indicator in pancreatic cancer.
Collapse
Affiliation(s)
- Zi-Ran Wei
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Chao Liang
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Dan Feng
- Department of Oncology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ya-Jun Cheng
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei-Min Wang
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - De-Jun Yang
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yue-Xiang Wang
- SIBS (Institute of Health Sciences)-Changzheng Hospital Joint Center for Translational Medicine, Institute of Health Sciences, Shanghai Changzheng Hospital, Institutes for Translational Medicine (CAS-SMMU), Key Laboratory of Stem Cell Biology, Institute of Health Sciences, SIBS, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Collaborative Innovation Center of Systems Biomedicine, Shanghai, China
| | - Qing-Ping Cai
- Department of Gastro-Intestine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
16
|
Huang L, Yu Z, Zhang Z, Ma W, Song S, Huang G. Interaction with Pyruvate Kinase M2 Destabilizes Tristetraprolin by Proteasome Degradation and Regulates Cell Proliferation in Breast Cancer. Sci Rep 2016; 6:22449. [PMID: 26926077 PMCID: PMC4772106 DOI: 10.1038/srep22449] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 02/17/2016] [Indexed: 12/29/2022] Open
Abstract
Pyruvate kinase M2 (PKM2), which is predominantly expressed in most cancers, plays a key role in the Warburg effect. However, how PKM2 functions as a tumor supportive protein has not been fully elucidated. Here, we identified tristetraprolin (TTP), an AU-rich, element-binding protein that regulates mRNA stability, as a new binding partner of PKM2. Our data reveal that PKM2 suppresses TTP protein levels by promoting its phosphorylation, ubiquitination, and proteasome degradation, reducing its mRNA turnover ability and ultimately impairing cell viability in breast cancer cells. The p38/mitogen-activated protein kinase (MAPK) pathway might be involved in PKM2-mediated TTP degradation, while treatment with the p38 inhibitor or siRNA abolished PKM2-induced TTP protein degradation. These findings demonstrate that PKM2-TTP association is crucial for regulating breast cancer cell proliferation and is therefore a potential therapeutic target in cancer.
Collapse
Affiliation(s)
- Liangqian Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhenhai Yu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhenchao Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Wenjing Ma
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shaoli Song
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
17
|
Newman R, McHugh J, Turner M. RNA binding proteins as regulators of immune cell biology. Clin Exp Immunol 2015. [PMID: 26201441 DOI: 10.1111/cei.12684] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sequence-specific RNA binding proteins (RBP) are important regulators of the immune response. RBP modulate gene expression by regulating splicing, polyadenylation, localization, translation and decay of target mRNAs. Increasing evidence suggests that RBP play critical roles in the development, activation and function of lymphocyte populations in the immune system. This review will discuss the post-transcriptional regulation of gene expression by RBP during lymphocyte development, with particular focus on the Tristetraprolin family of RBP.
Collapse
Affiliation(s)
- R Newman
- Babraham Institute, Cambridge, UK
| | - J McHugh
- Babraham Institute, Cambridge, UK
| | - M Turner
- Babraham Institute, Cambridge, UK
| |
Collapse
|
18
|
Selmi T, Alecci C, dell' Aquila M, Montorsi L, Martello A, Guizzetti F, Volpi N, Parenti S, Ferrari S, Salomoni P, Grande A, Zanocco-Marani T. ZFP36 stabilizes RIP1 via degradation of XIAP and cIAP2 thereby promoting ripoptosome assembly. BMC Cancer 2015; 15:357. [PMID: 25939870 PMCID: PMC4424499 DOI: 10.1186/s12885-015-1388-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 04/28/2015] [Indexed: 11/10/2022] Open
Abstract
Background ZFP36 is an mRNA binding protein that exerts anti-tumor activity in glioblastoma by triggering cell death, associated to an increase in the stability of the kinase RIP1. Methods We used cell death assays, size exclusion chromatography, Co-Immunoprecipitation, shRNA lentivectors and glioma neural stem cells to determine the effects of ZFP36 on the assembly of a death complex containing RIP1 and on the induction of necroptosis. Results Here we demonstrate that ZFP36 promotes the assembly of the death complex called Ripoptosome and induces RIP1-dependent death. This involves the depletion of the ubiquitine ligases cIAP2 and XIAP and leads to the association of RIP1 to caspase-8 and FADD. Moreover, we show that ZFP36 controls RIP1 levels in glioma neural stem cell lines. Conclusions We provide a molecular mechanism for the tumor suppressor role of ZFP36, and the first evidence for Ripoptosome assembly following ZFP36 expression. These findings suggest that ZFP36 plays an important role in RIP1-dependent cell death in conditions where IAPs are depleted. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1388-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tommaso Selmi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Claudia Alecci
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Miriam dell' Aquila
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Lucia Montorsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Andrea Martello
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Filippo Guizzetti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Nicola Volpi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Sandra Parenti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Sergio Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Paolo Salomoni
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, London WC1E 6BT, United Kingdom.
| | - Alexis Grande
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| | - Tommaso Zanocco-Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, 41125, Italy.
| |
Collapse
|
19
|
Ryu J, Yoon NA, Lee YK, Jeong JY, Kang S, Seong H, Choi J, Park N, Kim N, Cho WJ, Paek SH, Cho GJ, Choi WS, Park JY, Park JW, Kang SS. Tristetraprolin inhibits the growth of human glioma cells through downregulation of urokinase plasminogen activator/urokinase plasminogen activator receptor mRNAs. Mol Cells 2014; 38:156-62. [PMID: 25556371 PMCID: PMC4332028 DOI: 10.14348/molcells.2015.2259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 01/02/2023] Open
Abstract
Urokinase plasminogen activator (uPA) and urokinase plasminogen activator receptor (uPAR) play a major role in the infiltrative growth of glioblastoma. Downregulatoion of the uPA and uPAR has been reported to inhibit the growth glioblastoma. Here, we demonstrate that tristetraprolin (TTP) inhibits the growth of U87MG human glioma cells through downregulation of uPA and uPAR. Our results show that expression level of TTP is inversely correlated with those of uPA and uPAR in human glioma cells and tissues. TTP binds to the AU-rich elements within the 3' untranslated regions of uPA and uPAR and overexpression of TTP decreased the expression of uPA and uPAR through enhancing the degradation of their mRNAs. In addition, overexpression of TTP inhibited the growth and invasion of U87MG cells. Our findings implicate that TTP can be used as a promising therapeutic target to treat human glioma.
Collapse
Affiliation(s)
- Jinhyun Ryu
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Nal Ae Yoon
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Yeon Kyung Lee
- Department of Molecular Medicine, Gachon University of Medicine and Science, Incheon 406-840, Korea
| | - Joo Yeon Jeong
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Seokmin Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Hyemin Seong
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Jungil Choi
- Gyeongnam Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology (KIT), Jinju 660-844, Korea
| | - Nammi Park
- Department of Physiology, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Nayoung Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 138-736, Korea
| | - Wha Ja Cho
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 682-060, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Gyeong Jae Cho
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Wan Sung Choi
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 136-703, Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 660-751, Korea
| |
Collapse
|
20
|
Xu J, Zhang T, Wang T, You L, Zhao Y. PIM kinases: an overview in tumors and recent advances in pancreatic cancer. Future Oncol 2014; 10:865-76. [PMID: 24799066 DOI: 10.2217/fon.13.229] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The PIM kinases represent a family of serine/threonine kinases, which is composed of three different members (PIM1, PIM2 and PIM3). Aberrant expression of PIM kinases is observed in variety of tumors, including pancreatic cancer. The PIM kinases play pivotal roles in the regulation of cell cycle, apoptosis, properties of stem cells, metabolism, autophagy, drug resistance and targeted therapy. The roles of PIM kinases in pancreatic cancer include the regulation of proliferation, apoptosis, cell cycle, formation, angiogenesis and prediction prognosis. Blocking the activities of PIM kinases could prevent pancreatic cancer development. PIM kinases may be a novel target for cancer therapy.
Collapse
Affiliation(s)
- Jianwei Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | | | | | | | | |
Collapse
|
21
|
Feldman ME, Yarden Y. Steering tumor progression through the transcriptional response to growth factors and stroma. FEBS Lett 2014; 588:2407-14. [PMID: 24873881 DOI: 10.1016/j.febslet.2014.05.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 02/04/2023]
Abstract
Tumor progression can be understood as a collaborative effort of mutations and growth factors, which propels cell proliferation and matrix invasion, and also enables evasion of drug-induced apoptosis. Concentrating on EGFR, we discuss downstream signaling and the initiation of transcriptional events in response to growth factors. Specifically, we portray a wave-like program, which initiates by rapid disappearance of two-dozen microRNAs, followed by an abrupt rise of immediate early genes (IEGs), relatively short transcripts encoding transcriptional regulators. Concurrent with the fall of IEGs, some 30-60 min after stimulation, a larger group, the delayed early genes, is up-regulated and its own fall overlaps the rise of the final wave of late response genes. This late wave persists and determines long-term phenotype acquisition, such as invasiveness. Key regulatory steps in the orderly response to growth factors provide a trove of potential oncogenes and tumor suppressors.
Collapse
Affiliation(s)
- Morris E Feldman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
22
|
Sayegh ET, Kaur G, Bloch O, Parsa AT. Systematic review of protein biomarkers of invasive behavior in glioblastoma. Mol Neurobiol 2013; 49:1212-44. [PMID: 24271659 DOI: 10.1007/s12035-013-8593-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 11/11/2013] [Indexed: 12/26/2022]
Abstract
Glioblastoma (GBM) is an aggressive and incurable brain tumor with a grave prognosis. Recurrence is inevitable even with maximal surgical resection, in large part because GBM is a highly invasive tumor. Invasiveness also contributes to the failure of multiple cornerstones of GBM therapy, including radiotherapy, temozolomide chemotherapy, and vascular endothelial growth factor blockade. In recent years there has been significant progress in the identification of protein biomarkers of invasive phenotype in GBM. In this article, we comprehensively review the literature and survey a broad spectrum of biomarkers, including proteolytic enzymes, extracellular matrix proteins, cell adhesion molecules, neurodevelopmental factors, cell signaling and transcription factors, angiogenic effectors, metabolic proteins, membrane channels, and cytokines and chemokines. In light of the marked variation seen in outcomes in GBM patients, the systematic use of these biomarkers could be used to form a framework for better prediction, prognostication, and treatment selection, as well as the identification of molecular targets for further laboratory investigation and development of nascent, directed therapies.
Collapse
Affiliation(s)
- Eli T Sayegh
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, 676 N. St. Clair Street, Suite 2210, Chicago, IL, 60611-2911, USA
| | | | | | | |
Collapse
|
23
|
Abstract
Cytoplasmic polyadenylation element-binding protein 1 (CPEB1) is an mRNA-binding protein present in both neurons and glia. CPEB1 is capable of both repressing mRNA translation and activating it depending upon its phosphorylation state. CPEB1-bound mRNAs are held in translational dormancy until CPEB1 is phosphorylated, leading to the cytoplasmic polyadenylation of the bound mRNA that triggers translation. Here, we show that CPEB1 can bind to and regulate translation of the mRNA-encoding metadherin (MTDH, also known as AEG-1 and Lyric) in the rat glioblastoma cell line CNS1. MTDH/AEG-1 is being revealed as a critical signaling molecule in tumor progression, playing roles in invasion, metastasis, and chemoresistance. By using a mutant of CPEB1 that cannot be phosphorylated (thereby holding target mRNAs in translational arrest), we show that inhibiting CPEB1-mediated translation blocks MTDH/AEG-1 expression in vitro and inhibits glioblastomas tumor growth in vivo. CPEB1-mediated translation is likely to impact several signaling pathways that may promote tumor progression, but we present evidence suggesting a role in directed cell migration in glioblastoma cells. In addition, reporter mRNA containing CPEB1-binding sites is transported to the leading edge of migrating cells and translated, whereas the same mRNA with point mutations in the binding sites is synthesized perinuclearly. Our findings show that CPEB1 is hyperactive in rat glioblastoma cells and is regulating an important cohort of mRNAs whose increased translation is fueling the progression of tumor proliferation and dispersal in the brain. Thus, targeting CPEB1-mediated mRNA translation might be a sound therapeutic approach.
Collapse
Affiliation(s)
- Dawn M Kochanek
- Department of Molecular, Cellular & Developmental Biology, 260 Whitney Ave, KBT 338, Box 208103, New Haven, CT 06520, USA
| | | |
Collapse
|
24
|
Krivtsov AV, Figueroa ME, Sinha AU, Stubbs MC, Feng Z, Valk PJM, Delwel R, Döhner K, Bullinger L, Kung AL, Melnick AM, Armstrong SA. Cell of origin determines clinically relevant subtypes of MLL-rearranged AML. Leukemia 2012; 27:852-60. [PMID: 23235717 DOI: 10.1038/leu.2012.363] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mixed lineage leukemia (MLL)-fusion proteins can induce acute myeloid leukemias (AMLs) from either hematopoietic stem cells (HSCs) or granulocyte-macrophage progenitors (GMPs), but it remains unclear whether the cell of origin influences the biology of the resultant leukemia. MLL-AF9-transduced single HSCs or GMPs could be continuously replated, but HSC-derived clones were more likely than GMP-derived clones to initiate AML in mice. Leukemia stem cells derived from either HSCs or GMPs had a similar immunophenotype consistent with a maturing myeloid cell (LGMP). Gene expression analyses demonstrated that LGMP inherited gene expression programs from the cell of origin including high-level Evi-1 expression in HSC-derived LGMP. The gene expression signature of LGMP derived from HSCs was enriched in poor prognosis human MLL-rearranged AML in three independent data sets. Moreover, global 5'-mC levels were elevated in HSC-derived leukemias as compared with GMP-derived leukemias. This mirrored a difference seen in 5'-mC between MLL-rearranged human leukemias that are either EVI1 positive or EVI1 negative. Finally, HSC-derived leukemias were more resistant to chemotherapy than GMP-derived leukemias. These data demonstrate that the cell of origin influences the gene expression profile, the epigenetic state and the drug response in AML, and that these differences can account for clinical heterogeneity within a molecularly defined group of leukemias.
Collapse
Affiliation(s)
- A V Krivtsov
- Division of Hematology/Oncology, Children's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|