1
|
Pan J, Li Y, Gao W, Jiang Q, Geng L, Ding J, Li S, Li J. Transcription factor Sp1 transcriptionally enhances GSDME expression for pyroptosis. Cell Death Dis 2024; 15:66. [PMID: 38238307 PMCID: PMC10796635 DOI: 10.1038/s41419-024-06455-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Gasdermin-E (GSDME), the executioner of pyroptosis when cleaved by caspase 3, plays a crucial role in tumor defense and the response to chemotherapy drugs in cells. So far, there are poorly known mechanisms for the expression regulation of GSDME during cell death. Here, we identify the transcription factor Sp1 (Specificity protein 1) as a positive regulator of GSDME-mediated pyroptosis. Sp1 directly interacts with the GSDME promoter at -36 ~ -28 site and promotes GSDME gene transcription. Further, Sp1 knockdown or inhibition suppresses GSDME expression, thus reducing chemotherapy drugs (topotecan, etoposide, doxorubicin, sorafinib and cisplatin) induced cell pyroptosis. The regulation process synergizes with STAT3 (Signal transducer and activator of transcription 3) activity and antagonizes with DNA methylation but barely affects GSDMD-mediated pyroptosis or TNF-induced necroptosis. Our current finding reveals a new regulating mechanism of GSDME expression, which may be a viable target for the intervention of GSDME-dependent inflammatory diseases and cancer therapy.
Collapse
Affiliation(s)
- Jiasong Pan
- Department of Neurology, Huashan Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Yuanyuan Li
- Department of Neurology, Huashan Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Wenqing Gao
- Department of Neurology, Huashan Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Qizhou Jiang
- Division of Natural Science, Duke Kunshan University, Jiangsu, China
| | - Lu Geng
- Department of Neurology, Huashan Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Suhua Li
- Division of Natural Science, Duke Kunshan University, Jiangsu, China.
| | - Jixi Li
- Department of Neurology, Huashan Hospital, State Key Laboratory of Genetic Engineering and School of Life Sciences, Fudan University, Shanghai, China.
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China.
| |
Collapse
|
2
|
Zhou Q, Li L, Sha F, Lei Y, Tian X, Chen L, Chen Y, Liu H, Guo Y. PTTG1 Reprograms Asparagine Metabolism to Promote Hepatocellular Carcinoma Progression. Cancer Res 2023; 83:2372-2386. [PMID: 37159932 DOI: 10.1158/0008-5472.can-22-3561] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/14/2023] [Accepted: 05/04/2023] [Indexed: 05/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and has a poor prognosis. Pituitary tumor transforming gene 1 (PTTG1) is highly expressed in HCC, suggesting it could play an important role in hepatocellular carcinogenesis. Here, we evaluated the impact of PTTG1 deficiency on HCC development using a diethylnitrosamine (DEN)-induced HCC mouse model and a hepatitis B virus (HBV) regulatory X protein (HBx)-induced spontaneous HCC mouse model. PTTG1 deficiency significantly suppressed DEN- and HBx-induced hepatocellular carcinogenesis. Mechanistically, PTTG1 promoted asparagine synthetase (ASNS) transcription by binding to its promoter, and asparagine (Asn) levels were correspondingly increased. The elevated levels of Asn subsequently activated the mTOR pathway to facilitate HCC progression. In addition, asparaginase treatment reversed the proliferation induced by PTTG1 overexpression. Furthermore, HBx promoted ASNS and Asn metabolism by upregulating PTTG1 expression. Overall, PTTG1 is involved in the reprogramming of Asn metabolism to promote HCC progression and may serve as a therapeutic and diagnostic target for HCC. SIGNIFICANCE PTTG1 is upregulated in hepatocellular carcinoma and increases asparagine production to stimulate mTOR activity and promote tumor progression.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Leijia Li
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Feifei Sha
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Yiming Lei
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Xuan Tian
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Lingjun Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Yan Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Huiling Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| | - Yunwei Guo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Li JD, Farah AA, Huang ZG, Zhai GQ, Wang RG, Liu JL, Wang QJ, Zhang GL, Lei ZL, Dang YW, Li SH. Clinical significance and potential regulatory mechanism of overexpression of pituitary tumor-transforming gene transcription factor in bladder cancer. BMC Cancer 2022; 22:713. [PMID: 35768832 PMCID: PMC9241226 DOI: 10.1186/s12885-022-09810-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/21/2022] [Indexed: 11/30/2022] Open
Abstract
Background Pituitary tumor transforming gene-1 (PTTG1) transcription factor is identified as carcinogenic and associated with tumor invasiveness, but its role in bladder cancer (BLCA) remains obscure. This research is intended to analyze the aberrant expression and clinical significance of PTTG1 in BLCA, explore the relationship between PTTG1 and tumor microenvironment characteristics and predict its potential transcriptional activity in BLCA tissue. Methods We compared the expression discrepancy of PTTG1 mRNA in BLCA and normal bladder tissue, using the BLCA transcriptomic datasets from GEO, ArrayExpress, TCGA, and GTEx. In-house immunohistochemical staining was implemented to determine the PTTG1 protein intensity. The prognostic value of PTTG1 was evaluated using the Kaplan-Meier Plotter. CRISPR screen data was utilized to estimate the effect PTTG1 interference has on BLCA cell lines. We predicted the abundance of the immune cells in the BLCA tumor microenvironment using the microenvironment cell populations-counter and ESTIMATE algorithms. Single-cell RNA sequencing data was applied to identify the major cell types in BLCA, and the dynamics of BLCA progression were revealed using pseudotime analysis. PTTG1 target genes were predicted by CistromeDB. Results The elevated expression level of PTTG1 was confirmed in 1037 BLCA samples compared with 127 non-BLCA samples, with a standardized mean difference value of 1.04. Higher PTTG1 expression status exhibited a poorer BLCA prognosis. Moreover, the PTTG1 Chronos genetic effect scores were negative, indicating that PTTG1 silence may inhibit the proliferation and survival of BLCA cells. With PTTG1 mRNA expression level increasing, higher natural killer, cytotoxic lymphocyte, and monocyte lineage cell infiltration levels were observed. A total of four candidate targets containing CHEK2, OCIAD2, UBE2L3, and ZNF367 were determined ultimately. Conclusions PTTG1 mRNA over-expression may become a potential biomarker for BLCA prognosis. Additionally, PTTG1 may correlate with the BLCA tumor microenvironment and exert transcriptional activity by targeting CHEK2, OCIAD2, UBE2L3, and ZNF367 in BLCA tissue. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09810-y.
Collapse
Affiliation(s)
- Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Abdirahman Ahmed Farah
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Gao-Qiang Zhai
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Rui-Gong Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Jia-Lin Liu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Qin-Jie Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Guan-Lan Zhang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Zi-Long Lei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Sheng-Hua Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Rd, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China.
| |
Collapse
|
4
|
Pituitary Tumor-Transforming Gene 1/Delta like Non-Canonical Notch Ligand 1 Signaling in Chronic Liver Diseases. Int J Mol Sci 2022; 23:ijms23136897. [PMID: 35805898 PMCID: PMC9267054 DOI: 10.3390/ijms23136897] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023] Open
Abstract
The management of chronic liver diseases (CLDs) remains a challenge, and identifying effective treatments is a major unmet medical need. In the current review we focus on the pituitary tumor transforming gene (PTTG1)/delta like non-canonical notch ligand 1 (DLK1) axis as a potential therapeutic target to attenuate the progression of these pathological conditions. PTTG1 is a proto-oncogene involved in proliferation and metabolism. PTTG1 expression has been related to inflammation, angiogenesis, and fibrogenesis in cancer and experimental fibrosis. On the other hand, DLK1 has been identified as one of the most abundantly expressed PTTG1 targets in adipose tissue and has shown to contribute to hepatic fibrosis by promoting the activation of hepatic stellate cells. Here, we extensively analyze the increasing amount of information pointing to the PTTG1/DLK1 signaling pathway as an important player in the regulation of these disturbances. These data prompted us to hypothesize that activation of the PTTG1/DLK1 axis is a key factor upregulating the tissue remodeling mechanisms characteristic of CLDs. Therefore, disruption of this signaling pathway could be useful in the therapeutic management of CLDs.
Collapse
|
5
|
PCLAF promotes neuroblastoma G1/S cell cycle progression via the E2F1/PTTG1 axis. Cell Death Dis 2022; 13:178. [PMID: 35210406 PMCID: PMC8873510 DOI: 10.1038/s41419-022-04635-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 12/20/2022]
Abstract
PCLAF (PCNA clamp-associated factor), also known as PAF15/ KIAA0101, is overexpressed in most human cancers and is a predominant regulator of tumor progression. However, its biological function in neuroblastoma remains unclear. PCLAF is extremely overexpressed in neuroblastoma and is associated with poor prognosis. Through the analysis of various data sets, we found that the high expression of PCLAF is positively correlated with increased stage and high risk of neuroblastoma. Most importantly, knocking down PCLAF could restrict the proliferation of neuroblastoma cells in vitro and in vitro. By analyzing RNA-seq data, we found that the enrichment of cell cycle-related pathway genes was most significant among the differentially expressed downregulated genes after reducing the expression of PCLAF. In addition, PCLAF accelerated the G1/S transition of the neuroblastoma cell cycle by activating the E2F1/PTTG1 signaling pathway. In this study, we reveal the mechanism by which PCLAF facilitates cell cycle progression and recommend that the PCLAF/E2F1/PTTG1 axis is a therapeutic target in neuroblastoma.
Collapse
|
6
|
Wang X, Younis S, Cen J, Wang Y, Krizhanovskii C, Andersson L, Welsh N. ZBED6 counteracts high-fat diet-induced glucose intolerance by maintaining beta cell area and reducing excess mitochondrial activation. Diabetologia 2021; 64:2292-2305. [PMID: 34296320 PMCID: PMC8423654 DOI: 10.1007/s00125-021-05517-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/01/2021] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS ZBED6 (zinc finger, BED-type containing 6) is known to regulate muscle mass by suppression of Igf2 gene transcription. In insulin-producing cell lines, ZBED6 maintains proliferative capacity at the expense of differentiation and beta cell function. The aim was to study the impact of Zbed6 knockout on beta cell function and glucose tolerance in C57BL/6 mice. METHODS Beta cell area and proliferation were determined in Zbed6 knockout mice using immunohistochemical analysis. Muscle and fat distribution were assessed using micro-computed tomography. Islet gene expression was assessed by RNA sequencing. Effects of a high-fat diet were analysed by glucose tolerance and insulin tolerance tests. ZBED6 was overexpressed in EndoC-βH1 cells and human islet cells using an adenoviral vector. Beta cell cell-cycle analysis, insulin release and mitochondrial function were studied in vitro using propidium iodide staining and flow cytometry, ELISA, the Seahorse technique, and the fluorescent probes JC-1 and MitoSox. RESULTS Islets from Zbed6 knockout mice showed lowered expression of the cell cycle gene Pttg1, decreased beta cell proliferation and decreased beta cell area, which occurred independently from ZBED6 effects on Igf2 gene expression. Zbed6 knockout mice, but not wild-type mice, developed glucose intolerance when given a high-fat diet. The high-fat diet Zbed6 knockout islets displayed upregulated expression of oxidative phosphorylation genes and genes associated with beta cell differentiation. In vitro, ZBED6 overexpression resulted in increased EndoC-βH1 cell proliferation and a reduced glucose-stimulated insulin release in human islets. ZBED6 also reduced mitochondrial JC-1 J-aggregate formation, mitochondrial oxygen consumption rates (OCR) and mitochondrial reactive oxygen species (ROS) production, both at basal and palmitate + high glucose-stimulated conditions. ZBED6-induced inhibition of OCR was not rescued by IGF2 addition. ZBED6 reduced levels of the mitochondrial regulator PPAR-γ related coactivator 1 protein (PRC) and bound its promoter/enhancer region. Knockdown of PRC resulted in a lowered OCR. CONCLUSIONS/INTERPRETATION It is concluded that ZBED6 is required for normal beta cell replication and also limits excessive beta cell mitochondrial activation in response to an increased functional demand. ZBED6 may act, at least in part, by restricting PRC-mediated mitochondrial activation/ROS production, which may lead to protection against beta cell dysfunction and glucose intolerance in vivo.
Collapse
Affiliation(s)
- Xuan Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Shady Younis
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Jing Cen
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Yun Wang
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Camilla Krizhanovskii
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Leif Andersson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
- Department of Veterinary Integrative Biosciences, Texas A & M University, College Station, TX, USA.
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Wang W, Cui J, Ma H, Lu W, Huang J. Targeting Pyrimidine Metabolism in the Era of Precision Cancer Medicine. Front Oncol 2021; 11:684961. [PMID: 34123854 PMCID: PMC8194085 DOI: 10.3389/fonc.2021.684961] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022] Open
Abstract
Metabolic rewiring is considered as a primary feature of cancer. Malignant cells reprogram metabolism pathway in response to various intrinsic and extrinsic drawback to fuel cell survival and growth. Among the complex metabolic pathways, pyrimidine biosynthesis is conserved in all living organism and is necessary to maintain cellular fundamental function (i.e. DNA and RNA biosynthesis). A wealth of evidence has demonstrated that dysfunction of pyrimidine metabolism is closely related to cancer progression and numerous drugs targeting pyrimidine metabolism have been approved for multiple types of cancer. However, the non-negligible side effects and limited efficacy warrants a better strategy for negating pyrimidine metabolism in cancer. In recent years, increased studies have evidenced the interplay of oncogenic signaling and pyrimidine synthesis in tumorigenesis. Here, we review the recent conceptual advances on pyrimidine metabolism, especially dihydroorotate dehydrogenase (DHODH), in the framework of precision oncology medicine and prospect how this would guide the development of new drug precisely targeting the pyrimidine metabolism in cancer.
Collapse
Affiliation(s)
- Wanyan Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiayan Cui
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hui Ma
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jin Huang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
8
|
Sun R, Liu J, Nie S, Li S, Yang J, Jiang Y, Cheng W. Construction of miRNA-mRNA Regulatory Network and Prognostic Signature in Endometrial Cancer. Onco Targets Ther 2021; 14:2363-2378. [PMID: 33854334 PMCID: PMC8039850 DOI: 10.2147/ott.s272222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/10/2021] [Indexed: 01/04/2023] Open
Abstract
Introduction This bioinformatic study confirmed a new miRNA-mRNA regulatory network and a prognostic signature in endometrial cancer (EC). Materials and Methods We downloaded RNA-seq and miRNA-seq data of EC from the TCGA database, then used EdegR package to screen differentially expressed miRNAs and mRNAs (DE-miRNAs and DE-mRNAs). Then, we constructed a regulatory network of EC-associated miRNAs and hub genes by Cytoscape, and determined the expression of unexplored miRNAs in EC tissues and normal adjacent tissues by quantitative Real-Time PCR (qRT-PCR). A prognostic signature model and a predictive nomogram were constructed. Finally, we explored the association between the prognostic model and the immune cell infiltration. Results A total of 11,531 DE-mRNAs and 236 DE-miRNAs, as well as 275 and 118 candidate DEGs for upregulated and downregulated DE-miRNAs were screened out. The miRNA-mRNA network included 5 downregulated and 13 upregulated DE-miRNAs. qRT-PCR proved that the expression levels of miRNA-18a-5p, miRNA-18b-5p, miRNA-449c-5p and miRNA-1224-5p and their target genes (NR3C1, CTGF, MYC, and TNS1) were consistent with our predictions. Univariate and multivariate Cox proportional hazards regression analyses of the hub genes revealed a significant prognostic value of NR3C1, EZH2, AND GATA4, and these genes were closely related to eight types of immune infiltration cells. Conclusion We identified three genes as candidate biomarkers for EC, which may provide a theoretical basis for targeted therapy.
Collapse
Affiliation(s)
- Rui Sun
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Sipei Nie
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Siyue Li
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Jing Yang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Yi Jiang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
9
|
Deng P, Tan M, Zhou W, Chen C, Xi Y, Gao P, Ma Q, Liang Y, Chen M, Tian L, Xie J, Liu M, Luo Y, Li Y, Zhang L, Wang L, Zeng Y, Pi H, Yu Z, Zhou Z. Bisphenol A promotes breast cancer cell proliferation by driving miR-381-3p-PTTG1-dependent cell cycle progression. CHEMOSPHERE 2021; 268:129221. [PMID: 33352510 DOI: 10.1016/j.chemosphere.2020.129221] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/22/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a high-production-volume industrial chemical that facilitates the development of breast cancer. However, the molecular mechanism associated with BPA-induced breast cancer cell proliferation and migration remains elusive. In our study, we exposed MCF-7 cells to different concentrations of BPA (0.1, 1 and 10 μM) for 24, 48, or 72 h. We found that BPA exposure significantly promoted MCF-7 cell proliferation and migration but not invasion. To elucidate the mechanisms, the differentially expressed genes between the BPA and control groups were investigated with the Gene Expression Omnibus (GEO) database through GEO2R. Kyoto Encyclopedia of Genes and Genomes (KEGG) and pathway action network analyses demonstrated the important role of the cell cycle pathway in the effects of BPA exposure on MCF-7 cells. Importantly, analysis with the cytoHubba plugin of Cytoscape software coupled with analysis of enriched genes in the cell cycle pathway identified PTTG1 and CDC20 (two hub genes) as key targets associated with BPA-induced MCF-7 cell proliferation and migration. Interestingly, BPA significantly increased the protein expression levels of PTTG1 but not CDC20. Knockdown of PTTG1 inhibited the BPA-induced increase in proliferation and maintained cell cycle progression. In addition, we confirmed that the increased expression of PTTG1 upon BPA exposure was caused by miR-381-3p inhibition. Moreover, we verified that miR-381-3p expression was low and inversely correlated with PTTG1 expression in breast cancer tissues. Together, these findings demonstrate that BPA promotes high PTTG1 expression and alters the cell cycle to enhance MCF-7 cell proliferation by inhibiting miR-381-3p expression.
Collapse
Affiliation(s)
- Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Miduo Tan
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Wei Zhou
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yu Xi
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yidan Liang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Mengyu Liu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Youlong Zeng
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
10
|
Sergeeva O, Zatsepin T. RNA Helicases as Shadow Modulators of Cell Cycle Progression. Int J Mol Sci 2021; 22:2984. [PMID: 33804185 PMCID: PMC8001981 DOI: 10.3390/ijms22062984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
The progress of the cell cycle is directly regulated by modulation of cyclins and cyclin-dependent kinases. However, many proteins that control DNA replication, RNA transcription and the synthesis and degradation of proteins can manage the activity or levels of master cell cycle regulators. Among them, RNA helicases are key participants in RNA metabolism involved in the global or specific tuning of cell cycle regulators at the level of transcription and translation. Several RNA helicases have been recently evaluated as promising therapeutic targets, including eIF4A, DDX3 and DDX5. However, targeting RNA helicases can result in side effects due to the influence on the cell cycle. In this review, we discuss direct and indirect participation of RNA helicases in the regulation of the cell cycle in order to draw attention to downstream events that may occur after suppression or inhibition of RNA helicases.
Collapse
Affiliation(s)
- Olga Sergeeva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30b1, 121205 Moscow, Russia;
| | - Timofei Zatsepin
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30b1, 121205 Moscow, Russia;
- Department of Chemistry, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
11
|
Pawlonka J, Rak B, Ambroziak U. The regulation of cyclin D promoters - review. Cancer Treat Res Commun 2021; 27:100338. [PMID: 33618151 DOI: 10.1016/j.ctarc.2021.100338] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 11/25/2022]
Abstract
Cyclins are key regulators of cell cycle progression and survival. Particularly cyclins D (cyclin D1, D2, and D3) act in response to the mitogenic stimulation and are pivotal mediators between proliferative pathways and the nuclear cell cycle machinery. Dysregulation of cyclins expression results in impaired development, abnormal cell growth or tumorigenesis. In this review we summarize current knowledge about regulatory role of the cyclin D promoters, transcriptional factors: regulators, co-activators and adaptor proteins necessary to their activation. We focused on the intracellular signaling pathways vital to cell growth, differentiation and apoptosis including transcription factor families: activator protein 1 (AP1), nuclear factor (NFκB), signal transducer and activator of transcription (STAT), cAMP response element-binding protein (CREB) and Sp/NF-Y, with a special insight into the tissue specific cyclin representation.
Collapse
Affiliation(s)
- Jan Pawlonka
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw
| | - Beata Rak
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw; Department of Genomic Medicine, Medical University of Warsaw, Warsaw.
| | - Urszula Ambroziak
- Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw
| |
Collapse
|
12
|
Hinojosa-Amaya JM, Lam-Chung CE, Cuevas-Ramos D. Recent Understanding and Future Directions of Recurrent Corticotroph Tumors. Front Endocrinol (Lausanne) 2021; 12:657382. [PMID: 33986726 PMCID: PMC8111286 DOI: 10.3389/fendo.2021.657382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Corticotroph tumors (CTs) are pituitary neoplasms arising from the Tpit lineage, which may or not express adrenocorticotrophic hormone (ACTH). Functioning CTs cause Cushing's disease (CD), which has high morbidity and mortality due to hypercortisolemia. "Non-functioning" or silent CTs (SCT) and the Crooke's cell subtypes do not cause CD and may be asymptomatic until manifested by compressive symptoms and are more frequently found as macroadenoma. Both tend toward more aggressive behavior, recurrence, and a higher rate of malignant transformation to pituitary carcinoma. Tumorigenesis involves genetic, epigenetic, and post-transcriptional disruption of cell-cycle regulators, which increase cell proliferation, POMC overexpression, ACTH transcription, and/or hypersecretion. Furthermore, functioning CTs develop resistance to glucocorticoid-mediated negative feedback on ACTH secretion, through increased expression of testicular orphan nuclear receptor 4 (TR4), heat-shock protein 90 (HSP90), and loss-of-function mutation of CDK5 and ABL enzyme substrate 1 (CABLES1) gene. Overt autonomous hypercortisolemia is difficult to control, and multiple diagnostic studies and therapeutic modalities are commonly required. Cell-cycle regulation depends mainly on p27, cyclin E, cyclin-dependent kinases (CDKs), and the retinoblastoma protein (Rb)/E2F1 transcription factor complex. Gain-of-function mutations of ubiquitin-specific protease (USP) 8, USP48, and BRAF genes may subsequently cause overexpression of epithelial growth factor receptor (EGFR), and enhance POMC transcription, cell proliferation, and tumor growth. Epigenetic changes through micro RNAs and decreased DNA deacetylation by histone deacetylase type 2 (HDAC2), may also affect tumor growth. All the former mechanisms may become interesting therapeutic targets for CTs, aside from temozolomide, currently used for aggressive tumors. Potential therapeutic agents are EGFR inhibitors such as gefitinib and lapatinib, the purine analog R-roscovitine by dissociation of CDK2/Cyclin E complex, the HSP90 inhibitor silibinin (novobiocin), to reduce resistance to glucocorticoid-mediated negative feedback, and BRAF inhibitors vemurafenib and dabrafenib in BRAF V600E positive tumors. This review summarizes the molecular mechanisms related to CTs tumorigenesis, their diagnostic approach, and provides an update of the potential novel therapies, from the lab bench to the clinical translation.
Collapse
Affiliation(s)
- José Miguel Hinojosa-Amaya
- Pituitary Clinic, Endocrinology Division, Department of Medicine, Hospital Universitario “Dr. José E. González” UANL, Monterrey, Mexico
| | - César Ernesto Lam-Chung
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Daniel Cuevas-Ramos
- Neuroendocrinology Clinic, Department of Endocrinology and Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- *Correspondence: Daniel Cuevas-Ramos,
| |
Collapse
|
13
|
Unravelling the diagnostic and therapeutic potentialities of a novel RNA aptamer isolated against human pituitary tumour transforming gene 1 (PTTG1) protein. Anal Chim Acta 2020; 1138:181-190. [PMID: 33161980 DOI: 10.1016/j.aca.2020.09.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Human Pituitary Tumour Transforming Gene 1 (PTTG1) is an oncoprotein involved in maintaining chromosome stability and acts as a biomarker for a panel of cancers. In this study, we endeavoured to generate an RNA aptamer against PTTG1. The RNA aptamer, SECURA-3 has an estimated equilibrium dissociation constant of 16.41 ± 6.4 nM. The aptamer was successfully harnessed in several diagnostic platforms including ELASA, aptamer-based dot blot and aptamer-based western blot. SECURA-3 was also unveiled as a potential probe that could replace its counterpart antibody in the histostaining-based detection of PTTG1 in HeLa and MCF-7 formalin-fixed paraffin-embedded cell blocks. In the aspect of therapeutics, SECURA-3 RNA aptamer demonstrates an antagonistic effect by antagonizing the interaction between PTTG1 and CXCR2, as revealed in the in vitro competitive nitrocellulose filter binding assay and dual-luciferase reporter assay in HeLa cells. As the first anti-PTTG1 aptamer, SECURA-3 RNA aptamer has immense diagnostic and therapeutic properties.
Collapse
|
14
|
Demin DE, Uvarova AN, Klepikova AV, Schwartz AM. The Influence of the Minor Short Isoform of Securin (PTTG1) on Transcription is Significantly Different from the Impact of the Full Isoform. Mol Biol 2020. [DOI: 10.1134/s0026893320010045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
15
|
Hu X, Lin J, Jiang M, He X, Wang K, Wang W, Hu C, Shen Z, He Z, Lin H, Wu D, Wang M. HIF-1α Promotes the Metastasis of Esophageal Squamous Cell Carcinoma by Targeting SP1. J Cancer 2020; 11:229-240. [PMID: 31892989 PMCID: PMC6930417 DOI: 10.7150/jca.35537] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/08/2019] [Indexed: 02/07/2023] Open
Abstract
Background: In microenvironment of malignant tumors, Hypoxia-Inducible Factors (HIF), most importantly HIF-1α, play an important role in regulation of adaptive biological response to hypoxia, promoting angiogenesis and metastasis. However, the underlying mechanism that HIF-1α regulates metastasis needs to be further clarified. Methods: The expressions of HIF-1α and SP1 were detected in 182 samples of esophageal squamous cell carcinoma (ESCC) and adjacent normal tissues by immunohistochemistry (IHC), and the correlation between the expression levels of HIF-1α and SP1 was analyzed. The expression of HIF-1α in ESCC cell lines TE1 and KYSE30 was then detected using qRT-PCR and western blot. The potential binding sites of HIF-1α on the SP1 promoter were analyzed using UCSC and JASPAR databases, verified by chromosomal immunoprecipitation (ChIP) assay and qRT-PCR. The effects of HIF-1α and SP1 on ESCC cell migration and invasion were then tested with Transwell and Matrigel experiments. Results: The expression of HIF-1α in cancer tissues is higher than adjacent normal tissues, and is correlated with metastasis, recurrence and poor prognosis. Upon silencing HIF-1α by siRNA, the invasion and migration ability of ESCC cells were significantly inhibited, which could be restored by the overexpression of SP1. Hypoxic conditions significantly increased the expression of HIF-1α and SP1 at both protein and mRNA levels in ESCC cells. HIF-1α enhanced SP1 transcription through binding to the promoter region. The expression of protein and mRNA levels of SP1 was decreased by silencing HIF-1α in cells. In contrast, overexpression of HIF-1α significantly increased the mRNA and protein levels of SP1. The expression of SP1 in ESCC was positively correlated with the protein expression of HIF-1α and poor prognosis. Conclusion: The results of our study indicate that HIF-1α promotes metastasis of ESCC by targeting SP1 in a hypoxic microenvironment. Further study on this mechanism may elucidate the possibility of HIF-1α and SP1 as new targets for the treatment of ESCC.
Collapse
Affiliation(s)
- Xueting Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Jiatong Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Ming Jiang
- Department of Thoracic Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China 510120
| | - Xiaotian He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Kefeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Wenjian Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Chuwen Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Zhiwen Shen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Zhanghai He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Duoguang Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| | - Minghui Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120.,Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China 510120
| |
Collapse
|
16
|
Xiong Z, Li X, Yang Q. PTTG has a Dual Role of Promotion-Inhibition in the Development of Pituitary Adenomas. Protein Pept Lett 2019; 26:800-818. [PMID: 37020362 DOI: 10.2174/0929866526666190722145449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 11/22/2022]
Abstract
Pituitary Tumor Transforming Gene (PTTG) of human is known as a checkpoint gene in the middle and late stages of mitosis, and is also a proto-oncogene that promotes cell cycle progression. In the nucleus, PTTG works as securin in controlling the mid-term segregation of sister chromatids. Overexpression of PTTG, entering the nucleus with the help of PBF in pituitary adenomas, participates in the regulation of cell cycle, interferes with DNA repair, induces genetic instability, transactivates FGF-2 and VEGF and promotes angiogenesis and tumor invasion. Simultaneously, overexpression of PTTG induces tumor cell senescence through the DNA damage pathway, making pituitary adenoma possessing the potential self-limiting ability. To elucidate the mechanism of PTTG in the regulation of pituitary adenomas, we focus on both the positive and negative function of PTTG and find out key factors interacted with PTTG in pituitary adenomas. Furthermore, we discuss other possible mechanisms correlate with PTTG in pituitary adenoma initiation and development and the potential value of PTTG in clinical treatment.
Collapse
Affiliation(s)
- Zujian Xiong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
17
|
Kong J, Wang T, Zhang Z, Yang X, Shen S, Wang W. Five Core Genes Related to the Progression and Prognosis of Hepatocellular Carcinoma Identified by Analysis of a Coexpression Network. DNA Cell Biol 2019; 38:1564-1576. [PMID: 31633379 DOI: 10.1089/dna.2019.4932] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The molecular mechanism of tumorigenesis of the prevalent cancer hepatocellular carcinoma (HCC) is unclear. In this study, through weighted gene coexpression network analysis, a coexpression network was constructed by selecting the top 25% most variant genes in the dataset GSE62232. The average linkage hierarchical clustering identified 24 modules, and among them, the pink module associated with prognosis of HCC was screened. Five gene candidates (PCNA, RFC4, PTTG1, H2AFZ, and RRM1) with a common network in the module were screened after the protein-protein interaction network complex was combined with the coexpression network. After progression and survival analysis, all candidates were identified as real core genes. According to the Human Protein Atlas and the Oncomine database, these genes were dysregulated in HCC samples. The receiver operating characteristic curve proved that the expression levels of the core genes had high diagnostic efficacy. The results of gene set enrichment analysis and functional enrichment analysis demonstrated the importance of the cell cycle-related pathways in HCC progression and prognosis. In conclusion, the five real core genes and cell cycle-related pathways identified in this study could greatly improve the knowledge about HCC progression and contribute to HCC treatment.
Collapse
Affiliation(s)
- Junjie Kong
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Tao Wang
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Zifei Zhang
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Xianwei Yang
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Shu Shen
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| | - Wentao Wang
- Department of Liver Surgery, Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, P.R. China
| |
Collapse
|
18
|
Lian SH, Song JD, Huang Y. PBF, a Proto-oncogene in Esophageal Carcinoma. Open Med (Wars) 2019; 14:748-756. [PMID: 31637306 PMCID: PMC6795029 DOI: 10.1515/med-2019-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 07/26/2019] [Indexed: 11/15/2022] Open
Abstract
Emerging evidence shows that the pituitary tumour-transforming gene (PTTG)-binding factor (PBF) functions as a proto-oncogene in some tumors. However, the precise functions of PBF in tumorigenesis and its action mechanisms remain largely unknown. Here for the first time we demonstrated that PBF was associated with a tumor-related cell phenotype in esophageal carcinoma (ESCA) and identified the involved signaling pathways. PBF was up-regulated in ESCA tissues (Data from GEPIA) and cells. Then we down-regulated PBF in ESCA cell lines, Eca-109 and TE-1, by using RNAi technology. Cell function analysis suggested that down-regulation of PBF could inhibit tumor-related cell phenotypes, including proliferation, motility, apoptosis and cell cycle, in Eca-109 and TE-1 cells. Mechanism investigation suggested that apoptosis induced by PBF knockdown may be mediated by the activation of the mitochondrial apoptosis pathway and cell cycle arrest. AKT/mTOR and Wnt3a/β-catenin, key pathways in regulating tumor proliferation and metastasis, were found to be inactivated by the down-regulation of PBF in ESCA cells. In conclusion, our study demonstrates that PBF functions as a proto-oncogene in ESCA in vitro, which may be mediated through AKT/mTOR and Wnt3a/β-catenin pathways.
Collapse
Affiliation(s)
- Shi-hai Lian
- Department of Cardiothoracic surgery, Zaozhuang Municipal Hospital, Zaozhuang 277000, Shandong Province, China
| | - Jun-ding Song
- Department of Cardiothoracic surgery, Zaozhuang Municipal Hospital, Zaozhuang 277000, Shandong Province, China
| | - Yi Huang
- Department of Cardiothoracic surgery, Zaozhuang Municipal Hospital, 41# Longtou Rd, Zaozhuang 277100, Shandong Province, China
| |
Collapse
|
19
|
Välimäki N, Schalin-Jäntti C, Karppinen A, Paetau A, Kivipelto L, Aaltonen LA, Karhu A. Genetic and Epigenetic Characterization of Growth Hormone-Secreting Pituitary Tumors. Mol Cancer Res 2019; 17:2432-2443. [PMID: 31578227 DOI: 10.1158/1541-7786.mcr-19-0434] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/25/2019] [Accepted: 09/27/2019] [Indexed: 11/16/2022]
Abstract
Somatic driver mechanisms of pituitary adenoma pathogenesis have remained incompletely characterized; apart from mutations in the stimulatory Gα protein (Gαs encoded by GNAS) causing activated cAMP synthesis, pathogenic variants are rarely found in growth hormone-secreting pituitary tumors (somatotropinomas). The purpose of the current work was to clarify how genetic and epigenetic alterations contribute to the development of somatotropinomas by conducting an integrated copy number alteration, whole-genome and bisulfite sequencing, and transcriptome analysis of 21 tumors. Somatic mutation burden was low, but somatotropinomas formed two subtypes associated with distinct aneuploidy rates and unique transcription profiles. Tumors with recurrent chromosome aneuploidy (CA) were GNAS mutation negative (Gsp- ). The chromosome stable (CS) -group contained Gsp+ somatotropinomas and two totally aneuploidy-free Gsp- tumors. Genes related to the mitotic G1-S-checkpoint transition were differentially expressed in CA- and CS-tumors, indicating difference in mitotic progression. Also, pituitary tumor transforming gene 1 (PTTG1), a regulator of sister chromatid segregation, showed abundant expression in CA-tumors. Moreover, somatotropinomas displayed distinct Gsp genotype-specific methylation profiles and expression quantitative methylation (eQTM) analysis revealed that inhibitory Gα (Gαi) signaling is activated in Gsp+ tumors. These findings suggest that aneuploidy through modulated driver pathways may be a causative mechanism for tumorigenesis in Gsp- somatotropinomas, whereas Gsp+ tumors with constitutively activated cAMP synthesis seem to be characterized by DNA methylation activated Gαi signaling. IMPLICATIONS: These findings provide valuable new information about subtype-specific pituitary tumorigenesis and may help to elucidate the mechanisms of aneuploidy also in other tumor types.
Collapse
Affiliation(s)
- Niko Välimäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Camilla Schalin-Jäntti
- Endocrinology, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Atte Karppinen
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, HUSLAB, University of Helsinki, Helsinki, Finland
| | - Leena Kivipelto
- Department of Neurosurgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland.,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| | - Auli Karhu
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland. .,Applied Tumor Genomics, Research Programs Unit, FI-00014 University of Helsinki, Finland
| |
Collapse
|
20
|
Ma K, Sun X, Ma L, Zhang S. Expression of Serum PTTG1 in Laryngeal Carcinoma and Its Correlation to Prognosis. Clin Exp Otorhinolaryngol 2019; 13:64-68. [PMID: 31446750 PMCID: PMC7010490 DOI: 10.21053/ceo.2019.00395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023] Open
Abstract
Objectives. The purpose of this study was to investigate serum pituitary tumor transforming gene (PTTG1) expression in laryngeal carcinoma and its relationship with the clinical pathological characteristics and prognosis. Methods. Expression of serum PTTG1 was measured by enzyme-linked immunosorbent assay in 110 patients with laryngeal carcinoma and 60 patients with vocal cord polyps. Expression of the serum PTTG1 levels relationship with the clinicopathological characteristics and prognosis were analyzed. Results. In laryngeal carcinoma patients’ serum, the PTTG1 median concentration was 141.43 pg/mL (interquartile range [IQR], 111.387 to 160.837 pg/mL), significantly higher than that of the vocal cord polyp group of 94.01 pg/mL (IQR, 81.26 to 108.59 pg/mL), and the difference was statistically significant (z=–6.715, P<0.001). PTTG1 expression with lymph node metastasis, clinical stage, and patients with laryngeal carcinoma was significantly correlated with the tumor differentiation degree (P<0.05). The total survival rate of the PTTG1 high expression group was significantly lower than the low expression group, and the difference of total survival time of the two groups was statistically significant (P<0.001). Conclusion. The PTTG1 expression level can be used as an index for evaluating prognosis of laryngeal cancer. High PTTG1 expression is one of the factors of poor prognosis of laryngeal carcinoma patients.
Collapse
Affiliation(s)
- Kunpeng Ma
- Department of Otorhinolaryngology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiuxia Sun
- Department of Otorhinolaryngology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Limin Ma
- Department of Plastic Surgery, Plastic Surgery Hospital of Weifang Medical University, Weifang, China
| | - Shenglin Zhang
- Department of Network Information Center, Weifang Medical University, Weifang, China
| |
Collapse
|
21
|
EZH2 cooperates with E2F1 to stimulate expression of genes involved in adrenocortical carcinoma aggressiveness. Br J Cancer 2019; 121:384-394. [PMID: 31363169 PMCID: PMC6738105 DOI: 10.1038/s41416-019-0538-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 11/08/2022] Open
Abstract
Background EZH2 is overexpressed and associated with poor prognosis in adrenocortical carcinoma (ACC) and its inhibition reduces growth and aggressiveness of ACC cells in culture. Although EZH2 was identified as the methyltransferase that deposits the repressive H3K27me3 histone mark, it can cooperate with transcription factors to stimulate gene transcription. Methods We used bioinformatics approaches on gene expression data from three cohorts of patients and a mouse model of EZH2 ablation, to identify targets and mode of action of EZH2 in ACC. This was followed by ChIP and functional assays to evaluate contribution of identified targets to ACC pathogenesis. Results We show that EZH2 mostly works as a transcriptional inducer in ACC, through cooperation with the transcription factor E2F1 and identify three positive targets involved in cell cycle regulation and mitosis i.e., RRM2, PTTG1 and ASE1/PRC1. Overexpression of these genes is associated with poor prognosis, suggesting a potential role in acquisition of aggressive ACC features. Pharmacological and siRNA-mediated inhibition of RRM2 blocks cell proliferation, induces apoptosis and inhibits cell migration, suggesting that it may be an interesting target in ACC. Conclusions Altogether, these data show an unexpected role of EZH2 and E2F1 in stimulating expression of genes associated with ACC aggressiveness.
Collapse
|
22
|
Demin DE, Bogolyubova AV, Zlenko DV, Uvarova AN, Deikin AV, Putlyaeva LV, Belousov PV, Mitkin NA, Korneev KV, Sviryaeva EN, Kulakovskiy IV, Tatosyan KA, Kuprash DV, Schwartz AM. The Novel Short Isoform of Securin Stimulates the Expression of Cyclin D3 and Angiogenesis Factors VEGFA and FGF2, but Does Not Affect the Expression of MYC Transcription Factor. Mol Biol 2018. [DOI: 10.1134/s0026893318030032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Fuertes M, Sapochnik M, Tedesco L, Senin S, Attorresi A, Ajler P, Carrizo G, Cervio A, Sevlever G, Bonfiglio JJ, Stalla GK, Arzt E. Protein stabilization by RSUME accounts for PTTG pituitary tumor abundance and oncogenicity. Endocr Relat Cancer 2018; 25:665-676. [PMID: 29622689 DOI: 10.1530/erc-18-0028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/05/2018] [Indexed: 11/08/2022]
Abstract
Increased levels of the proto-oncogene pituitary tumor-transforming gene 1 (PTTG) have been repeatedly reported in several human solid tumors, especially in endocrine-related tumors such as pituitary adenomas. Securin PTTG has a critical role in pituitary tumorigenesis. However, the cause of upregulation has not been found yet, despite analyses made at the gene, promoter and mRNA level that show that no mutations, epigenetic modifications or other mechanisms that deregulate its expression may explain its overexpression and action as an oncogene. We describe that high PTTG protein levels are induced by the RWD-containing sumoylation enhancer (RWDD3 or RSUME), a protein originally identified in the same pituitary tumor cell line in which PTTG was also cloned. We demonstrate that PTTG and RSUME have a positive expression correlation in human pituitary adenomas. RSUME increases PTTG protein in pituitary tumor cell lines, prolongs the half-life of PTTG protein and regulates the PTTG induction by estradiol. As a consequence, RSUME enhances PTTG transcription factor and securin activities. PTTG hyperactivity on the cell cycle resulted in recurrent and unequal divisions without cytokinesis, and the consequential appearance of aneuploidies and multinucleated cells in the tumor. RSUME knockdown diminishes securin PTTG and reduces its tumorigenic potential in a xenograft mouse model. Taken together, our findings show that PTTG high protein steady state levels account for PTTG tumor abundance and demonstrate a critical role of RSUME in this process in pituitary tumor cells.
Collapse
Affiliation(s)
- M Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - M Sapochnik
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - L Tedesco
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - S Senin
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - A Attorresi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - P Ajler
- Servicio de NeurocirugíaHospital Italiano, Buenos Aires, Argentina
| | - G Carrizo
- Servicio de NeurocirugíaHospital Italiano, Buenos Aires, Argentina
| | - A Cervio
- Departamento de NeurocirugíaFundación Para la Lucha Contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - G Sevlever
- Departamento de NeurocirugíaFundación Para la Lucha Contra las Enfermedades Neurológicas de la Infancia (FLENI), Buenos Aires, Argentina
| | - J J Bonfiglio
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
| | - G K Stalla
- Department of Clinical ResearchMax Planck Institute of Psychiatry, Munich, Germany
| | - E Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular y CelularFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
Fu D, Zhang Y, Cui H. Long noncoding RNA CCAT2 is activated by E2F1 and exerts oncogenic properties by interacting with PTTG1 in pituitary adenomas. Am J Cancer Res 2018; 8:245-255. [PMID: 29511595 PMCID: PMC5835692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/04/2017] [Indexed: 06/08/2023] Open
Abstract
Pituitary adenomas, arising from the pituitary gland cells, are one of the most frequent tumors found in the sella region. However, the molecular mechanisms involved in the carcinogenesis and progression of pituitary adenomas is still not understood in detail. Long noncoding RNA (lncRNA) colon cancer-associated transcript 2 (CCAT2), a newly identified lncRNA, has been reported to be abnormally expressed in some cancers. In the present study, we found that CCAT2 was significantly upregulated in pituitary adenomas tissues. Elevated CCAT2 expression was correlated with poor prognosis in patients with pituitary adenomas. Moreover, CCAT2 expression was activated by E2F1. Loss-of-function and gain-of-function assays showed that CCAT2 positively regulated pituitary adenoma cell proliferation, migration, and invasion. Further investigation demonstrated that CCAT2 interacted with PTTG1, and promoted its stability. Furthermore, CCAT2 affected the expression of downstream genes regulated by PTTG1, including SOX2, DLK1, MMP2, and MMP13. Cumulatively, CCAT2 functions as an oncogene in pituitary adenomas and its overexpression contributes to pituitary adenoma carcinogenesis and progression.
Collapse
Affiliation(s)
- Dongxia Fu
- The Second Department of Endocrinology, Cangzhou Central Hospital, Hebei Medical UniversityCangzhou, China
| | - Yunna Zhang
- The Second Department of Endocrinology, Cangzhou Central Hospital, Hebei Medical UniversityCangzhou, China
| | - Haibin Cui
- The First Department of Oncology Surgery, Cangzhou Central Hospital, Hebei Medical UniversityCangzhou, China
| |
Collapse
|
25
|
Araki T, Liu NA. Cell Cycle Regulators and Lineage-Specific Therapeutic Targets for Cushing Disease. Front Endocrinol (Lausanne) 2018; 9:444. [PMID: 30147673 PMCID: PMC6096271 DOI: 10.3389/fendo.2018.00444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/18/2018] [Indexed: 11/22/2022] Open
Abstract
Cell cycle proteins are critical to pituitary development, but their contribution to lineage-specific tumorigenesis has not been well-elucidated. Emerging evidence from in vitro human tumor analysis and transgenic mouse models indicates that G1/S-related cell cycle proteins, particularly cyclin E, p27, Rb, and E2F1, drive molecular mechanisms that underlie corticotroph-specific differentiation and development of Cushing disease. The aim of this review is to summarize the literature and discuss the complex role of cell cycle regulation in Cushing disease, with a focus on identifying potential targets for therapeutic intervention in patients with these tumors.
Collapse
Affiliation(s)
- Takako Araki
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Takako Araki
| | - Ning-Ai Liu
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
26
|
RNF20 Suppresses Tumorigenesis by Inhibiting the SREBP1c-PTTG1 Axis in Kidney Cancer. Mol Cell Biol 2017; 37:MCB.00265-17. [PMID: 28827316 DOI: 10.1128/mcb.00265-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/29/2017] [Indexed: 12/21/2022] Open
Abstract
Elevated lipid metabolism promotes cancer cell proliferation. Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancers, characterized by ectopic lipid accumulation. However, the relationship between aberrant lipid metabolism and tumorigenesis in ccRCC is not thoroughly understood. Here, we demonstrate that ring finger protein 20 (RNF20) acts as a tumor suppressor in ccRCC. RNF20 overexpression repressed lipogenesis and cell proliferation by inhibiting sterol regulatory element-binding protein 1c (SREBP1c), and SREBP1 suppression, either by knockdown or by the pharmacological inhibitor betulin, attenuated proliferation and cell cycle progression in ccRCC cells. Notably, SREBP1c regulates cell cycle progression by inducing the expression of pituitary tumor-transforming gene 1 (PTTG1), a novel target gene of SREBP1c. Furthermore, RNF20 overexpression reduced tumor growth and lipid storage in xenografts. In ccRCC patients, RNF20 downregulation and SREBP1 activation are markers of poor prognosis. Therefore, RNF20 suppresses tumorigenesis in ccRCC by inhibiting the SREBP1c-PTTG1 axis.
Collapse
|
27
|
Yang C, Li J, Yu L, Zhang Z, Xu F, Jiang L, Zhou X, He S. Regulation of RIP3 by the transcription factor Sp1 and the epigenetic regulator UHRF1 modulates cancer cell necroptosis. Cell Death Dis 2017; 8:e3084. [PMID: 28981102 PMCID: PMC5682651 DOI: 10.1038/cddis.2017.483] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022]
Abstract
Receptor-interacting kinase-3 (RIP3) is a key regulator of necroptosis. It has been shown that the expression of RIP3 is silenced in most cancer cells and tissues due to genomic methylation. However, the regulatory mechanisms controlling RIP3 expression in cancer cells have not been fully elucidated. Here, we report that Sp1, a well-characterized zinc-finger transcription factor, directly regulates RIP3 expression in cancer cells. Knockdown of endogenous Sp1 significantly decreases the transcription of Rip3, thereby further inhibiting necroptosis. The re-expression of Sp1 restores the necroptotic response. In addition, knockdown of epigenetic regulator UHRF1 (ubiquitin-like, containing PHD and RING finger domains 1) in RIP3-null cancer cells reduces the methylation level of the Rip3 promoter. This effect is sufficient to trigger the expression of RIP3 in RIP3-null cancer cells. The induced expression of RIP3 by UHRF1 RNAi depends on the presence of Sp1. Remarkably, the ectopic expression of RIP3 in RIP3-null cancer cells results in a decrease in tumor growth in mice. Therefore, our findings offer insights into RIP3 expression control in cancer cells and suggest an inhibitory effect of RIP3 on tumorigenesis.
Collapse
Affiliation(s)
- Chengkui Yang
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Jun Li
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Lu Yu
- Department of Emergency Medicine, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Zili Zhang
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Feng Xu
- Department of Emergency Medicine, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Lang Jiang
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Xiuxia Zhou
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Sudan He
- Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
28
|
Pappas L, Xu XL, Abramson DH, Jhanwar SC. Genomic instability and proliferation/survival pathways in RB1-deficient malignancies. Adv Biol Regul 2017; 64:20-32. [PMID: 28242412 DOI: 10.1016/j.jbior.2017.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 01/31/2017] [Indexed: 06/06/2023]
Abstract
Genomic instability (GIN) is a hallmark of most cancer cells. However, compared to most human cancer cell types, the retinoblastoma tumor cells show a relatively stable genome. The fundamental basis of this genomic stability has yet to be elucidated, and the role of certain proteins involved in cell cycle regulation may be the key to the development of these specific genotypes. We examined whether thyroid hormone receptor beta 1 and 2 (TRβ1 and TRβ2), known to regulate tumorigenesis, and PTTG1, a mitotic checkpoint protein, play a role in maintaining genomic stability in retinoblastoma. In order to elucidate the role of these proteins in development of aneuploidy/polyploidy, an indicator of GIN, we first studied comparative GIN in retinoblastomas and multiple RB mutant cancer cell lines using single nucleotide polymorphism (SNP) analysis. We then utilized pLKO lentiviral vectors to selectively modify expression of the targeted cell cycle proteins and interpret their effect on downstream cell cycle proteins and their relative effects on the development of polyploidy in multiple tumor cell lines. The SNP analysis showed that retinoblastomas displayed relatively fewer genomic copy number changes as compared to other RB1-deficient cancer cell lines. Both TRβ1 and TRβ2 knockdown led to accumulation of E2F1 and PTTG1 and increased GIN as demonstrated by an increase in polyploidy. Downregulation of PTTG1 led to a relative decrease in GIN while upregulation of PTTG1 led to a relative increase in GIN. Knockdown of E2F1 led to a downstream decrease in PTTG1 expression. Rb-knockdown also upregulated E2F1 and PTTG1 leading to increased GIN. We showed that Rb is necessary for PTTG1 inhibition and genomic stability. A relatively stable genome in retinoblastoma tumor cells is maintained by TRβ1 and TRβ2-mediated PTTG1 inhibition, counteracting Rb-deficiency-related GIN. TRβ1, TRβ2 and Rb-KD all led to the downstream PTTG1 accumulation, apparently through an activation of E2F1 resulting in extensive genomic instability as seen in other Rb-deficient tumors.
Collapse
Affiliation(s)
- Lara Pappas
- Departments of Pathology, Pediatrics, Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Xiaoliang Leon Xu
- Departments of Pathology, Pediatrics, Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - David H Abramson
- Departments of Pathology, Pediatrics, Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Suresh C Jhanwar
- Departments of Pathology, Pediatrics, Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA.
| |
Collapse
|
29
|
Li Y, Guo A, Feng Y, Zhang Y, Wang J, Jing L, Yan Y, Jing L, Liu Z, Ma L, Diao A. Sp1 transcription factor promotes TMEPAI gene expression and contributes to cell proliferation. Cell Prolif 2016; 49:710-719. [PMID: 27625141 DOI: 10.1111/cpr.12292] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/11/2016] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVES TMEPAI (transmembrane prostate androgen-induced protein) has been reported to be overexpressed during tumour progression; however, little is known concerning transcriptional mechanisms regulating TMEPAI gene expression. MATERIALS AND METHODS In this study, the TMEPAI gene promoter has been identified and characterized, and the effects of Sp1 on TMEPAI-induced viability of A549 cells were evaluated, using MTT and colony formation assays. RESULTS We found that the sequence between -298 and +24 consists of basal promoter activity for TMEPAI. Further analysis indicated that two Sp1-binding sites are crucial for maintaining basal transcriptional activity of the TMEPAI promoter, and chromatin immunoprecipitation assays confirmed direct binding of Sp1 to the TMEPAI promoter. In addition, Sp1 up-regulated TMEPAI protein expression, as well as Sp1 promoting TMEPAI-induced cell proliferation. CONCLUSIONS These results indicate that the sequence between -298 and +24 consists of the basal promoter activity for TMEPAI. Sp1 promotes TMEPAI expression and contributes to cell proliferation.
Collapse
Affiliation(s)
- Yuyin Li
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ailong Guo
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yajuan Feng
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yueying Zhang
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jianjun Wang
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Lifang Jing
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yali Yan
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Lei Jing
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Zhenxing Liu
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Long Ma
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Aipo Diao
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, School of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
30
|
The PTTG1-targeting miRNAs miR-329, miR-300, miR-381, and miR-655 inhibit pituitary tumor cell tumorigenesis and are involved in a p53/PTTG1 regulation feedback loop. Oncotarget 2016; 6:29413-27. [PMID: 26320179 PMCID: PMC4745736 DOI: 10.18632/oncotarget.5003] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/27/2015] [Indexed: 12/23/2022] Open
Abstract
Deregulation of the pituitary tumor transforming gene (PTTG1), a newly discovered oncogene, is a hallmark of various malignancies, including pituitary tumors. However, the mechanisms regulating PTTG1 expression are still needed to be explored. MicroRNAs (miRNAs) are a novel class of small RNA molecules that act as posttranscriptional regulators of gene expression and can play a significant role in tumor development. Here, we identified a series of miRNAs, namely, miR-329, miR-300, miR-381 and miR-655, which could target PTTG1 messenger RNA and inhibit its expression. Interestingly, all four miRNAs significantly that are downregulated in pituitary tumors were mapped to the 14q32.31 locus, which acts as a tumor suppressor in several cancers. Functional studies show that the PTTG1-targeting miRNAs inhibit proliferation, migration and invasion but induce apoptosis in GH3 and MMQ cells. Furthermore, overexpression of a PTTG1 expression vector lacking the 3′UTR partially reverses the tumor suppressive effects of these miRNAs. Next, we identified the promoter region of PTTG1-targeting miRNAs with binding sites for p53. In our hands, p53 transcriptionally activated the expression of these miRNAs in pituitary tumor cells. Finally, we found that PTTG1 could inhibit p53 transcriptional activity to the four miRNAs. These data indicate the existence of a feedback loop between PTTG1 targeting miRNAs, PTTG1 and p53 that promotes pituitary tumorigenesis. Together, these findings suggest that these PTTG1-targeting miRNAs are important players in the regulation of pituitary tumorigenesis and that these miRNAs may serve as valuable therapeutic targets for cancer treatment.
Collapse
|
31
|
Xie B, Qin Z, Liu S, Nong S, Ma Q, Chen B, Liu M, Pan T, Liao DJ. Cloning of Porcine Pituitary Tumor Transforming Gene 1 and Its Expression in Porcine Oocytes and Embryos. PLoS One 2016; 11:e0153189. [PMID: 27058238 PMCID: PMC4825983 DOI: 10.1371/journal.pone.0153189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/24/2016] [Indexed: 11/30/2022] Open
Abstract
The maternal-to-embryonic transition (MET) is a complex process that occurs during early mammalian embryogenesis and is characterized by activation of the zygotic genome, initiation of embryonic transcription, and replacement of maternal mRNA with embryonic mRNA. The objective of this study was to reveal the temporal expression and localization patterns of PTTG1 during early porcine embryonic development and to establish a relationship between PTTG1 and the MET. To achieve this goal, reverse transcription-polymerase chain reaction (RT-PCR) was performed to clone porcine PTTG1. Subsequently, germinal vesicle (GV)- and metaphase II (MII)-stage oocytes, zygotes, 2-, 4-, and 8-cell-stage embryos, morulas, and blastocysts were produced in vitro and their gene expression was analyzed. The results revealed that the coding sequence of porcine PTTG1 is 609-bp in length and that it encodes a 202-aa polypeptide. Using qRT-PCR, PTTG1 mRNA expression was observed to be maintained at high levels in GV- and MII-stage oocytes. The transcript levels in oocytes were also significantly higher than those in embryos from the zygote to blastocyst stages. Immunohistochemical analyses revealed that porcine PTTG1 was primarily localized to the cytoplasm and partially localized to the nucleus. Furthermore, the PTTG1 protein levels in MII-stage oocytes and zygotes were significantly higher than those in embryos from the 2-cell to blastocyst stage. After fertilization, the level of this protein began to decrease gradually until the blastocyst stage. The results of our study suggest that porcine PTTG1 is a new candidate maternal effect gene (MEG) that may participate in the processes of oocyte maturation and zygotic genome activation during porcine embryogenesis.
Collapse
Affiliation(s)
- Bingkun Xie
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
- * E-mail:
| | - Zhaoxian Qin
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - Shuai Liu
- Hebei Research Institute for Family Planning, Shijiazhang, Hebei, P. R. China
| | - Suqun Nong
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - Qingyan Ma
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - Baojian Chen
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - Mingjun Liu
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - Tianbiao Pan
- Guangxi Key Laboratory of Livestock Genetic Improvement, Guangxi Institute of Animal Sciences, Nanning, Guangxi, P. R. China
| | - D. Joshua Liao
- Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| |
Collapse
|
32
|
Huang Z, Huang L, Shen S, Li J, Lu H, Mo W, Dang Y, Luo D, Chen G, Feng Z. Sp1 cooperates with Sp3 to upregulate MALAT1 expression in human hepatocellular carcinoma. Oncol Rep 2015; 34:2403-12. [PMID: 26352013 DOI: 10.3892/or.2015.4259] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/03/2015] [Indexed: 01/14/2023] Open
Abstract
Long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), also known as nuclear-enriched transcript 2 (NEAT2), is highly conserved among mammals and highly expressed in the nucleus. It was first identified in lung cancer as a prognostic marker for metastasis but is also associated with several other solid tumors. In hepatocellular carcinoma (HCC), MALAT1 is a novel biomarker for predicting tumor recurrence after liver transplantation. The mechanism of overexpression in tumor progression remains unclear. In the present study, we investigated the role of specificity protein 1/3 (Sp1/3) in regulation of MALAT1 transcription in HCC cells. The results showed a high expression of Sp1, Sp3 and MALAT1 in HCC vs. paired non-tumor liver tissues, which was associated with the AFP level (Sp1, r=7.44, P=0.0064; MALAT1, r=12.37, P=0.0004). Co-silencing of Sp1 and Sp3 synergistically repressed MALAT1 expression. Sp1 binding inhibitor, mithramycin A (MIT), also inhibited MALAT1 expression in HCC cells. In conclusion, the upstream of MALAT1 contains five Sp1/3 binding sites, which may be responsible for MALAT1 transcription. Inhibitors, such as MIT, provide a potential therapeutic strategy for HCC patients with MALAT1 overexpression.
Collapse
Affiliation(s)
- Ziling Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lanshan Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Siqiao Shen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jia Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Huiping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Weijia Mo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yiwu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Dianzhong Luo
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhenbo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
33
|
Noll JE, Vandyke K, Hewett DR, Mrozik KM, Bala RJ, Williams SA, Kok CH, Zannettino AC. PTTG1 expression is associated with hyperproliferative disease and poor prognosis in multiple myeloma. J Hematol Oncol 2015; 8:106. [PMID: 26445238 PMCID: PMC4595141 DOI: 10.1186/s13045-015-0209-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023] Open
Abstract
Background Multiple myeloma (MM) is an incurable haematological malignancy characterised by the clonal proliferation of malignant plasma cells within the bone marrow. We have previously identified pituitary tumour transforming gene 1 (Pttg1) as a gene that is significantly upregulated in the haematopoietic compartment of the myeloma-susceptible C57BL/KaLwRij mouse strain, when compared with the myeloma-resistant C57BL/6 mouse. Over-expression of PTTG1 has previously been associated with malignant progression and an enhanced proliferative capacity in solid tumours. Methods In this study, we investigated PTTG1 gene and protein expression in MM plasma cells from newly diagnosed MM patients. Gene expression profiling was used to identify gene signatures associated with high PTTG1 expression in MM patients. Additionally, we investigated the effect of short hairpin ribonucleic acid (shRNA)-mediated PTTG1 knockdown on the proliferation of the murine myeloma plasma cell line 5TGM1 in vitro and in vivo. Results PTTG1 was found to be over-expressed in 36–70 % of MM patients, relative to normal controls, with high PTTG1 expression being associated with poor patient outcomes (hazard ratio 2.49; 95 % CI 1.28 to 4.86; p = 0.0075; log-rank test). In addition, patients with high PTTG1 expression exhibited increased expression of cell proliferation-associated genes including CCNB1, CCNB2, CDK1, AURKA, BIRC5 and DEPDC1. Knockdown of Pttg1 in 5TGM1 cells decreased cellular proliferation, without affecting cell cycle distribution or viability, and decreased expression of Ccnb1, Birc5 and Depdc1 in vitro. Notably, Pttg1 knockdown significantly reduced MM tumour development in vivo, with an 83.2 % reduction in tumour burden at 4 weeks (p < 0.0001, two-way ANOVA). Conclusions This study supports a role for increased PTTG1 expression in augmenting tumour development in a subset of MM patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0209-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacqueline E Noll
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Kate Vandyke
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,SA Pathology, Adelaide, Australia.
| | - Duncan R Hewett
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Rachel J Bala
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Sharon A Williams
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Chung H Kok
- Leukaemia Research Group, Cancer Theme, SAHMRI, Adelaide, Australia.
| | - Andrew Cw Zannettino
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,Discipline of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide, Cancer Theme, Level 5 South, SAHMRI, PO Box 11060, Adelaide, SA, 5001, Australia.
| |
Collapse
|
34
|
Lanigan F, Brien GL, Fan Y, Madden SF, Jerman E, Maratha A, Aloraifi F, Hokamp K, Dunne EJ, Lohan AJ, Flanagan L, Garbe JC, Stampfer MR, Fridberg M, Jirstrom K, Quinn CM, Loftus B, Gallagher WM, Geraghty J, Bracken AP. Delineating transcriptional networks of prognostic gene signatures refines treatment recommendations for lymph node-negative breast cancer patients. FEBS J 2015; 282:3455-73. [DOI: 10.1111/febs.13354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/02/2015] [Accepted: 06/17/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Fiona Lanigan
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Gerard L. Brien
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Yue Fan
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - Stephen F. Madden
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - Emilia Jerman
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Ashwini Maratha
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - Fatima Aloraifi
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Karsten Hokamp
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Eiseart J. Dunne
- The Smurfit Institute of Genetics; Trinity College Dublin; Ireland
| | - Amanda J. Lohan
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - Louise Flanagan
- Department of Histopathology; St Vincent's University Hospital; Dublin Ireland
| | - James C. Garbe
- Life Science Division; Lawrence Berkeley National Laboratory; Berkeley CA USA
| | - Martha R. Stampfer
- Life Science Division; Lawrence Berkeley National Laboratory; Berkeley CA USA
| | - Marie Fridberg
- Department of Clinical Sciences, Division of Oncology and Pathology; Lund University; Sweden
| | - Karin Jirstrom
- Department of Clinical Sciences, Division of Oncology and Pathology; Lund University; Sweden
| | - Cecily M. Quinn
- Department of Histopathology; St Vincent's University Hospital; Dublin Ireland
| | - Brendan Loftus
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - William M. Gallagher
- UCD School of Biomolecular and Biomedical Science; UCD Conway Institute; University College Dublin; Ireland
| | - James Geraghty
- Department of Histopathology; St Vincent's University Hospital; Dublin Ireland
| | | |
Collapse
|
35
|
Castilla C, Flores ML, Medina R, Pérez-Valderrama B, Romero F, Tortolero M, Japón MA, Sáez C. Prostate cancer cell response to paclitaxel is affected by abnormally expressed securin PTTG1. Mol Cancer Ther 2014; 13:2372-83. [PMID: 25122070 DOI: 10.1158/1535-7163.mct-13-0405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PTTG1 protein, the human securin, has a central role in sister chromatid separation during mitosis, and its altered expression has been reported in many tumor types. Paclitaxel is a widely used chemotherapeutic drug, whose mechanism of action is related to its ability to arrest cells in mitosis and the subsequent induction of the intrinsic apoptotic pathway. By using two prostate cancer cell lines with different responses to paclitaxel treatment, we have identified two situations in which PTTG1 influences cell fate differentially. In slippage-prone PC3 cells, both PTTG1 downregulation and overexpression induce an increase in mitotic cells that is associated with diminished apoptosis after paclitaxel treatment. In LNCaP cells, however, PTTG1 downregulation prevents mitotic entry and, subsequently, inhibits mitosis-associated, paclitaxel-induced apoptosis. In contrast, PTTG1 overexpression induces an increase in mitotic cells and apoptosis after paclitaxel treatment. We have also identified a role for Mcl-1 protein in preventing apoptosis during mitosis in PC3 cells, as simultaneous PTTG1 and Mcl-1 silencing enhances mitosis-associated apoptosis after paclitaxel treatment. The finding that a more efficient mitotic arrest alone in PC3 cells is not enough to increase apoptosis was also confirmed with the observation that a selected paclitaxel-resistant PC3 cell line showed an apoptosis-resistant phenotype associated with increased mitosis upon paclitaxel treatment. These findings could contribute to identify putative responsive and nonresponsive cells and help us to approach incomplete responses to paclitaxel in the clinical setting.
Collapse
Affiliation(s)
- Carolina Castilla
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - M Luz Flores
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Rafael Medina
- Department of Urology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Francisco Romero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - María Tortolero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Miguel A Japón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| | - Carmen Sáez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| |
Collapse
|
36
|
Lee HJ, Choi JH, Jung J, Kim JK, Lee SS, Kim GJ. Changes in PTTG1 by human TERT gene expression modulate the self-renewal of placenta-derived mesenchymal stem cells. Cell Tissue Res 2014; 357:145-57. [PMID: 24816985 DOI: 10.1007/s00441-014-1874-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 03/13/2014] [Indexed: 01/01/2023]
Abstract
In addition to their differentiation potential, self-renewal capability is an important characteristic of stem cells. The limited self-renewal activity of mesenchymal stem cells is the greatest obstacle to the application of stem cell therapy in regenerative medicine. The human TERT gene enhances the self-renewal of MSCs, but the mechanism of self-renewal and the interactions among TERT-gene-related molecules remain unknown. The objectives of this study were to generate immortalized MSCs derived from MSCs isolated from placenta (naive) by human TERT gene transfection with the AMAXA gene delivery system, to compare their characteristics, and to investigate whether increased TERT expression affected the pituitary tumor transforming gene (PTTG1; also known as securin), which is involved in chromosome segregation during mitosis. TERT-immortalized cells (TERT+) with a prolonged life span displayed high PTTG1 expression. TERT+ cells also retained the stemness capacity and multipotency of naive cells and displayed high PTTG1 expression. However, down-regulation of PTTG1 by treatment with short interfering RNA induced cell senescence and decreased telomerase activity. Moreover, TERT bound to PTTG1 formed complexes with chaperones such as Ku70 and heat shock protein 90. Thus, placental MSCs immortalized by TERT gene transfection display differentiation potential and exhibit enhanced self-renewal through a balanced interaction of PTTG1 and chaperones. The interaction between TERT and PTTG1 by association of Ku70 might be important for the enhancement of the limited self-renewal activity of MSCs and for understanding the regulatory mechanisms of self-renewal.
Collapse
Affiliation(s)
- Hyun-Jung Lee
- Department of Biomedical Science, CHA University, 606-16 Yeoksam1-dong, Kangnam-Gu, Seoul, 135-097, Republic of Korea
| | | | | | | | | | | |
Collapse
|
37
|
PTTG1 overexpression in adrenocortical cancer is associated with poor survival and represents a potential therapeutic target. Surgery 2014; 154:1405-16; discussion 1416. [PMID: 24238056 DOI: 10.1016/j.surg.2013.06.058] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/28/2013] [Indexed: 11/20/2022]
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is associated with poor survival rates. The objective of the study was to analyze ACC gene expression profiling data for prognostic biomarkers and therapeutic targets. METHODS We profiled 44 ACC and 4 normal adrenals on Affymetrix U133 Plus 2 expression microarrays. Pathway and transcriptional enrichment analysis was performed. Protein levels were determined by Western blot. Drug efficacy was assessed against ACC cell lines. Previously published expression datasets were analyzed for validation. RESULTS Pathway enrichment analysis identified marked dysregulation of cyclin-dependent kinases and mitosis. Overexpression of PTTG1, which encodes securin, a negative regulator of p53, was identified as a marker of poor survival. Median survival for patients with tumors expressing high PTTG1 levels (log2 ratio of PTTG1 to average β-actin <-3.04) was 1.8 years compared with 9.0 years if tumors expressed lower levels of PTTG1 (P < .0001). Analysis of a previously published dataset confirmed the association of high PTTG1 expression with a poor prognosis. Treatment of 2 ACC cell lines with vorinostat decreased securin levels and inhibited cell growth (median inhibition concentrations of 1.69 μmol/L and 0.891 μmol/L, for SW-13 and H295R, respectively). CONCLUSION Overexpression of PTTG1 is correlated with poor survival in ACC. PTTG1/securin is a prognostic biomarker and warrants investigation as a therapeutic target.
Collapse
|
38
|
Pttg1/securin is required for the branching morphogenesis of the mammary gland and suppresses mammary tumorigenesis. Proc Natl Acad Sci U S A 2014; 111:1008-13. [PMID: 24395789 DOI: 10.1073/pnas.1318124111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pituitary tumor transforming gene 1 (Pttg1) encodes the mammalian securin, which is an inhibitor of separase (a protease required for the separation of sister chromatids in mitosis and meiosis). PTTG1 is overexpressed in a number of human cancers and has been suggested to be an oncogene. However, we found that, in Pttg1-mutant females, the mammary epithelial cells showed increased proliferation and precocious branching morphogenesis. In accord with these phenotypic changes, progesterone receptor, cyclin D1, and Mmp2 were up-regulated whereas p21 (Cdkn1a) was down-regulated. These molecular changes provide explanation for the observed developmental defects, and suggest that Pttg1 is a tumor suppressor. Indeed, mice lacking Pttg1 developed spontaneous mammary tumors. Furthermore, in human breast tumors, PTTG1 protein levels were down-regulated and the reduction was significantly correlated with the tumor grade.
Collapse
|
39
|
Analysis of regulatory networks constructed based on gene coexpression in pituitary adenoma. J Genet 2013; 92:489-97. [DOI: 10.1007/s12041-013-0299-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
40
|
Chen PY, Yen JH, Kao RH, Chen JH. Down-regulation of the oncogene PTTG1 via the KLF6 tumor suppressor during induction of myeloid differentiation. PLoS One 2013; 8:e71282. [PMID: 23977008 PMCID: PMC3745464 DOI: 10.1371/journal.pone.0071282] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/26/2013] [Indexed: 01/04/2023] Open
Abstract
The aberrant expression of proto-oncogenes is involved in processes that are responsible for cellular proliferation and the inhibition of myeloid differentiation in acute myeloid leukemia (AML). Pituitary Tumor-Transforming gene 1 (PTTG1), an oncogenic transcription factor, is abundantly expressed in various human cancers and hematopoietic malignancies. However, its expression in normal leukocytes and most normal tissues is very low or undetectable. The mechanism by which PTTG1 overexpression modifies myeloid cell development and promotes leukemogenesis remain unclear. To investigate the mechanistic links between PTTG1 overexpression and leukemia cell differentiation, we utilized phorbol 12-myristate 13-acetate (PMA), a well-known agent that triggers monocyte/macrophage differentiation, to analyze the expression patterns of PTTG1 in PMA-induced myeloid differentiation. We found that PTTG1 is down-regulated at the transcriptional level in PMA-treated HL-60 and THP1 cells. In addition, we identified a binding site for a tumor suppressor protein, Kruppel-like factor 6 (KLF6), in the PTTG1 promoter. We found that KLF6 could directly bind and repress PTTG1 expression. In HL-60 and THP1 cells, KLF6 mRNA and protein levels are up-regulated with a concordant reduction of PTTG1 expression upon treatment with PMA. Furthermore, KLF6 knockdown by shRNA abolished the suppression of PTTG1 and reduced the activation of the differentiation marker CD11b in PMA-primed cells. The protein kinase C (PKC) inhibitor and the MAPK/ERK kinase (MEK) inhibitor significantly blocked the potentiation of PMA-mediated KLF6 induction and the down-regulation of PTTG1, indicating that PTTG1 is suppressed via the activation of PKC/ERK/KLF6 pathway. Our findings suggest that drugs that increase the KLF6 inhibition of PTTG1 may have a therapeutic application in AML treatment strategies.
Collapse
Affiliation(s)
- Pei-Yi Chen
- Institute of Medical Science, Tzu Chi University, Hualien, Taiwan
- Center of Medical Genetics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Jui-Hung Yen
- Institute of Medical Science, Tzu Chi University, Hualien, Taiwan
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Ruey-Ho Kao
- Department of Hematology-Oncology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Ji-Hshiung Chen
- Institute of Medical Science, Tzu Chi University, Hualien, Taiwan
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
- * E-mail:
| |
Collapse
|
41
|
PTTG acts as a STAT3 target gene for colorectal cancer cell growth and motility. Oncogene 2013; 33:851-61. [PMID: 23416975 PMCID: PMC3930149 DOI: 10.1038/onc.2013.16] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 12/19/2012] [Accepted: 12/21/2012] [Indexed: 12/29/2022]
Abstract
Pituitary tumor transforming gene (PTTG), the index mammalian securin, is abundantly expressed in several tumors and regulates tumor growth and progression. Molecular mechanisms elucidating PTTG regulation and actions remain elusive. Here, we provide evidence that PTTG acts as a STAT3 target gene. Total STAT3 and Tyr705 phosphorylated STAT3 were concordantly expressed with PTTG in human colorectal tumors (n=97 and n=95 respectively, P<0.001). STAT3 specifically bound the human PTTG promoter and induced PTTG transcriptional activity (2-fold) as assessed by chromatin immunoprecipitation and luciferase reporter assays. STAT3 transfection increased PTTG mRNA and protein abundance 2-fold in HCT116 human colon cancer cells, and induction was further enhanced (3-fold) by constitutively active STAT3 (STAT3-C), while strongly abrogated by dominant negative STAT3 (STAT3-DN). Attenuating PTTG expression by siRNA in STAT3 HCT116 stable transfectants suppressed cell growth and colony formation in vitro, and PTTG cell knockout also constrained activated STAT3-induced explanted murine tumor growth in vivo. STAT3 increased HCT116 cell migration and invasion up to 5-fold, whereas cell mobility was abolished by STAT3-DN (>85%). Impairing PTTG expression by siRNA also strongly suppressed STAT3-faciliated cell migration and invasion by up to 90%. Knocking out PTTG in STAT3-C HCT116 stable transfectants strongly decreased tumor metastases in nude mice, indicating the requirement of PTTG for STAT3-promoted metastasis. These results elucidate a mechanism for tumor cell PTTG regulation, whereby STAT3 induces PTTG expression to facilitate tumor growth and metastasis; and further support the rationale for targeting PTTG to abrogate colorectal cancer growth.
Collapse
|
42
|
The role of human pituitary transforming gene-1 in transcriptional and cytoskeletal regulation of cancer cells. Tzu Chi Med J 2012. [DOI: 10.1016/j.tcmj.2012.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
43
|
Karra H, Pitkänen R, Nykänen M, Talvinen K, Kuopio T, Söderström M, Kronqvist P. Securin predicts aneuploidy and survival in breast cancer. Histopathology 2012; 60:586-96. [DOI: 10.1111/j.1365-2559.2011.04107.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
44
|
Moreno-Mateos MA, Espina ÁG, Torres B, Gámez del Estal MM, Romero-Franco A, Ríos RM, Pintor-Toro JA. PTTG1/securin modulates microtubule nucleation and cell migration. Mol Biol Cell 2011; 22:4302-11. [PMID: 21937724 PMCID: PMC3216656 DOI: 10.1091/mbc.e10-10-0838] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PTTG1 is associated with the cis face of the Golgi apparatus and the centrosome, forming a complex with proteins involved in microtubule nucleation. PTTG1 depletion produces a delay in centrosomal and noncentrosomal microtubule nucleation and causes defects in both cell polarization and migration. Pituitary tumor transforming gene 1 (PTTG1), also known as securin, has been implicated in many biological functions, including inhibition of sister chromatid separation, DNA repair, organ development, and regulation of the expression and secretion of angiogenic and metastatic factors. Although most of these functions of securin seem to depend on the localization of PTTG1 in the nucleus of the cell, a fraction of the protein has been also detected in the cytoplasm. Here we demonstrate that, in different cell types, a portion of cytoplasmic PTTG1 is associated with the cis face of the Golgi apparatus and that this localization depends on PTTG1 phosphorylation status. In this organelle, PTTG1 forms a complex with proteins involved in microtubule nucleation, including GM130, AKAP450, and γ-tubulin. RNA interference–mediated depletion of PTTG1 produces a delay in centrosomal and noncentrosomal microtubule nucleation. Cells lacking PTTG1 show severe defects in both cell polarization and migration in wound-healing assays. To our knowledge, this is the first study reporting the role of PTTG1 in microtubule nucleation and cell polarization, two processes directly involved in cell migration. We believe that these findings will contribute to understanding the mechanisms underlying PTTG1-mediated biological functions.
Collapse
Affiliation(s)
- Miguel A Moreno-Mateos
- Centro Andaluz de Biología Molecular y Medicina Regenerativa, Consejo Superior de Investigaciones Científicas, 41092 Seville, Spain
| | | | | | | | | | | | | |
Collapse
|
45
|
Tong Y, Zhao W, Zhou C, Wawrowsky K, Melmed S. PTTG1 attenuates drug-induced cellular senescence. PLoS One 2011; 6:e23754. [PMID: 21858218 PMCID: PMC3157437 DOI: 10.1371/journal.pone.0023754] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 07/24/2011] [Indexed: 12/15/2022] Open
Abstract
As PTTG1 (pituitary tumor transforming gene) abundance correlates with adverse outcomes in cancer treatment, we determined mechanisms underlying this observation by assessing the role of PTTG1 in regulating cell response to anti-neoplastic drugs. HCT116 cells devoid of PTTG1 (PTTG1−/−) exhibited enhanced drug sensitivity as assessed by measuring BrdU incorporation in vitro. Apoptosis, mitosis catastrophe or DNA damage were not detected, but features of senescence were observed using low doses of doxorubicin and TSA. The number of drug-induced PTTG1−/− senescent cells increased ∼4 fold as compared to WT PTTG1-replete cells (p<0.001). p21, an important regulator of cell senescence, was induced ∼3 fold in HCT116 PTTG1−/− cells upon doxorubicin or Trichostatin A treatment. Binding of Sp1, p53 and p300 to the p21 promoter was enhanced in PTTG1−/− cells after treatment, suggesting transcriptional regulation of p21. p21 knock down abrogated the observed senescent effects of these drugs, indicating that PTTG1 likely suppresses p21 to regulate drug-induced senescence. PTTG1 also regulated SW620 colon cancer cells response to doxorubicin and TSA mediated by p21. Subcutaneously xenografted PTTG1−/− HCT116 cells developed smaller tumors and exhibited enhanced responses to doxorubicin. PTTG1−/− tumor tissue derived from excised tumors exhibited increased doxorubicin-induced senescence. As senescence is a determinant of cell responses to anti-neoplastic treatments, these findings suggest PTTG1 as a tumor cell marker to predict anti-neoplastic treatment outcomes.
Collapse
Affiliation(s)
- Yunguang Tong
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Weijiang Zhao
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Cuiqi Zhou
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kolja Wawrowsky
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shlomo Melmed
- Department of Medicine, Cedars-Sinai Medical Center, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
46
|
Targeting zebrafish and murine pituitary corticotroph tumors with a cyclin-dependent kinase (CDK) inhibitor. Proc Natl Acad Sci U S A 2011; 108:8414-9. [PMID: 21536883 DOI: 10.1073/pnas.1018091108] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cushing disease caused by adrenocorticotropin (ACTH)-secreting pituitary adenomas leads to hypercortisolemia predisposing to diabetes, hypertension, osteoporosis, central obesity, cardiovascular morbidity, and increased mortality. There is no effective pituitary targeted pharmacotherapy for Cushing disease. Here, we generated germline transgenic zebrafish with overexpression of pituitary tumor transforming gene (PTTG/securin) targeted to the adenohypophyseal proopiomelanocortin (POMC) lineage, which recapitulated early features pathognomonic of corticotroph adenomas, including corticotroph expansion and partial glucocorticoid resistance. Adult Tg:Pomc-Pttg fish develop neoplastic coticotrophs and pituitary cyclin E up-regulation, as well as metabolic disturbances mimicking hypercortisolism caused by Cushing disease. Early development of corticotroph pathologies in Tg:Pomc-Pttg embryos facilitated drug testing in vivo. We identified a pharmacologic CDK2/cyclin E inhibitor, R-roscovitine (seliciclib; CYC202), which specifically reversed corticotroph expansion in live Tg:Pomc-Pttg embryos. We further validated that orally administered R-roscovitine suppresses ACTH and corticosterone levels, and also restrained tumor growth in a mouse model of ACTH-secreting pituitary adenomas. Molecular analyses in vitro and in vivo showed that R-roscovitine suppresses ACTH expression, induces corticotroph tumor cell senescence and cell cycle exit by up-regulating p27, p21 and p57, and downregulates cyclin E expression. The results suggest that use of selective CDK inhibitors could effectively target corticotroph tumor growth and hormone secretion.
Collapse
|
47
|
Sylvestre EL, Pennetier S, Bureau M, Robert C, Sirard MA. Investigating the potential of genes preferentially expressed in oocyte to induce chromatin remodeling in somatic cells. Cell Reprogram 2011; 12:519-28. [PMID: 20936903 DOI: 10.1089/cell.2010.0012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The oocyte capacity to rejuvenate a differentiated nucleus to restart the proper embryonic program has been highly conserved between vertebrate species. In view of the recent progress to induce pluripotency in somatic cells with stemness genes, we investigated the potential of oocyte genes to contribute to chromatin rearrangements in somatic cells. We selected conserved genes that are naturally expressed mainly in oocytes and that were susceptible to play a role in reprogramming during early embryogenesis. We induced their expression by transient transfection in HEK293 cells. We then assessed whether they had a global impact on epigenetic events such as histone core modifications, and also on transcription and expression of pluripotency-associated transcription factors. Nucleoplasmin 2 (NPM2), activation-induced cytidine deaminase (AICDA), and Geminin (GMNN) overexpression induced differences in histone core modifications (methylation and acetylation). AICDA and NPM2 also influenced RNA neosynthesis. NPM2, GMNN, and STELLA induced overexpression of well-known pluripotency transcription factors. Overall, AICDA, GMNN, NPM2, and STELLA influenced at least one of the aspects analyzed. Their potential could be useful in increasing the cell receptivity to pluripotency induction.
Collapse
|
48
|
Chintharlapalli S, Papineni S, Lee SO, Lei P, Jin UH, Sherman SI, Santarpia L, Safe S. Inhibition of pituitary tumor-transforming gene-1 in thyroid cancer cells by drugs that decrease specificity proteins. Mol Carcinog 2011; 50:655-67. [PMID: 21268135 DOI: 10.1002/mc.20738] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 11/22/2010] [Accepted: 12/14/2010] [Indexed: 02/06/2023]
Abstract
Methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate (CDODA-Me) and the corresponding 2-trifluoromethyl analog (CF(3)DODA-Me) are derived synthetically from the triterpenoid glycyrrhetinic acid, a major component of licorice. CDODA-Me and CF(3)DODA-Me inhibited growth of highly invasive ARO, DRO, K-18, and HTh-74 thyroid cancer cells and this was due, in part, to decreased expression of specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4 that are overexpressed in these cells. CDODA-Me and CF(3)DODA-Me also decreased expression of Sp-dependent genes, such as survivin and vascular endothelial growth factor (VEGF), and induced apoptosis. In addition, pituitary tumor-transforming gene-1 (PTTG-1) protein and mRNA levels were also decreased in thyroid cancer cells treated with CDODA-Me or CF(3)DODA-Me and this was accompanied by decreased expression of PTTG-1-dependent c-Myc and fibroblast growth factor-2 (FGF-2) genes. RNA interference studies against Sp1, Sp3, and Sp4 proteins showed that in thyroid cancer cells, PTTG-1 was an Sp-dependent gene. This study demonstrates for the first time that drugs, such as CDODA-Me and CF(3)DODA-Me, that decrease Sp protein expression also downregulate PTTG-1 in thyroid cancer cells and therefore have potential for clinical treatment of thyroid cancer and other endocrine neoplasias where PTTG-1 is a major pro-oncogenic factor.
Collapse
|
49
|
Abstract
The pituitary tumor-transforming gene (PTTG1) encodes a multifunctional protein (PTTG) that is overexpressed in numerous tumours, including pituitary, thyroid, breast and ovarian carcinomas. PTTG induces cellular transformation in vitro and tumourigenesis in vivo, and several mechanisms by which PTTG contributes to tumourigenesis have been investigated. Also known as the human securin, PTTG is involved in cell cycle regulation, controlling the segregation of sister chromatids during mitosis. This review outlines current information regarding PTTG structure, expression, regulation and function in the pathogenesis of neoplasia. Recent progress concerning the use of PTTG as a prognostic marker or therapeutic target will be considered. In addition, the PTTG binding factor (PBF), identified through its interaction with PTTG, has also been established as a proto-oncogene that is upregulated in several cancers. Current knowledge regarding PBF is outlined and its role both independently and alongside PTTG in endocrine and related cancers is discussed.
Collapse
|
50
|
Feeney KM, Wasson CW, Parish JL. Cohesin: a regulator of genome integrity and gene expression. Biochem J 2010; 428:147-61. [PMID: 20462401 DOI: 10.1042/bj20100151] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Following DNA replication, chromatid pairs are held together by a proteinacious complex called cohesin until separation during the metaphase-to-anaphase transition. Accurate segregation is achieved by regulation of both sister chromatid cohesion establishment and removal, mediated by post-translational modification of cohesin and interaction with numerous accessory proteins. Recent evidence has led to the conclusion that cohesin is also vitally important in the repair of DNA lesions and control of gene expression. It is now clear that chromosome segregation is not the only important function of cohesin in the maintenance of genome integrity.
Collapse
Affiliation(s)
- Katherine M Feeney
- Bute Medical School, University of St Andrews, St Andrews, Fife KY16 9TS, Scotland, U.K
| | | | | |
Collapse
|