1
|
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
|
2
|
aarF domain containing kinase 5 gene promotes invasion and migration of lung cancer cells through ADCK5-SOX9-PTTG1 pathway. Exp Cell Res 2020; 392:112002. [PMID: 32277958 DOI: 10.1016/j.yexcr.2020.112002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 02/08/2023]
Abstract
AarF domain containing kinase 5 (ADCK5) is a member of an atypical kinase family and overexpressed in many carcinomas including lung cancer, while the function of this protein has not been elucidated. Here we investigated the mechanism of ADCK5 involved in regulating invasion and migration of lung cancer cells, and showed that ADCK5 might regulate the expression of tumor oncogene human pituitary tumor transforming gene-1 (PTTG1) by phosphorylating transcription factor SOX9, therefore enhancing the migration and invasion capabilities of lung cancer cells. Mutagenesis of potential serine phosphorylation sites on SOX9 indicated that serine 181 might be required to maintain transcription activation of SOX9 as well as increase PTTG1 levels. The serine 181 site of SOX9 is in a motif that is targeted by ADCK5. The ADCK5-SOX9-PTTG1 pathway might be a potential therapeutic target for lung cancer.
Collapse
|
3
|
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
|
4
|
Gounden V, Rampursat YD, Jialal I. Secretory tumors of the pituitary gland: a clinical biochemistry perspective. ACTA ACUST UNITED AC 2018; 57:150-164. [DOI: 10.1515/cclm-2018-0552] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
Abstract
The pituitary gland is responsible for the production and/or secretion of various hormones that play a vital role in regulating endocrine function within the body. Secretory tumors of the anterior pituitary predominantly, pituitary adenomas, collectively account for 10%–25% of central nervous system tumors requiring surgical treatment. The most common secretory tumors are prolactinomas, which can be diagnosed by basal prolactin levels. Acromegaly can be diagnosed by basal insulin growth-like factor 1 levels and the failure of growth hormone (GH) to suppress during an oral glucose tolerance test. Cushing disease can be diagnosed by demonstrating hypercortisolemia evidenced by increased salivary cortisol levels in the evening, increased urine free cortisol excretion and failure of plasma cortisol to suppress following oral dexamethasone given overnight (1.0 mg). We also discuss the diagnosis of the rarer thyroid-stimulating hormone and gonadotrophin secretory tumors. Morbidity is associated with tumor occurrence, clinical sequelae as well as the related medical, surgical and radiological management. This review focuses on the pathogenesis of secretory tumors of the anterior pituitary with emphasis on molecular mechanisms associated with tumorigenesis and the major role of the clinical chemistry laboratory in diagnosis and management of these tumors.
Collapse
Affiliation(s)
- Verena Gounden
- Department of Chemical Pathology , University of KwaZulu Natal and National Health Laboratory Services, Inkosi Albert Luthuli Central Hospital , Durban , South Africa
| | - Yashna D. Rampursat
- Department of Chemical Pathology , University of KwaZulu Natal and National Health Laboratory Services, Inkosi Albert Luthuli Central Hospital , Durban , South Africa
| | - Ishwarlal Jialal
- California North-State University, College of Medicine , Elk Grove, CA 95757 , USA
- Director, Section of Clinical Chemistry, VA Medical Center , Sacramento, CA , USA
| |
Collapse
|
5
|
Ma K, Ma L, Jian Z. Pituitary tumor-transforming 1 expression in laryngeal cancer and its association with prognosis. Oncol Lett 2018; 16:1107-1114. [PMID: 30061937 DOI: 10.3892/ol.2018.8745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 02/12/2018] [Indexed: 01/08/2023] Open
Abstract
The purpose of the present study was to investigate the association between the expression of pituitary tumor-transforming 1 (PTTG1) and the expression of matrix metalloproteinase (MMP)-2 and MMP-9 in laryngeal carcinoma tissues, and to elucidate the association between PTTG1 expression and the prognosis of patients with laryngeal cancer. Immunohistochemical staining was used to detect PTTG1 expression in laryngeal cancer and normal tumor-adjacent laryngeal tissues. Western blotting was used to determine the levels of PTTG1 and MMP-2 and -9 in laryngeal carcinoma tissues and to assess their correlation. In addition, the associations between PTTG1 expression and the clinical parameters of laryngeal cancer and patient survival were determined. The immunohistochemistry results revealed that the positive expression rates of PTTG1, MMP-2 and MMP-9 in the laryngeal cancer tissues were significantly higher than those in the carcinoma-adjacent normal laryngeal tissues (all P<0.05). In addition, expression levels of PTTG1, MMP-2 and MMP-9 were significantly associated with lymph node metastasis, histological grade and clinical stage (P<0.05). Furthermore, the levels of PTTG1 were positively correlated with the levels of MMP-2 and MMP-9 in laryngeal cancer tissues (P<0.05). In summary, the expression levels of PTTG1, MMP-2 and MMP-9 are closely associated with the biological behaviors of laryngeal cancer tissues, showing that they serve important roles in the occurrence and development of laryngeal cancer, and may be useful as biological indicators of laryngeal tissue invasion, metastasis and patient prognosis.
Collapse
Affiliation(s)
- Kunpeng Ma
- Department of Otorhinolaryngology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Limin Ma
- Department of Plastic Surgery, Plastic Surgery Hospital of Weifang Medical University, Weifang, Shandong 261041, P.R. China
| | - Zhaocheng Jian
- Department of Vascular Interventional Radiology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| |
Collapse
|
6
|
Yan T, Shi X, Fu J. Identification of peptide-mediated interactions between human PTTG and SH3 domains in pALL gene expression profile. J Mol Graph Model 2017; 76:11-16. [PMID: 28667917 DOI: 10.1016/j.jmgm.2017.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 11/17/2022]
Abstract
Human pituitary tumor-transforming gene (PTTG) plays an essential role in the development and progression of pediatric acute lymphoblastic leukemia (pALL). PTTG has two SH3-binding peptide motifs that can be recognized by a variety of SH3-containing proteins in the pALL through peptide-mediated interactions. In this study, the gene expression profile of pALL was examined in detail by integrating computational modeling and experimental assay, aiming to identify those potential partner proteins of human PTTG. The binding potency of domain candidates to peptide motifs was ranked using knowledge-based scoring and fluorescence titration. A number of SH3 domains found in a variety of pALL proteins were identified as potent binders with moderate or high affinity for PTTG. It is revealed that the PTTG peptide motifs show different affinity profiles for various candidate proteins, indicating that the PTTG selectivity is optimized across pALL gene expression space. The PTTG peptides were then mutated rationally to target the SH3 domains of identified partner proteins by competing with the native peptide motifs.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Pediatrics, The Second People's Hospital of Huai'an, Huai'an 223002, PR China.
| | - Xiangxiang Shi
- Department of Pediatrics, The Second People's Hospital of Huai'an, Huai'an 223002, PR China
| | - Jing Fu
- ICU, The Second People's Hospital of Huai'an, Huai'an 223002, PR China
| |
Collapse
|
7
|
Liu J, Wang D, Li Y, Yao H, Zhang N, Zhang X, Zhong F, Huang Y. Integrated In Silico-In Vitro Identification and Characterization of the SH3-Mediated Interaction between Human PTTG and its Cognate Partners in Medulloblastoma. Cell Biochem Biophys 2017. [PMID: 28646413 DOI: 10.1007/s12013-017-0810-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The human pituitary tumor-transforming gene is an oncogenic protein which serves as a central hub in the cellular signaling network of medulloblastoma. The protein contains two vicinal PxxP motifs at its C terminus that are potential binding sites of peptide-recognition SH3 domains. Here, a synthetic protocol that integrated in silico analysis and in vitro assay was described to identify the SH3-binding partners of pituitary tumor-transforming gene in the gene expression profile of medulloblastoma. In the procedure, a variety of structurally diverse, non-redundant SH3 domains with high gene expression in medulloblastoma were compiled, and their three-dimensional structures were either manually retrieved from the protein data bank database or computationally modeled through bioinformatics technique. The binding capability of these domains towards the two PxxP-containing peptides m1p: 161LGPPSPVK168 and m2p: 168KMPSPPWE175 of pituitary tumor-transforming gene were ranked by structure-based scoring and fluorescence-based assay. Consequently, a number of SH3 domains, including MAP3K and PI3K, were found to have moderate or high affinity for m1p and/or m2p. Interestingly, the two overlapping peptides exhibits a distinct binding profile to these identified domain partners, suggesting that the binding selectivity of m1p and m2p is optimized across the medulloblastoma expression spectrum by competing for domain candidates. In addition, two redesigned versions of m1p peptide ware obtained via a structure-based rational mutation approach, which exhibited an increased affinity for the domain as compared to native peptide.
Collapse
Affiliation(s)
- Jiangang Liu
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Dapeng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yanyan Li
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hui Yao
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Nan Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xuewen Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fangping Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yulun Huang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
8
|
Su X, Chen J, Ni L, Shi W, Shi J, Liu X, Zhang Y, Gong P, Zhu H, Huang Q. Inhibition of PTTG1 expression by microRNA suppresses proliferation and induces apoptosis of malignant glioma cells. Oncol Lett 2016; 12:3463-3471. [PMID: 27900021 DOI: 10.3892/ol.2016.5035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 02/19/2016] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the role of pituitary tumor-transforming gene 1 (PTTG1) in the proliferation, invasion and apoptosis of human malignant glioma U251 cells. Firstly, 2 microRNAs (miRNAs) targeting PTTG1 messenger (m)RNA were ligated into a pcDNA6.2-GW/EmGFP-miR expression vector. The recombinant plasmids, miRNA-1 and miRNA-2 (miR-2), were transfected into U251 cells using the liposome method. PTTG1 mRNA and protein levels were evaluated using quantitative polymerase chain reaction and western blot analysis. The proliferation and invasion abilities of U251 cells were determined using methylthiazol tetrazolium and Matrigel assays. Flow cytometry analysis with Annexin V/propidium iodide double staining was used to determine the percentage of apoptotic cells. PTTG1 expression was effectively suppressed by miR-2. U251 cell growth was inhibited between 10.7 and 34.7% in the miR-2 group compared with the blank group. The Matrigel assay demonstrated that the percentage of infiltrating U251 cells was significantly lower in the miR-2 group (12.3±1.0%) compared to the blank group (24.7±1.4%; P<0.001) and the negative control group (24.0±2.0%; P<0.05). A higher percentage of apoptotic U251 cells were observed in the miR-2 group compared with the blank group (53.6 vs. 32.4%) using flow cytometry due to cycle arrests at the G2/M phase. The miR-2-transfected U251 cells were subcutaneously injected into nude mice, and these mice possessed a decreased tumor tissue growth rate and higher percentage of apoptotic cells compared with the blank and negative control groups. In conclusion, PTTG1 gene expression in human malignant glioma U251 cells was effectively suppressed by exogenous miR-2. The downregulation of PTTG1 induced glioma cell apoptosis and cell cycle arrest at the G2/M phase, which inhibited cell proliferation, reverse invasion and infiltration of glioma cells.
Collapse
Affiliation(s)
- Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianguo Chen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lanchun Ni
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wei Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jinlong Shi
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaojiang Liu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yu Zhang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Peipei Gong
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hui Zhu
- Comprehensive Surgical Laboratory, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qingfeng Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
9
|
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
|
10
|
Prognostic implications of securin expression and sub-cellular localization in human breast cancer. Cell Oncol (Dordr) 2016; 39:319-31. [DOI: 10.1007/s13402-016-0277-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2016] [Indexed: 02/06/2023] Open
|
11
|
Pituitary tumor-transforming gene-1 serves as an independent prognostic biomarker for gastric cancer. Gastric Cancer 2016; 19:107-15. [PMID: 25627474 DOI: 10.1007/s10120-015-0459-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/29/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pituitary tumor-transforming gene-1 (PTTG1) is a transcription factor that can affect transcriptional activity, angiogenesis, and cell senescence. We examined PTTG1 mRNA and protein expression in gastric cancer (GC) cell lines and tissues to determine its value as a biomarker for GC diagnosis and therapy. METHODS PTTG1 mRNA expression from 78 GC cases and paired adjacent normal mucosa (PCR cohort) as well as from five gastric cell lines was assessed using qRT-PCR. Nuclear and cytoplasmic RNA were extracted from two gastric cell lines to determine PTTG1 mRNA localization. PTTG1 protein expression from 98 GC cases, their paired adjacent normal mucosa, and 23 gastric intraepithelial neoplasia (GIN) cases was examined using immunohistochemistry (IHC cohort). The correlation between PTTG1 mRNA and protein expression and GC clinicopathological parameters was analyzed. RESULTS PTTG1 mRNA expression in GC tissues and cell lines was significantly increased compared with adjacent normal gastric mucosa and normal gastric mucous cell lines (p < 0.05). PTTG1 expression was nuclear and cytoplasmic, with higher cytoplasmic expression. PTTG1 immunostaining significantly differed in GC (95.66 ± 20.65), GIN (84.00 ± 34.16), and normal adjacent mucosa (28 ± 22.25) (p < 0.001). Multivariate Cox regression analysis revealed that PTTG1 mRNA and protein expression are independent prognostic factors for GC patient survival. CONCLUSION Our results suggest that PTTG1 is a promising target for GC diagnosis and therapy.
Collapse
|
12
|
Wondergem B, Zhang Z, Huang D, Ong CK, Koeman J, Hof DV, Petillo D, Ooi A, Anema J, Lane B, Kahnoski RJ, Furge KA, Teh BT. Expression of the PTTG1 Oncogene Is Associated with Aggressive Clear Cell Renal Cell Carcinoma. Cancer Res 2012; 72:4361-71. [DOI: 10.1158/0008-5472.can-11-2330] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Tseng HH, Chuah QY, Yang PM, Chen CT, Chao JC, Lin MD, Chiu SJ. Securin enhances the anti-cancer effects of 6-methoxy-3-(3',4',5'-trimethoxy-benzoyl)-1H-indole (BPR0L075) in human colorectal cancer cells. PLoS One 2012; 7:e36006. [PMID: 22563433 PMCID: PMC3338557 DOI: 10.1371/journal.pone.0036006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 03/29/2012] [Indexed: 12/15/2022] Open
Abstract
BPR0L075 [6-methoxy-3-(3′,4′,5′-trimethoxy-benzoyl)-1H-indole] is a novel anti-microtubule drug with anti-tumor and anti-angiogenic activities in vitro and in vivo. Securin is required for genome stability, and is expressed abundantly in most cancer cells, promoting cell proliferation and tumorigenesis. In this study, we found that BPR0L075 efficiently induced cell death of HCT116 human colorectal cancer cells that have higher expression levels of securin. The cytotoxicity of BPR0L075 was attenuated in isogenic securin-null HCT116 cells. BPR0L075 induced DNA damage response, G2/M arrest, and activation of the spindle assembly checkpoint in HCT116 cells. Interestingly, BPR0L075 induced phosphorylation of securin. BPR0L075 withdrawal resulted in degradation of securin, mitotic exit, and mitotic catastrophe, which were attenuated in securin-null cells. Inhibition of cdc2 decreased securin phosphorylation, G2/M arrest and cell death induced by BPR0L075. Moreover, BPR0L075 caused cell death through a caspase-independent mechanism and activation of JNK and p38 MAPK pathways. These findings provided evidence for the first time that BPR0L075 treatment is beneficial for the treatment of human colorectal tumors with higher levels of securin. Thus, we suggest that the expression levels of securin may be a predictive factor for application in anti-cancer therapy with BPR0L075 in human cancer cells.
Collapse
Affiliation(s)
- Ho-Hsing Tseng
- Department of Life Science, Tzu Chi University, Hualien, Taiwan R.O.C.
| | - Qiu-Yu Chuah
- Department of Life Science, Tzu Chi University, Hualien, Taiwan R.O.C.
| | - Pei-Ming Yang
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan R.O.C.
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan R.O.C.
| | - Jung-Chi Chao
- Department of Life Science, Tzu Chi University, Hualien, Taiwan R.O.C.
| | - Ming-Der Lin
- Department of Molecular Biology and Human Genetic, Tzu Chi University, Hualien, Taiwan R.O.C.
| | - Shu-Jun Chiu
- Department of Life Science, Tzu Chi University, Hualien, Taiwan R.O.C.
- * E-mail:
| |
Collapse
|
14
|
Lewy GD, Sharma N, Seed RI, Smith VE, Boelaert K, McCabe CJ. The pituitary tumor transforming gene in thyroid cancer. J Endocrinol Invest 2012; 35:425-33. [PMID: 22522436 DOI: 10.3275/8332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The pituitary tumor transforming gene (PTTG) is a multifunctional proto-oncogene that is over-expressed in various tumors including thyroid carcinomas, where it is a prognostic indicator of tumor recurrence. PTTG has potent transforming capabilities in vitro and in vivo, and many studies have investigated the potential mechanisms by which PTTG contributes to tumorigenesis. As the human securin, PTTG is involved in critical mechanisms of cell cycle regulation, whereby aberrant expression induces aneuploidy. PTTG may further contribute to tumorigenesis through its role in DNA damage response pathways and via complex interactions with hormones and growth factors. Furthermore, PTTG over-expression negatively impacts upon the efficacy of radioiodine therapy in thyroid cancer, through repression of expression and function of the sodium iodide symporter. Given its various roles at all disease stages, PTTG appears to be an important oncogene in thyroid cancer. This review discusses the current knowledge of PTTG with particular focus on its role in thyroid cancer.
Collapse
Affiliation(s)
- G D Lewy
- School of Clinical and Experimental Medicine, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | | | | | | | | | | |
Collapse
|
15
|
Lai PC, Fang TC, Chiu TH, Huang YT. Overexpression of Securin in Human Transitional Cell Carcinoma Specimens. Tzu Chi Med J 2010. [DOI: 10.1016/s1016-3190(10)60067-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
16
|
|
17
|
Sánchez-Ortiga R, Sánchez Tejada L, Peiró Cabrera G, Moreno-Pérez O, Arias Mendoza N, Ignacio Aranda López F, Picó Alfonso A. Papel de pituitary tumour-transforming gene (PTTG) en los adenomas hipofisarios. ACTA ACUST UNITED AC 2010; 57:28-34. [DOI: 10.1016/s1575-0922(10)70006-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 12/10/2009] [Indexed: 01/23/2023]
|
18
|
Abstract
In pituitary tumorigenesis there is cross-talk between fine deregulation of intracellular pathways and complex microenvironmental factors, processes that can be modulated at various levels. The signaling pathways of growth, angiogenic factors and hormones are intricate; therefore, alterations induced upon node-molecules can lead to aberrant proliferation. The demonstrated overactivity of AKT and MAPK pathways qualifies them as valuable targets for inhibition mediated by somatostatin analogues. An increasing body of evidence suggests clinically significant implications of PTTG1 in correlation with aggressive phenotypes or survival rate, thus PTTG1 is an interesting candidate biomarker for malignancy, tumor staging and subsequent therapeutic interventions. Future work should focus on understanding the molecular mechanisms that control pituitary tumor transformation, where intracellular signaling molecules will constitute not only diagnostic/prognostic markers but also novel therapeutic targets.
Collapse
Affiliation(s)
- Cristiana Pistol Tanase
- Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, Sector 5, Bucharest, Romania.
| | | | | |
Collapse
|
19
|
Yan S, Zhou C, Lou X, Xiao Z, Zhu H, Wang Q, Wang Y, Lu N, He S, Zhan Q, Liu S, Xu N. PTTG Overexpression Promotes Lymph Node Metastasis in Human Esophageal Squamous Cell Carcinoma. Cancer Res 2009; 69:3283-90. [DOI: 10.1158/0008-5472.can-08-0367] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Morris MA, Dawson CW, Wei W, O'Neil JD, Stewart SE, Jia J, Bell AI, Young LS, Arrand JR. Epstein–Barr virus-encoded LMP1 induces a hyperproliferative and inflammatory gene expression programme in cultured keratinocytes. J Gen Virol 2008; 89:2806-2820. [DOI: 10.1099/vir.0.2008/003970-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SCC12F cells are a line of keratinocytes that retain the capacity for terminal differentiation in vitro. We showed previously that the Epstein–Barr virus (EBV)-encoded oncogene latent membrane protein 1 (LMP1) altered SCC12F morphology in vitro, downregulated cell–cell-adhesion molecule expression and promoted cell motility. In organotypic raft culture, LMP1-expressing cells failed to stratify and formed poorly organized structures which displayed impaired terminal differentiation. To understand better the mechanism(s) by which LMP1 induces these effects, we generated SCC12F cells in which LMP1 expression is inducible. Following induction, these cells exhibited phenotypic changes similar to those observed previously and allowed us to investigate the effects of LMP1 expression on cellular pathways associated with growth, differentiation and morphology. Using microarrays and a number of confirmatory techniques, we identified sets of differentially expressed genes that are characteristically expressed in inflammatory and hyperproliferative epidermis, including chemokines, cytokines and their receptors, growth factors involved in promoting epithelial cell motility and proliferation and signalling molecules that regulate actin filament reorganization and cell movement. Among the genes whose expression was differentially induced significantly by LMP1, the induction of IL-1β and IL-1α was of particular interest, as many of the LMP1-regulated genes identified are established targets of these cytokines. Our findings suggest that alterations in the IL-1 signalling network may be responsible for many of the changes in host-cell gene expression induced in response to LMP1. Identification of these LMP1-regulated genes helps to define the mechanism(s) by which this oncoprotein influences cellular pathways that regulate terminal differentiation, cell motility and inflammation.
Collapse
Affiliation(s)
- Mhairi A. Morris
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Christopher W. Dawson
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Wenbin Wei
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - John D. O'Neil
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Suzanne E. Stewart
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Junying Jia
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Andrew I. Bell
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - Lawrence S. Young
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| | - John R. Arrand
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
21
|
Kim JW, Song JY, Lee JM, Lee JK, Lee NW, Yeom BW, Lee KW. Expression of pituitary tumor-transforming gene in endometrial cancer as a prognostic marker. Int J Gynecol Cancer 2008; 18:1352-9. [DOI: 10.1111/j.1525-1438.2007.01168.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The pituitary tumor-transforming gene (PTTG) is a novel oncogene expressed abundantly in most tumors, regulates basic fibroblast growth factor secretion, and induces angiogenesis. The objective of this study is to compare the expression rate of PTTG in endometrial cells, to correlate the level of expression of PTTG with the clinicopathologic parameters and overall survival, and to evaluate the possible use of PTTG as a prognostic marker of endometrial cancer. Forty patients diagnosed with endometrial cancer, 20 patients with endometrial hyperplasia, and 20 patients with normal endometrial tissues were included in the study. Immunohistochemical analyses on paraffin-embedded blocks were performed using a polyclonal anti-PTTG antibody. The decrease in expression of cytoplasmic and nuclear PTTG seen for endometrial cancer cells was statistically significant (P< 0.05). Cytoplasmic PTTG expression correlated with expression of progesterone receptor (P= 0.009) and FGF-2 (P= 0.007) but not with other parameters such as the expression of estrogen receptor, tumor grade, and surgical stage. Nuclear PTTG expression did not correlate with any parameters. The mean survival of patients with positive and negative cytoplasmic PTTG expression was 40.8 and 48.6 months (P= 0.78). In nuclear PTTG expression, the survival was 20.0 and 51.8 months, respectively (P= 0.04). Cytoplasmic PTTG expression was not associated with survival. Patients with nuclear PTTG overexpression showed a significant decrease in survival. The use of PTTG as a prognostic marker for endometrial cancer needs further investigation.
Collapse
|
22
|
Tong Y, Ben-Shlomo A, Zhou C, Wawrowsky K, Melmed S. Pituitary tumor transforming gene 1 regulates Aurora kinase A activity. Oncogene 2008; 27:6385-95. [PMID: 18663361 DOI: 10.1038/onc.2008.234] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pituitary tumor transforming gene 1 (PTTG1), a transforming gene highly expressed in several cancers, is a mammalian securin protein regulating both G1/S and G2/M phases. Using protein array screening, we showed PTTG1 interacting with Aurora kinase A (Aurora-A), and confirmed the interaction using co-immunoprecipitation, His-tagged pull-down assays and intracellular immunofluorescence colocalization. PTTG1 transfection into HCT116 cells prevented Aurora-A T288 autophosphorylation, inhibited phosphorylation of the histone H3 Aurora-A substrate and resulted in abnormally condensed chromatin. PTTG1-null cell proliferation was more sensitive to Aurora-A knock down and to Aurora kinase Inhibitor III treatment. The results indicate that PTTG1 and Aurora-A interact to regulate cellular responses to anti-neoplastic drugs. PTTG1 knockdown is therefore a potential approach to improve the efficacy of tumor Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Y Tong
- Department of Medicine, Cedars-Sinai Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90048, USA
| | | | | | | | | |
Collapse
|
23
|
Abstract
Pituitary tumor-transforming gene-1 (PTTG1) is overexpressed in a variety of endocrine-related tumors, especially pituitary, thyroid, breast, ovarian, and uterine tumors, as well as nonendocrine-related cancers involving the central nervous, pulmonary, and gastrointestinal systems. Forced PTTG1 expression induces cell transformation in vitro and tumor formation in nude mice. In some tumors, high PTTG1 levels correlate with invasiveness, and PTTG1 has been identified as a key signature gene associated with tumor metastasis. Increasing evidence supports a multifunctional role of PTTG1 in cell physiology and tumorigenesis. Physiological PTTG1 properties include securin activity, DNA damage/repair regulation and involvement in organ development and metabolism. Tumorigenic mechanisms for PTTG1 action involve cell transformation and aneuploidy, apoptosis, and tumorigenic microenvironment feedback. This paper reviews recent advances in our understanding of PTTG1 structure and regulation and addresses known mechanisms of PTTG1 action. Recent knowledge gained from PTTG1-null mouse models and transgenic animals and their potential application to subcellular therapeutic targeting PTTG1 are discussed.
Collapse
Affiliation(s)
- George Vlotides
- Department of Medicine, Cedars-Sinai Medical Center, University of California School of Medicine, Los Angeles, California 90048, USA
| | | | | |
Collapse
|
24
|
Tong Y, Tan Y, Zhou C, Melmed S. Pituitary tumor transforming gene interacts with Sp1 to modulate G1/S cell phase transition. Oncogene 2007; 26:5596-605. [PMID: 17353909 PMCID: PMC2736684 DOI: 10.1038/sj.onc.1210339] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pituitary tumor transforming gene (PTTG1) was isolated from rat pituitary tumor cells, and subsequently identified as a securin protein as well as a transcription factor. We show here a global transcriptional effect of PTTG1 in human choriocarcinoma JEG-3 cells by simultaneously assessing 20,000 gene promoters using chromatin immunoprecipitation (ChIP)-on-Chip experiments. Seven hundred and forty-six gene promoters (P<0.001) were found enriched, with functions relating to cell cycle, metabolic control and signal transduction. Significant interaction between PTTG1 and Sp1 (P<0.000001) was found by transcriptional pattern analysis of ChIP data and further confirmed by immunoprecipitation and pull-down assays. PTTG1 acts coordinately with Sp1 to induce cyclin D3 expression approximately threefold, and promotes G1/S-phase transition independently of p21. PTTG1 deletion was also protective for anchorage-independent cell colony formation. The results show that PTTG1 exhibits properties of a global transcription factor, and specifically modulates the G1/S-phase transition by interacting with Sp1. This novel signaling pathway may be required for PTTG1 transforming activity.
Collapse
Affiliation(s)
- Y Tong
- Department of Medicine, Cedars-Sinai Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
25
|
Boelaert K, Smith VE, Stratford AL, Kogai T, Tannahill LA, Watkinson JC, Eggo MC, Franklyn JA, McCabe CJ. PTTG and PBF repress the human sodium iodide symporter. Oncogene 2007; 26:4344-56. [PMID: 17297475 DOI: 10.1038/sj.onc.1210221] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The ability of the thyroid to accumulate iodide provides the basis for radioiodine ablation of differentiated thyroid cancers and their metastases. Most thyroid tumours exhibit reduced iodide uptake, although the mechanisms accounting for this remain poorly understood. Pituitary tumour transforming gene (PTTG) is a proto-oncogene implicated in the pathogenesis of thyroid tumours. We now show that PTTG and its binding factor PBF repress expression of sodium iodide symporter (NIS) messenger RNA (mRNA), and inhibit iodide uptake. This process is mediated at least in part through fibroblast growth factor-2. In detailed studies of the NIS promoter in rat FRTL-5 cells, PTTG and PBF demonstrated specific inhibition of promoter activity via the human upstream enhancer element (hNUE). Within this approximately 1 kb element, a complex PAX8-upstream stimulating factor 1 (USF1) response element proved critical both to basal promoter activity and to PTTG and PBF repression of NIS. In particular, repression by PTTG was contingent upon the USF1, but not the PAX8, site. Finally, in human primary thyroid cells, PTTG and PBF similarly repressed the NIS promoter via hNUE. Taken together, our data suggest that the reported overexpression of PTTG and PBF in differentiated thyroid cancer has profound implications for activity of the NIS gene, and hence significantly impacts upon the efficacy of radioiodine treatment.
Collapse
Affiliation(s)
- K Boelaert
- Department of Medicine, Division of Medical Sciences, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Malik MT, Kakar SS. Regulation of angiogenesis and invasion by human Pituitary tumor transforming gene (PTTG) through increased expression and secretion of matrix metalloproteinase-2 (MMP-2). Mol Cancer 2006; 5:61. [PMID: 17096843 PMCID: PMC1654177 DOI: 10.1186/1476-4598-5-61] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 11/10/2006] [Indexed: 11/24/2022] Open
Abstract
Background Pituitary tumor transforming gene (PTTG) is a novel oncogene that is expressed at higher level in most of the tumors analyzed to date compared to normal tissues. Existence of a relationship between PTTG levels and tumor angiogenesis and metastasis has been reported. However, the mechanisms by which PTTG achieve these functions remain unknown. In the present study, we investigated the effect of overexpression of PTTG on secretion and expression of metastasis-related metalloproteinase-2 (MMP-2) in HEK293 cells, cell migration, invasion and tubule formation. Results Transient or stable transfection of HEK293 cells with PTTG cDNA showed a significant increase in secretion and expression of MMP-2 measured by zymography, reverse transcriptase (RT/PCR), ELISA, and MMP-2 gene promoter activity. Furthermore, in our studies, we showed that tumor developed in nude mice on injection of HEK293 cells that constitutively express PTTG expressed high levels of both MMP-2 mRNA and protein, and MMP-2 activity. Conditioned medium collected from the HEK293 cells overexpressing PTTG showed a significant increase in cell migration, invasion and tubule formation of human umbilical vein endothelial cells (HUVEC). Pretreatment of conditioned medium with MMP-2-specific antibody significantly decreased these effects, suggesting that PTTG may contribute to tumor angiogenesis and metastasis via activation of proteolysis and increase in invasion through modulation of MMP-2 activity and expression. Conclusion Our results provide novel information that PTTG contributes to cell migration, invasion and angiogenesis by induction of MMP-2 secretion and expression. Furthermore, we showed that tumors developed in nude mice on injection of HEK293 cells that constitutively express PTTG induce expression of MMP-2 and significantly increase its functional activity, suggesting a relationship between PTTG levels and MMP-2 which may play a critical role in regulation of tumor growth, angiogenesis and metastasis. Blocking of function of PTTG or down regulation of its expression in tumors may result in suppression of tumor growth and metastasis, through the down regulation of MMP-2 expression and activity. To our knowledge, this study is the first study demonstrating the modulation of MMP-2 expression and biological activity by PTTG.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Line
- Cell Movement/genetics
- Cell Movement/physiology
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Luciferases/genetics
- Luciferases/metabolism
- Matrix Metalloproteinase 2/genetics
- Matrix Metalloproteinase 2/metabolism
- Mice
- Mice, Nude
- Neoplasm Invasiveness
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplasm Proteins/physiology
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/physiopathology
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Securin
- Transfection
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Mohammad T Malik
- Department of Biochemistry and Molecular Biology, University of Louisville, Louisville, KY, USA
| | - Sham S Kakar
- Department of Medicine and James Graham Brown cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
27
|
Solbach C, Roller M, Eckerdt F, Peters S, Knecht R. Pituitary tumor-transforming gene expression is a prognostic marker for tumor recurrence in squamous cell carcinoma of the head and neck. BMC Cancer 2006; 6:242. [PMID: 17029632 PMCID: PMC1613251 DOI: 10.1186/1471-2407-6-242] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Accepted: 10/09/2006] [Indexed: 12/03/2022] Open
Abstract
Background The proto-oncogene pituitary tumor-transforming gene (PTTG) has been shown to be abundantly overexpressed in a large variety of neoplasms likely promoting neo-vascularization and tumor invasiveness. In this study, we investigated a potential role for PTTG mRNA expression as a marker to evaluate the future clinical outcome of patients diagnosed with primary cancer of the head and neck. Methods Tumor samples derived from primary tumors of 89 patients suffering from a squamous cell carcinoma were analyzed for PTTG mRNA-expression and compared to corresponding unaffected tissue. Expression levels were correlated to standard clinico-pathological parameters based on a five year observation period. Results In almost all 89 tumor samples PTTG was found to be overexpressed (median fold increase: 2.1) when compared to the unaffected tissue specimens derived from the same patient. The nodal stage correlated with PTTG transcript levels with significant differences between pN0 (median expression: 1.32) and pN+ (median expression: 2.12; P = 0.016). In patients who developed a tumor recurrence we detected a significantly higher PTTG expression in primary tumors (median expression: 2.63) when compared to patients who did not develop a tumor recurrence (median expression: 1.29; P = 0.009). Since the median expression of PTTG in patients with tumor stage T1/2N0M0 that received surgery alone without tumor recurrence was 0.94 versus 3.82 in patients suffering from a tumor recurrence (P = 0.006), PTTG expression might provide a feasible mean of predicting tumor recurrence. Conclusion Elevated PTTG transcript levels might be used as a prognostic biomarker for future clinical outcome (i.e. recurrence) in primary squamous cell carcinomas of the head and neck, especially in early stages of tumor development.
Collapse
Affiliation(s)
- Christine Solbach
- Department of Gynecology and Obstetrics, Johann Wolfgang Goethe University School of Medicine, D-60590, Frankfurt am Main, Germany.
| | | | | | | | | |
Collapse
|
28
|
Abstract
Conventional cytopathology is an excellent tool for distinguishing benign from malignant thyroid nodules with high sensitivity and specificity. However, significant numbers of cases are indeterminate, resulting in many ultimately unnecessary diagnostic thyroidectomies. Numerous molecular markers have been studied in an attempt to improve the diagnostic accuracy of thyroid fine-needle aspiration cytology. Several markers, such as galectin-3 and thyroid peroxidase, have been extensively assessed and shown not only to differentiate malignant tumors from benign thyroid lesions with high sensitivity and specificity, but also to identify tumors associated with poor outcome. More recently, four other genes (PTTG, PBF, BRAF and MUC1) have been identified that show real promise as potential molecular markers in thyroid cancer, offering discrimination between tumor subtypes and providing valuable prognostic information. However, larger, well-controlled studies are needed before their introduction into routine clinical practice. The search for molecular markers represents one of the most exciting areas in translational thyroid cancer research. We are optimistic that molecular markers will be used in the near future as adjuncts to conventional histological techniques to improve diagnostic accuracy of fine-needle aspiration cytology for thyroid lesions, particularly those that are cytologically indeterminate.
Collapse
Affiliation(s)
- D S Kim
- a University of Birmingham, Division of Medical Sciences, Institute of Biomedical Research, Birmingham, B15 2TH, UK.
| | - C J McCabe
- b University of Birmingham, Division of Medical Sciences, Institute of Biomedical Research, Birmingham, B15 2TH, UK.
| |
Collapse
|
29
|
Minematsu T, Suzuki M, Sanno N, Takekoshi S, Teramoto A, Osamura RY. PTTG overexpression is correlated with angiogenesis in human pituitary adenomas. Endocr Pathol 2006; 17:143-53. [PMID: 17159247 DOI: 10.1385/ep:17:2:143] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/19/2023]
Abstract
Human pituitary tumor transforming gene (hPTTG1) was recently identified as a protooncogene, which is a regulator of the cell cycle, as a homolog of yeast securin and a transcriptional activator of several angiogenic factors. Here we examined the relationships of hPTTG1 expression with cell proliferation, expression of the angiogenic factor, VEGF (vascular endothelial growth factor), and numbers of the blood vessels in the normal and/or adenomatous pituitary. With the exception of TSHoma, the expression of hPTTG1 was significantly higher in pituitary adenomas than in the normal pituitary gland. The cell proliferation activity was higher in pituitary adenomas than in the normal pituitary. Pituitary cell proliferation was significantly correlated with the level of hPTTG1 expression in the normal pituitary tissue, but there was no such correlation in the adenomas. The significant correlation of hPTTG1 with the VEGF expression and the numbers of the blood vessels was elucidated in pituitary adenomas. It is particularly noteworthy that immunohistochemical double staining indicated co-localization of VEGF in many hPTTG1-positive tumor cells. In conclusion, higher levels of hPTTG1 expression contribute to the pathobiology of pituitary adenomas by promoting angiogenesis rather than by activating cell proliferation, whereas hPTTG1 expression is related to mitotic activity in the normal pituitary gland.
Collapse
Affiliation(s)
- Takeo Minematsu
- Department of Pathology, Tokai University School of Medicine. Bohseidai, Isehara, Kanagawa 259-1193, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Stratford AL, Boelaert K, Tannahill LA, Kim DS, Warfield A, Eggo MC, Gittoes NJL, Young LS, Franklyn JA, McCabe CJ. Pituitary tumor transforming gene binding factor: a novel transforming gene in thyroid tumorigenesis. J Clin Endocrinol Metab 2005; 90:4341-9. [PMID: 15886233 DOI: 10.1210/jc.2005-0523] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT There are currently no clear markers for the detection of differentiated thyroid cancer and its recurrence. Pituitary tumor transforming gene (PTTG) is a protooncogene implicated in the pathogenesis of multiple tumor types, which stimulates fibroblast growth factor-2 secretion via PTTG binding factor (PBF). OBJECTIVE The aim of this study was to ascertain whether PBF expression is associated with thyroid cancer outcome. DESIGN PBF expression was measured at the mRNA and protein level. Tissue was collected during surgery, with normal samples being taken from the contralateral lobe. In vitro studies ascertained the ability of PBF to transform cells and form tumors in nude mice and its subcellular localization. SETTING The study was conducted at a primary care/referral center. PATIENTS Thyroid tumors were collected from a series of 27 patients undergoing surgical excision of papillary and follicular thyroid tumors. INTERVENTION No intervention was conducted. MAIN OUTCOME MEASURE The expression of PBF in thyroid cancers compared with normal thyroid, hypothesized before the investigation to be raised in tumors, was the main outcome measure. RESULTS PBF mRNA expression was higher in differentiated thyroid carcinomas than in normal thyroid (P < 0.001; n = 27) and was independently associated with tumor recurrence (P = 0.002; R(2) = 0.49). PTTG was able to up-regulate PBF mRNA expression in vitro (P < 0.001; n = 12), and stable overexpression of PBF in NIH3T3 cells resulted in significant colony formation (P < 0.001; n = 12). In vivo, stable sc overexpression of PBF induced tumor formation in athymic nude mice. CONCLUSIONS PBF is an additional prognostic indicator in differentiated thyroid cancer that is transforming in vitro and tumorigenic in vivo.
Collapse
Affiliation(s)
- Anna L Stratford
- Division of Medical Sciences, University of Birmingham, Birmingham, B15 2TH, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Chamaon K, Kirches E, Kanakis D, Braeuninger S, Dietzmann K, Mawrin C. Regulation of the pituitary tumor transforming gene by insulin-like-growth factor-I and insulin differs between malignant and non-neoplastic astrocytes. Biochem Biophys Res Commun 2005; 331:86-92. [PMID: 15845362 DOI: 10.1016/j.bbrc.2005.03.124] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Indexed: 10/25/2022]
Abstract
The reasons for overexpression of the oncogene pituitary tumor transforming gene (PTTG) in tumors are still not fully understood. A possible influence of the insulin-like growth factor I (Igf-I) may be of interest, since enhanced Igf-I signalling was reported in various human tumors. We examined the influence of Igf-I and insulin on PTTG expression in human astrocytoma cells in comparison to proliferating non-neoplastic rat embryonal astrocytes. PTTG mRNA expression and protein levels were increased in malignant astrocytes treated with Igf-I or insulin, whereas in rat embryonic astrocytes PTTG expression and protein levels increased only when cells were exposed to Igf-I. Enhanced transcription did not occur after treatment with inhibitors of phosphoinositol-3-kinase (PI3K) and mitogen-activated protein kinase (MAPK), blocking the two basic signalling pathways of Igf-I and insulin. In addition to this transcriptional regulation, both kinases directly bind to PTTG, suggesting a second regulatory route by phosphorylation. However, the interaction of endogenous PTTG with MAPK and PI3K, as well as PTTG phosphorylation were independent from Igf-I or insulin. The latter results were also found in human testis, which contains high PTTG levels as well as in nonneoplastic astrocytes. This suggest, that PI3K and MAPK signalling is involved in PTTG regulation not only in malignant astrocytomas but also in non-tumorous cells.
Collapse
Affiliation(s)
- Kathrin Chamaon
- Department of Neuropathology, University of Magdeburg, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Zhou C, Liu S, Zhou X, Xue L, Quan L, Lu N, Zhang G, Bai J, Wang Y, Liu Z, Zhan Q, Zhu H, Xu N. Overexpression of human pituitary tumor transforming gene (hPTTG), is regulated by beta-catenin /TCF pathway in human esophageal squamous cell carcinoma. Int J Cancer 2005; 113:891-8. [PMID: 15514942 DOI: 10.1002/ijc.20642] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Overexpression of human pituitary tumor transforming gene (PTTG) is wildly detected in many tumors, including esophageal cancer. Besides overexpression of PTTG in esophageal squamous cell carcinoma (ESCC) tissues and cells, we detected accumulation of cytoplasmic beta-catenin in ESCC. In our study, a putative TCF4-binding element (TBE) was identified in PTTG promoter region. The activity of PTTG promoter containing the TBE was activated by S37Abeta-catenin and inhibited by dominant-negative TCF. Furthermore, the activation by S37Abeta-catenin was mostly abrogated among PTTG promoter region without the TBE or with a mutant one. By using biotin-streptavidin pull-down assay, we also found that the TBE among PTTG promoter bound to TCF-4 protein. Moreover, levels of PTTG mRNA and protein were increased by S37Abeta-catenin. Finally, it is noticeable that we detected a correlation between beta-catenin localization and PTTG expression in 69 primary ESCC (p<0.01). In brief, our study shows that overexpression of PTTG in ESCC is likely due to the activation of beta-catenin/WNT signaling. As a target gene of beta-catenin/TCF pathway, PTTG may play an important role in tumorigenesis of human ESCC.
Collapse
Affiliation(s)
- Cuiqi Zhou
- Laboratory of Cell and Molecular Biology, Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tfelt-Hansen J, Yano S, Bandyopadhyay S, Carroll R, Brown EM, Chattopadhyay N. Expression of pituitary tumor transforming gene (PTTG) and its binding protein in human astrocytes and astrocytoma cells: function and regulation of PTTG in U87 astrocytoma cells. Endocrinology 2004; 145:4222-31. [PMID: 15178645 DOI: 10.1210/en.2003-1661] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human securin, pituitary tumor transforming gene (PTTG), is a protooncogene. Here we report expressions of PTTG and its interacting protein, PTTG-binding factor in human astrocytic cells. PTTG expression was higher in malignant cells than in primary astrocytes, whereas PTTG-binding factor was not. Using a xenotransplantable, glioma cell line (U87), we observed that knocking down PTTG mRNA by RNA silencing inhibited serum-induced proliferation by approximately 50%. Furthermore, in U87 cells PTTG expression was up-regulated by promalignant ligands epithelial growth factor (EGF) and TGFalpha, both at the protein and mRNA levels. PTTG induction by EGF receptor (EGFR) ligands could be blocked by the specific EGFR inhibitor, AG1478. Hepatocyte growth factor (HGF) also induced PTTG but to a lesser extent than EGF. Although EGF stimulates HGF secretion in U87 cells, the effect of EGF on PTTG mRNA expression is independent of HGF as neutralizing antibody against HGF failed to abolish EGF-induced up-regulation of PTTG mRNA. PTTG mRNA was unchanged by incubating U87 cells with the promalignant growth factor TGFbeta, apoptosis inducing TNFalpha and ligands for nuclear receptors, such as retinoic acid and retinoid X receptors and peroxisome proliferator-activated receptor-gamma, known for their growth-inhibitory and apoptosis-inducing effects on gliomas. In addition, 17beta-estradiol and Ca2+, known to activate PTTG expression, did not change PTTG mRNA levels in U87 cells. In summary, we show higher PTTG expression in astrocytoma than normal astrocytes and secondly, PTTG is involved in glioma cell growth. Finally, regulation of its expression has glioma-specific features and is selectively regulated by promalignant cytokines including EGFR ligands and HGF.
Collapse
Affiliation(s)
- Jacob Tfelt-Hansen
- Division of Endocrinology, Diabetes, and Hypertension,Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Hamid T, Kakar SS. PTTG/securin activates expression of p53 and modulates its function. Mol Cancer 2004; 3:18. [PMID: 15242522 PMCID: PMC479695 DOI: 10.1186/1476-4598-3-18] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2004] [Accepted: 07/08/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pituitary tumor transforming gene (PTTG) is a novel oncogene that is expressed abundantly in most tumors. Overexpression of PTTG induces cellular transformation and promotes tumor formation in nude mice. PTTG has been implicated in various cellular processes including sister chromatid separation during cell division as well as induction of apoptosis through p53-dependent and p53-independent mechanisms. The relationship between PTTG and p53 remains unclear, however. RESULTS Here we report the effects of overexpression of PTTG on the expression and function of p53. Our results indicate that overexpression of PTTG regulates the expression of the p53 gene at both the transcriptional and translational levels and that this ability of PTTG to activate the expression of p53 gene is dependent upon the p53 status of the cell. Deletion analysis of the p53 gene promoter revealed that only a small region of the p53 gene promoter is required for its activation by PTTG and further indicated that the activation of p53 gene by PTTG is an indirect effect that is mediated through the regulation of the expression of c-myc, which then interacts with the p53 gene promoter. Our results also indicate that overexpression of PTTG stimulates expression of the Bax gene, one of the known downstream targets of p53, and induces apoptosis in a human embryonic kidney cell line (HEK293). This stimulation of bax expression by PTTG is indirect and is mediated through modulation of p53 gene expression. CONCLUSIONS Overexpression of PTTG activates the expression of p53 and modulates its function, with this action of PTTG being mediated through the regulation of c-myc expression. PTTG also up-regulates the activity of the bax promoter and increases the expression of bax through modulation of p53 expression.
Collapse
Affiliation(s)
- Tariq Hamid
- Department of Medicine, University of Louisville, Louisville KY 40202, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville KY 40202, USA
| | - Sham S Kakar
- Department of Medicine, University of Louisville, Louisville KY 40202, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville KY 40202, USA
| |
Collapse
|
35
|
Clem AL, Hamid T, Kakar SS. Characterization of the role of Sp1 and NF-Y in differential regulation of PTTG/securin expression in tumor cells. Gene 2004; 322:113-21. [PMID: 14644503 DOI: 10.1016/j.gene.2003.08.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Pituitary tumor transforming gene (PTTG), also known as securin, is a regulator of cell division that is overexpressed in many tumors. Its expression is cell cycle regulated, although its transcriptional regulation is yet to be determined. The 5' RACE analysis of the human testis mRNA revealed the existence of a previously unreported transcription start site at 317 bp upstream of the translation start site (ATG). This gene is known to be composed of five exons and four introns, which is now changed to six exons and five introns. To map the promoter region, and to understand its regulation, we designed several fusion constructs of the 5' flanking region of PTTG including the sequence from nucleotide -1373 to -3 (relative to the translation start site) to a luciferase reporter gene. Transient transfection of these constructs in prostate cancer cell line (PC-3) and fibroblast cell line (HS27) confirmed the existence of promoter for PTTG between nucleotides -161 and -3 (in relation to translation start site). The 5' and 3' deletion analysis of the PTTG flanking region and electrophoretic mobility shift assays revealed binding of Sp1 and NF-Y transcription factors within nucleotides -540 to -500. Chromatin immunoprecipitation (ChIP) assays of the HS27 and PC-3 cells revealed the binding of Sp1 protein to PTTG promoter sequence in vivo. Site-directed mutagenesis of the Sp1 consensus sequence resulted in approximately 70% reduction of the overall transcriptional activation of the PTTG promoter, whereas mutation of the NF-Y sequence resulted in approximately 25% reduction. Deletion of both Sp1 and NF-Y consensus sequences resulted in 90% loss of PTTG promoter activity. It was further observed, by Western blot analysis, that the levels of Sp1 protein are higher in PC-3 cells when compared to levels in HS27 cells, possibly contributing to a tissue-specific effect. Our studies indicate an important role of Sp1 in transcription regulation of PTTG expression in tumors.
Collapse
Affiliation(s)
- Amy L Clem
- Department of Medicine and James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | |
Collapse
|
36
|
Abstract
The anterior pituitary gland integrates the repertoire of hormonal signals controlling thyroid, adrenal, reproductive, and growth functions. The gland responds to complex central and peripheral signals by trophic hormone secretion and by undergoing reversible plastic changes in cell growth leading to hyperplasia, involution, or benign adenomas arising from functional pituitary cells. Discussed herein are the mechanisms underlying hereditary pituitary hypoplasia, reversible pituitary hyperplasia, excess hormone production, and tumor initiation and promotion associated with normal and abnormal pituitary differentiation in health and disease.
Collapse
Affiliation(s)
- Shlomo Melmed
- Cedars-Sinai Medical Center, David Geffen School of Medicine, University of California, 8700 Beverly Boulevard, Room 2015, Los Angeles, California 90048, USA.
| |
Collapse
|
37
|
Mu YM, Oba K, Yanase T, Ito T, Ashida K, Goto K, Morinaga H, Ikuyama S, Takayanagi R, Nawata H. Human pituitary tumor transforming gene (hPTTG) inhibits human lung cancer A549 cell growth through activation of p21(WAF1/CIP1 ). Endocr J 2003; 50:771-81. [PMID: 14709851 DOI: 10.1507/endocrj.50.771] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Pituitary tumor transforming gene (PTTG) is a proto-oncogene cloned from rat GH4 cells. This gene was able to induce cell transformation in vitro and is also associated with p53-dependent and -independent apoptosis. In this study, we cloned human PTTG (hPTTG) from a pituitary tumor and then stably transfected the hPTTG into HeLa and A549 cells. An overexpression of hPTTG significantly inhibited cell growth, which was determined by the adherent cell growth properties, colony formation in soft agar and [3H] thymidine incorporation, respectively, in HeLa and A549 cells. The inhibitory effect on cell growth was associated with the activation of p21WAF1/CIP1 in A549 cells, but not in HeLa cells. The hPTTG overexpression increased both the p21WAF1/CIP1 mRNA and protein expression levels as determined by both Northern and Western blot analysis, respectively, in A549 cells. The increased expression of p21WAF1/CIP1 mRNA was regulated at the transcription level and was independent on p53 expression because the luciferase activity increased after the co-transfection of hPTTG and p21WAF1/CIP1 promoter fragments with and without a p53 binding sequence. The subcellular distribution of hPTTG was dependent on cell type, and was predominantly in the nucleus in HeLa, Cos-7 and DU145 cells, but showed a diffuse distribution in both the nucleus and cytoplasm in A549, DLD-1 and NIH3T3 cells. These results indicate that an overexpression of hPTTG inhibits the cell growth due to different mechanisms, which are p21WAF1/CIP1 -dependent and -independent.
Collapse
Affiliation(s)
- Yi-Ming Mu
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tfelt-Hansen J, Schwarz P, Terwilliger EF, Brown EM, Chattopadhyay N. Calcium-sensing receptor induces messenger ribonucleic acid of human securin, pituitary tumor transforming gene, in rat testicular cancer. Endocrinology 2003; 144:5188-93. [PMID: 12970167 DOI: 10.1210/en.2003-0520] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pituitary tumor transforming gene (PTTG), the human ortholog of securin, is an oncogene. Few normal tissues express PTTG, although in the testis, it is more abundantly expressed. In cancer, however, its wide expression has been directly correlated with the proliferation and angiogenesis, although very little is known about the overall regulation of the PTTG gene. In this study, we investigate the role of the calcium-sensing receptor (CaR), a G protein-coupled receptor (GPCR), in regulating PTTG in a widely used model of humoral hypercalcemia of malignancy, the rat H-500 Leydig cell testicular cancer. We show that extracellular calcium (Ca2+o) up-regulates PTTG mRNA. This up-regulation has a rapid onset, starting at 0.5 h, and remains up-regulated until 40 h. The up-regulation was also Ca2+o concentration dependent, with increases (mean +/- se) of 4.22 +/- 1.61-fold, 5.11 +/- 1.11-fold, and 5.64 +/- 1.92-fold at 5, 7.5, and 10 mm calcium, respectively, compared with 0.5 mm Ca2+o. This effect was abolished by overexpression of a dominant-negative CaR (R185Q), thereby confirming that the effect of high Ca2+o is CaR mediated. Another GPCR agonist, ADP, had no effect on PTTG expression. Because PTTG has been reported to induce angiogenesis, we investigated the effect of elevated Ca2+o on vascular endothelial growth factor (VEGF) expression. Indeed high calcium up-regulated VEGF mRNA by 1.59 +/- 0.22-fold. In conclusion, we show for the first time that a GPCR, the CaR, stimulates the synthesis of PTTG mRNA in a nonmetastasizing model for humoral hypercalcemia of malignancy and, in the process, might induce angiogenesis via VEGF.
Collapse
Affiliation(s)
- Jacob Tfelt-Hansen
- Endocrine-Hypertension Division, Department of Medicine and Membrane Biology Program, Brigham and Women's Hospital and Harvard Medical School, 221 Longwood Avenue, Boston, Massachusetts 02115, USA.
| | | | | | | | | |
Collapse
|
39
|
Jin ZY, Cheng RX, Zheng CL, Zheng H. Expression of PTTG and c-myc gene in human primary hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2003; 11:1677-1681. [DOI: 10.11569/wcjd.v11.i11.1677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the relationship between expressions of PTTG and c-myc genes and oncogenesis in human hepatocellular carcinomas(HCC).
METHODS In situ hybridization (DNA-RNA) and immunohistochemistry (SP method) methods were used to detect the expressions of PTTG and c-myc genes in 61 cases of human hepatocellular carcinomas.
RESULTS The distributions of positive cells of PTTG mRNA and PTTG protein in HCC were diffuse, aggregate or scattered. The positive stainings of PTTG mRNA and PTTG protein were plasma and submembrane types. The positive rates of PTTG mRNA and PTTG protein were 72.1% (44/61) and 78.7% (48/61) in HCC, and 93.4% (57/61) and 91.8% (56/61) in pericarcinomatous liver tissues. The expressions of PTTG mRNA and PTTG protein in HCC were significantly lower than those in paracancerous tissues (P<0.005, P<0.05). The expression of PTTG gene was significantly correlated with that of c-myc gene (P<0.005).
CONCLUSION Overexpression of PTTG is related to human hepatocellular carcinogenesis. C-myc gene activated by PTTG protein may play an important role in hepatocellular transformation and carcinogenesis.
Collapse
Affiliation(s)
- Zhong-Yuan Jin
- Department of Pathology, Xiangya School of Medicine, Zhongnan University, Changsha 410078, Hunan Province, China
| | - Rui-Xue Cheng
- Department of Pathology, Xiangya School of Medicine, Zhongnan University, Changsha 410078, Hunan Province, China
| | - Chang-Li Zheng
- Department of Pathology, Xiangya School of Medicine, Zhongnan University, Changsha 410078, Hunan Province, China
| | - Hui Zheng
- Department of Pathology, Xiangya School of Medicine, Zhongnan University, Changsha 410078, Hunan Province, China
| |
Collapse
|
40
|
Affiliation(s)
- Chris J McCabe
- Division of Medical Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | | |
Collapse
|
41
|
Yao YQ, Xu JS, Lee WM, Yeung WSB, Lee KF. Identification of mRNAs that are up-regulated after fertilization in the murine zygote by suppression subtractive hybridization. Biochem Biophys Res Commun 2003; 304:60-6. [PMID: 12705884 DOI: 10.1016/s0006-291x(03)00537-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Transcriptions occur in mouse preimplantation embryos as early as one-cell stage. However, our understanding on gene expression at this stage is lacking. The present study applied suppression subtractive hybridization (SSH) to compared gene expression profiles of mouse zygote and oocyte. Forty-four differentially expressed genes were selected and shown positive signals by reverse dot-blot hybridization. DNA sequences comparison of these putative clones with the GenBank/EMBL databases using BLAST search identified 38 clones with >90% identity to known genes and six novel clones with less than 70% homology to the databases. Eleven out of the 44 differentially expressed clones were either originally isolated from male embryo or testis-specific genes, suggesting that these genes may be derived from paternal genome. Five differentially expressed genes of interest, including bromodomain-containing protein BP75, spindlin, radixin, pituitary tumor-transforming gene (PTTG), and proteoglycan core protein (serglycin) were further studied by semi-quantitative RT-PCR. It is noted that spindlin which involves in cell division is highly expressed in zygote, suggesting that this protein may play an important role in zygotic gene activation (ZGA) and early stage development in 1-cell stage mouse embryos.
Collapse
Affiliation(s)
- Yuan-Qing Yao
- Department of Obstetrics and Gynaecology, The University of Hong Kong, Hong Kong, SAR, China
| | | | | | | | | |
Collapse
|
42
|
Sang N, Stiehl DP, Bohensky J, Leshchinsky I, Srinivas V, Caro J. MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300. J Biol Chem 2003; 278:14013-9. [PMID: 12588875 PMCID: PMC4518846 DOI: 10.1074/jbc.m209702200] [Citation(s) in RCA: 273] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hypoxia-inducible factors (HIF) are a family of heterodimeric transcriptional regulators that play pivotal roles in the regulation of cellular utilization of oxygen and glucose and are essential transcriptional regulators of angiogenesis in solid tumor and ischemic disorders. The transactivation activity of HIF complexes requires the recruitment of p300/CREB-binding protein (CBP) by HIF-1 alpha and HIF-2 alpha that undergo oxygen-dependent degradation. HIF activation in tumors is caused by several factors including mitogen-activated protein kinase (MAPK) signaling. Here we investigated the molecular basis for HIF activation by MAPK. We show that MAPK is required for the transactivation activity of HIF-1 alpha. Furthermore, inhibition of MAPK disrupts the HIF-p300 interaction and suppresses the transactivation activity of p300. Overexpression of MEK1, an upstream MAPK activator, stimulates the transactivation of both p300 and HIF-1 alpha. Interestingly, the C-terminal transactivation domain of HIF-1 alpha is not a direct substrate of MAPK, and HIF-1 alpha phosphorylation is not required for HIF-CAD/p300 interaction. Taken together, our data suggest that MAPK signaling facilitates HIF activation through p300/CBP.
Collapse
Affiliation(s)
| | | | | | | | | | - Jaime Caro
- To whom correspondence may be addressed: Cardeza Foundation and Dept. of Medicine, Thomas Jefferson University, 1015 Walnut St., Curtis Bldg., Rm. 809, Philadelphia, PA 19107. Tel.: 215-955-5118; Fax: 215-923-3836;
| |
Collapse
|
43
|
Stappenbeck TS, Mills JC, Gordon JI. Molecular features of adult mouse small intestinal epithelial progenitors. Proc Natl Acad Sci U S A 2003; 100:1004-9. [PMID: 12552106 PMCID: PMC298716 DOI: 10.1073/pnas.242735899] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The adult mouse small intestinal epithelium undergoes perpetual regeneration, fueled by a population of multipotential stem cells and oligopotential daughters located at the base of crypts of Lieberkühn. Although the morphologic features of small intestinal epithelial progenitors (SiEPs) are known, their molecular features are poorly defined. Previous impediments to purification and molecular characterization of SiEPs include lack of ex vivo clonigenic assays and the difficulty of physically retrieving them from their niche where they are interspersed between their numerous differentiated Paneth cell daughters. To overcome these obstacles, we used germ-free transgenic mice lacking Paneth cells to obtain a consolidated population of SiEPs with normal proliferative activity. These cells were harvested by laser capture microdissection. Functional genomics analysis identified 163 transcripts enriched in SiEPs compared with Paneth cell-dominated normal crypt base epithelium. The dataset was validated by (i) correlation with the organellar composition of SiEPs versus Paneth cells, (ii) similarities to databases generated from recent mouse hematopoietic and neural stem cell genome anatomy projects, and (iii) laser capture microdissectionreal-time quantitative RT-PCR studies of progenitor cell-containing populations retrieved from the small intestines, colons, and stomachs of conventionally raised mice. The SiEP profile has prominent representation of genes involved in c-myc signaling and in the processing, localization, and translation of mRNAs. This dataset, together with our recent analysis of gene expression in the gastric stem cell niche, discloses a set of molecular features shared by adult mouse gut epithelial progenitors.
Collapse
Affiliation(s)
- Thaddeus S Stappenbeck
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
44
|
Kanakis D, Kirches E, Mawrin C, Dietzmann K. Promoter mutations are no major cause of PTTG overexpression in pituitary adenomas. Clin Endocrinol (Oxf) 2003; 58:151-5. [PMID: 12580929 DOI: 10.1046/j.1365-2265.2003.01683.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The pituitary tumour transforming gene (PTTG) was proven to cause transformation of NIH3T3 fibroblasts, which produce tumours when transplanted into immunodeficient mice. PTTG is overexpressed in about 90% of pituitary adenomas. The reason for its overexpression is still unclear. DESIGN Because promoter mutations may play a role for an altered regulation of PTTG transcription in the pituitary adenomas, we analysed two promoter regions which were characterized previously as functionally important. PATIENTS Twenty-five patients of both sexes with pituitary adenomas, mainly null-cell adenomas, were included in this study. MEASUREMENTS Both DNA regions were amplified from paraffin sections by PCR and analysed for small deletions or insertions on polyacrylamide gels in all patients. In 16 cases both DNA regions were sequenced to detect base substitutions. RESULTS No deletions/insertions and no tumour-specific substitutions were found. In three homopolymeric regions a polymorphism was detected, which also occurred in control sequences. In addition, these tracts showed some degree of length instability. CONCLUSIONS Promoter mutations do not play a major role for the enhanced PTTG transcription in pituitary adenomas. Therefore, DNA-binding proteins, hypomethylation or other epigenetic factors may be responsible for PTTG overexpression.
Collapse
Affiliation(s)
- D Kanakis
- Department of Neuropathology, University of Magdeburg, Magdeburg, Germany.
| | | | | | | |
Collapse
|
45
|
Fagin JA. Minireview: branded from the start-distinct oncogenic initiating events may determine tumor fate in the thyroid. Mol Endocrinol 2002; 16:903-11. [PMID: 11981026 DOI: 10.1210/mend.16.5.0838] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Thyroid follicular neoplasms commonly have aneuploidy, presumably due to chromosomal instability. This property is associated with a greater malignant potential and worse prognosis. Recently, there has been considerable progress in our understanding of mechanisms that may account for chromosomal instability in cancer cells. Many tumors with chromosomal instability have abnormalities in the cell cycle checkpoint that monitors the fidelity of mitosis. Mutations of Bub1 or BubR1, genes coding for kinases involved in mitotic spindle assembly checkpoint signaling, are found in a small subset of aneuploid tumors. Other components of protein complexes responsible for attachment of kinetochores to microtubules, or for cohesion between sister chromatids, may also be subject to alterations during tumor progression. Here, we also discuss the evidence that certain oncogenic events, such as Ras mutations, may predispose cells to chromosomal instability by favoring inappropriate posttranslational changes in mitotic checkpoint components through activation of upstream kinases during tumor initiation or progression.
Collapse
Affiliation(s)
- James A Fagin
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0547, USA.
| |
Collapse
|
46
|
|
47
|
Yin H, Fujimoto N, Maruyama S, Asano K. Strain difference in regulation of pituitary tumor transforming gene (PTTG) in estrogen-induced pituitary tumorigenesis in rats. Jpn J Cancer Res 2001; 92:1034-40. [PMID: 11676853 PMCID: PMC5926607 DOI: 10.1111/j.1349-7006.2001.tb01057.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Recently a novel oncogene, PTTG (pituitary tumor transforming gene) was isolated from a rat pituitary tumor cell line whose expression is apparently correlated with pituitary tumorigenesis. In the rat, estradiol (E(2)) is known to induce anterior pituitary hyperplasia. The effects of E(2), however, vary greatly among rat strains. Therefore we examined the expression of PTTG and its regulation by E(2) in F344, Wistar, Brown-Norway and Donryu rats. Four-week-old females were ovariectomized and a pellet containing 10 mg of E(2) was given s.c. Total RNA was isolated from the pituitary gland and PTTG mRNA was measured with a competitive RT-PCR technique. The F344 strain was the most susceptible to E(2) induction of pituitary tumorigenesis, followed by Wistar and Brown-Norway, while no increase in pituitary weight was noted in Donryu rats. PTTG mRNA in the gland was induced by E(2) within 48 - 72 h in F344 and Wistar, but not in Brown-Norway or Donryu strains. These data suggest that PTTG expression may at least in part be responsible for strain differences in E(2)-induced pituitary tumorigenesis.
Collapse
Affiliation(s)
- H Yin
- Department of Cancer Research, RIRBM, Hiroshima University, Minami-ku, Hiroshima 734-8553
| | | | | | | |
Collapse
|
48
|
Abstract
In recent years, remarkable progress has been made in the understanding of the pathogenesis of pituitary tumors. Pituitary tumors originate from the uncontrolled proliferation of a single transformed cell in which an initiating event has caused a gain of proliferative function. After the initiation, promoting factors cooperate in the clonal expansion. Common oncogenes, such as ras, are only exceptionally involved. The only activating mutations identified so far are gsp mutations causing the constitutive activation of cAMP pathway. However, gsp-positive adenomas are not associated to a more aggressive tumoral phenotype. The oncogenic potential of gsp mutations is limited by a more rapid degradation of the mutant Gs(alpha) with respect to the wild-type protein, and by a faster removal of cAMP due to increased phosphodiesterase activity. Estrogen-inducible gene sequences with transforming properties (pituitary tumor-transforming gene (PTTG)) have been identified in human pituitary tumors. Human pituitary tumor-transforming gene (hPTTG) is involved both in early pituitary tumorigenesis, as it causes in vitro and in vivo transformation acting as a transcription activator, and in tumor progression, as it regulates the production of basic fibroblast growth factor (bFGF), a potent activator of angiogenesis and mitogenesis. Moreover, a role of cyclin D1 in pituitary tumorigenesis is emerging. The allelic loss of loci for unknown oncosuppressor genes are currently under investigation, while an exceedingly limited role for menin gene and RB1 has been demonstrated for sporadic pituitary tumors. Abnormal methylation that predisposing toward genetic instability may favor the allelic loss or the reduced expression of oncosuppressor genes, is also an emerging field of investigation. Several promoting factors, including the excessive action of physiological stimulators, the defective action of inhibitors, the susceptibility to respond to inappropriate stimuli and the locally produced growth factors, help in tumor progression. The study of homeobox genes that intervene in pituitary cell differentiation may help in expanding our knowledge in pituitary tumor cell genealogy.
Collapse
Affiliation(s)
- G Faglia
- Institute of Endocrine Sciences, Ospedale Maggiore IRCCS, University of Milan, Italy.
| | | |
Collapse
|
49
|
Abstract
Pituitary tumors constitute 10% of intracranial neoplasms and are mostly benign, monoclonal adenomas derived from single mutant cells. Pituitary oncogenes have been intensively studied and three of them, gsp, ccnd1, and PTTG are abundant in significant numbers of cases. gsp is present in approximately 40% of Caucasian patients with GH-secreting tumors and results from a mutated, constitutively active alpha subunit of Gs protein. Persistent activation of the cAMP-PKA-CREB pathway may lead to uncontrolled cell proliferation and GH secretion. ccnd1 is overexpressed cyclin D1, and cyclin D1 gene is amplified in some pituitary tumors. PTTG is expressed in most pituitary tumors. PTTG is localized to both the nucleus and cytoplasm and interacts with several protein partners. At least three tumorigenesis mechanisms are proposed for human PTTG. 1) PTTG and FGF form a positive feedback loop and stimulate tumor vascularity. 2) PTTG transactivates c-myc or other pro-proliferation genes. 3) PTTG overexpression causes aneuploidy. PTTG expression activates p53 and causes p53-dependent and -independent apoptosis. Due to lack of functional human pituitary cell cultures and appropriate animal models for pituitary tumors, many of the results reviewed here are obtained from heterologous systems.
Collapse
Affiliation(s)
- Run Yu
- Cedars‐Sinai Research Institute‐UCLA School of Medicine, Los Angeles, CA 90048
| | - Shlomo Melmed
- Cedars‐Sinai Research Institute‐UCLA School of Medicine, Los Angeles, CA 90048
| |
Collapse
|
50
|
Pei L. Identification of c-myc as a down-stream target for pituitary tumor-transforming gene. J Biol Chem 2001; 276:8484-91. [PMID: 11115508 DOI: 10.1074/jbc.m009654200] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pituitary tumor-transforming gene (PTTG) encodes a protein implicated in cellular transformation and transcriptional regulation. To identify downstream target genes, I established cell lines with tightly regulated inducible expression of PTTG. DNA arrays were used to analyze gene expression profiles after PTTG induction. I identified c-myc oncogene as a major PTTG target. Induction of PTTG resulted in increased cell proliferation through activation of c-myc. I showed that PTTG activates c-myc transcription in transfected cells. PTTG binds to c-myc promoter near the transcription initiation site in a protein complex containing the upstream stimulatory factor (USF1). I have defined the PTTG DNA-binding site and mapped PTTG DNA binding domain to a region between amino acids 61 and 118. Furthermore, I demonstrated that PTTG DNA binding is required for its transcriptional activation function. These results definitively established the role of PTTG as a transcription activator and indicate that PTTG is involved in cellular transformation and tumorigenesis through activation of c-myc oncogene.
Collapse
Affiliation(s)
- L Pei
- Division of Endocrinology and Metabolism, Cedars-Sinai Research Institute, UCLA School of Medicine, Los Angeles, California 90048, USA.
| |
Collapse
|