1
|
Li X, Ding N, Ma W, Zhang M. IFIH1-mediated post-transcriptional regulation of PTTG1 promotes proliferation and affects PHA-848125 sensitivity and prognosis in oropharyngeal carcinoma. Am J Cancer Res 2024; 14:2157-2171. [PMID: 38859832 PMCID: PMC11162656 DOI: 10.62347/ylcq4222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/28/2023] [Indexed: 06/12/2024] Open
Abstract
The pituitary tumor-transforming gene 1 (PTTG1) is an oncogene involved in chromosomal segregation, DNA repair, apoptosis, and metabolism. PTTG1 can be used for clinical diagnosis and treatment and is a potential target for oropharyngeal carcinoma. The proliferation and viability of Cal27 and FaDu cells were assessed using the CCK-8 assay. Real-time PCR and western blotting, respectively, were used to analyze the mRNA and protein expression levels of PTTG1 and IFIH1. The interaction between PTTG1 mRNA and the translational regulatory protein IFIH1 was analyzed using RNA pull-down, RNA immunoprecipitation, and luciferase reporter assays. PTTG1 protein was significantly overexpressed in oropharyngeal carcinoma, whereas PTTG1 mRNA was not. We hypothesized that a translation regulatory protein plays a post-transcriptional role in PTTG1. The IFIH1 protein specifically bound to the 42-52 nt region of PTTG1 mRNA, promoted the translation of PTTG1, and promoted the proliferation of oropharyngeal cancer cells. Administration of the PTTG1 inhibitor PHA-848125 and silencing of IFIH1 synergistically decreased the expression of PTTG1, inhibited the proliferation of oropharyngeal cancer cells, and indicated a good prognosis. We found that the IFIH1-PTTG1 axis could regulate the PHA-848125 response and functionally mediate inter-individual oropharyngeal cancer susceptibility and prognosis. This study aimed to confirm the upstream regulatory genes of PTTG1 and further investigate the specific interactions in this signaling pathway, which will provide a new approach for the treatment of oropharyngeal carcinoma.
Collapse
Affiliation(s)
- Xiaoli Li
- Department of Otolaryngology, The Fourth Affiliated Hospital of China Medical UniversityNo. 4 Chongshan East Road, Huanggu District, Shenyang 110032, Liaoning, P. R. China
| | - Nan Ding
- Clinical Laboratory, Hebei General HospitalNo. 348 Heping West Road, Xinhua District, Shijiazhuang 050051, Hebei, P. R. China
| | - Wenting Ma
- Department of Pathology, Hong Kong University of Shenzhen HospitalHaiyuan 1st Road, Futian District, Shenzhen 518053, Guangdong, P. R. China
| | - Ming Zhang
- Department of Otolaryngology, The Fourth Affiliated Hospital of China Medical UniversityNo. 4 Chongshan East Road, Huanggu District, Shenyang 110032, Liaoning, P. R. China
| |
Collapse
|
2
|
He W, Zhu H, Zhang S, Shu G, Lei H, Yin G, Ni X, Wang M, Wu Q. Promoter Methylation Changes in DNA Damage-Response Genes in Ovarian Cancer and Their Correlation with Prognosis. CLIN EXP OBSTET GYN 2024; 51. [DOI: 10.31083/j.ceog5105109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Background: Ovarian cancer has a poor prognosis, and DNA damage-response (DDR) genes are associated with both its occurrence and prognosis. However, previous studies have mostly focused on genetic mutations, with no clear conclusions on epigenetic factors such as DNA methylation. Methods: In this study, we comprehensively investigated the relationship between promoter methylation of DDR genes and ovarian cancer prognosis. We performed combined multidata analysis of the promoter methylation, expression, homologous recombination defieiency (HRD) score, and drug sensitivity of 377 DDR genes in ovarian cancer by utilizing The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. We then validated abnormal promoter methylation and its relationship with overall survival in clinical samples. Results: Our analysis identified 52 methylation-driven DDR genes that exhibited abnormal expression due to abnormal promoter methylation. These genes are mostly related to BRCA1-related DNA damage repair and cell cycle regulatory pathways. Further studies revealed six of these genes, BRCA1, PTTG1, TTK, AURKA, CDC6, and E2F1, to be significantly associated with HRD scores. Among them, E2F1, PTTG1, and CDC6 are associated with drug sensitivity. Finally, we verified in 81 ovarian cancer samples that methylation of the promoter of these three genes was significantly associated with patient survival. Conclusions: Our study identified a large number of methylation-driven aberrantly expressed DDR genes in ovarian cancer, some of which affect disease prognosis. Levels of methylation of these gene promoters may serve as potential prognostic markers.
Collapse
Affiliation(s)
- Wanhong He
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Haijun Zhu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Sufen Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Han Lei
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Xiaohua Ni
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, 410013 Changsha, Hunan, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, 200237 Shanghai, China
| |
Collapse
|
3
|
Liu X, Zeng W, Zheng D, Tang M, Zhou W. Clinical significance of securin expression in solid cancers: A PRISMA-compliant meta-analysis of published studies and bioinformatics analysis based on TCGA dataset. Medicine (Baltimore) 2022; 101:e30440. [PMID: 36123907 PMCID: PMC9478268 DOI: 10.1097/md.0000000000030440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Numerous studies have investigated the clinical significance of securin expression in solid cancers; however, the results have been inconsistent. Hence, we performed a meta-analysis of published studies to assess the clinical value of securin expression in patients with solid cancers. METHODS The Chinese National Knowledge Infrastructure, Web of Science, PubMed, and EMDASE databases were searched for eligible studies (from inception up to April 2021). Bioinformatics analysis based on The Cancer Genome Atlas dataset was also performed to evaluate the prognostic value of securin expression. RESULTS A total of 25 articles with 26 studies were included in the meta-analysis. The results of the meta-analysis implied that high securin expression was positively correlated with unfavorable overall survival (OS) (hazard ratio = 1.52, 95% CI, 1.33-1.73; P < .001) and lymph node metastasis (odd ratio = 2.96, 95% CI, 2.26-3.86; P < .001). Consistently, our bioinformatics analysis showed that increased securin expression was associated with worse OS and shorter disease-free survival in cancer patients. CONCLUSION Our study indicated that securin overexpression was positively associated with metastasis and inversely related to the prognosis of patients with solid cancers. However, additional high-quality studies should be conducted to validate these findings.
Collapse
Affiliation(s)
- Xiang Liu
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital, University of South China, Hengyang, China
| | - Wei Zeng
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital, University of South China, Hengyang, China
| | - Dayang Zheng
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital, University of South China, Hengyang, China
| | - Min Tang
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital, University of South China, Hengyang, China
| | - Wangyan Zhou
- Department of Medical Humanities and Education Department, the First Affiliated Hospital, University of South China, Hengyang, China
- * Correspondence: Wangyan Zhou, Department of Medical Humanities and Education Department, the First Affiliated Hospital, University of South China, No. 69 Chuanshan Road, Hengyang 421001, China (e-mail: )
| |
Collapse
|
4
|
Demin DE, Stasevich EM, Murashko MM, Tkachenko EA, Uvarova AN, Schwartz AM. Full and D-BOX-Deficient PTTG1 Isoforms: Effects on Cell Proliferation. Mol Biol 2022. [DOI: 10.1134/s0026893322060061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
5
|
Lu L, Xu X, Du X, Zeng T, Yang T, Chen Y, Tao Z, Zhong S, Wen J, Zhou C. Transcriptome analyses to reveal the dynamic change mechanism of pigeon magnum during one egg-laying cycle. Mol Reprod Dev 2020; 87:1141-1151. [PMID: 33084116 DOI: 10.1002/mrd.23428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 09/12/2020] [Indexed: 12/14/2022]
Abstract
We analyzed the transcriptome of pigeon magnum in three stages (C1: pre-ovulation, C2: post-ovulation, C3: 5-6 days after ovulation) to elucidate the molecular and cellular events associated with morphological changes during the laying cycle. We observed that C1 was highly developed, apoptosis rate was highest in C2, and C3 attained the smallest size. Through RNA-sequencing, we obtained 54,764,938 (97.2%) high-quality clean reads that aligned to 20,767 genes. Gene expression profile analysis showed the greatest difference between C1 and C3; 3966 differentially expressed genes (DEGs) were identified, of which 2250 genes were upregulated and 1716 genes were downregulated in C1. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed that protein processing and transport activities were prominent in C1, and upregulated genes included those related to signal recognition particle (SRP), signal recognition particle receptor (SRPR), translocon, GRP78, RRBP1, TRAP, TRAM1, and OST. Egg white protein-related gene expression was highest, with OVALY being the most highly expressed. In C2, apoptosis-related gene expression was higher than in C1, and fatty acid metabolism was active, which may be correlated with magnum tissue regression. Collagen- and laminin-related gene expression was prominent in C1 and C3, indicating roles in egg white protein generation and magnum reconstruction. PR gene expression was highest and exhibited drastic change in the three groups, indicating that PR and its regulation may be involved in changes in magnum morphology and function. Through the identification and functional analysis of DEGs and other crucial genes, this may contribute to understand the egg white protein production, magnum tissue regression, and magnum regeneration mechanisms.
Collapse
Affiliation(s)
- Lizhi Lu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Xiaoqin Xu
- Institute of Ecology, China West Normal University, Nanchong, Sichuan, China
| | - Xue Du
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Tao Zeng
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Tingbang Yang
- Institute of Ecology, China West Normal University, Nanchong, Sichuan, China
| | - Yao Chen
- Institute of Ecology, China West Normal University, Nanchong, Sichuan, China
| | - Zhengrong Tao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Science, Hangzhou, Zhejiang, China
| | - Shengliang Zhong
- PingYang XingLiang Pigeon Farming Co. Ltd., Wenzhou, Zhejiang, China
| | - Jihui Wen
- PingYang AoFeng Pigeon Farming Co. Ltd., Wenzhou, Zhejiang, China
| | - Caiquan Zhou
- Institute of Ecology, China West Normal University, Nanchong, Sichuan, China
| |
Collapse
|
6
|
Lin P, Pan Y, Chen H, Jiang L, Liao Y. Key genes of renal tubular necrosis: a bioinformatics analysis. Transl Androl Urol 2020; 9:654-664. [PMID: 32420172 PMCID: PMC7215026 DOI: 10.21037/tau.2019.11.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background To explore the key genes in renal tubular necrosis. Methods Microarray datasets GSE69644, GSE27168, and GSE2027 were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified and we performed functional enrichment analysis. The network of protein interaction and gene interaction was constructed, and the module analysis was conducted using Cytoscape. Results A total of 543 DEGs and 13 hub genes were identified. The correlation analysis between the hub genes and the clinical characteristics of tubular necrosis indicated that the patients with high expression of SPAG5 and BIRC5 had better renal function. Patients with high expression of KIF14, KIF20A, MAD2L1, CKAP2, CDC25C, and CENPEN had poor renal function. Four of those hub genes participate in the cell cycle, apoptosis, and mismatch repair by regulating important genes in the pathway. Conclusions Our study suggests that CDC25C, MAD2L, BIRC5, and EXO1 participate in the cell cycle, apoptosis, and mismatch repair during renal tubule necrosis (RTN) and have an impact on renal function.
Collapse
Affiliation(s)
- Peng Lin
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yongqing Pan
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hang Chen
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Ling Jiang
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Yunhua Liao
- Department of Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
7
|
Khazaei G, Shamsabadi FT, Yamchi A, Golalipour M, Jhingan GD, Shahbazi M. Proteomics evaluation of MDA-MB-231 breast cancer cells in response to RNAi-induced silencing of hPTTG. Life Sci 2019; 239:116873. [PMID: 31521689 DOI: 10.1016/j.lfs.2019.116873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022]
Abstract
AIMS Breast cancer is the most common cancer in women worldwide. Several genes are up-regulated in breast cancer such as human pituitary tumor transforming gene (hPTTG). This study aims to evaluate cell proliferation and the downstream expression pattern of hPTTG1 gene at the mRNA and protein levels after specific down-regulation of hPTTG1 by siRNA. MAIN METHODS The human breast cancer MDA-MB-231 cell line was transfected with siRNA against hPTTG1. The mRNA and protein expression levels were examined by Real-time PCR and Western blot, respectively. The cell proliferation was assayed by MTS. To investigate the pattern of protein expression, total cellular protein was analyzed by 2D gel electrophoresis and mass spectroscopy. Subsequently, the possible biological consequences were determined by the bioinformatics databases. KEY FINDINGS Subsequent of hPTTG1 silencing in the MDA_MB-231 cells, the proliferation of cells decreased obviously. In response to hPTTG1 silencing, the levels mRNA and protein were effectively down-regulated 80% and 50%, respectively, at 48 h post-transfection. The proteomics evidenced that PTTG1 increased the expression of 5 proteins. The reduced expression of PTTG1 was functionally involved in hypoxia (NPM1, ENO1), cell proliferation and apoptosis (ENO1, NPM1, NME1, STMN1), and metastasis (NPM1, NME1). SIGNIFICANCE We identified the hPTTG1-regulated proteins and its molecular mechanism in pathogenesis of breast cancer. Further study emphasis is to understand the association of hPTTG1 with other genes in cancer progression. This novel modality might also consider for identification of targeted drugs, prognosis and follow up in breast cancer gene therapy.
Collapse
Affiliation(s)
- Ghasem Khazaei
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Fatemeh T Shamsabadi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Biotechnology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ahad Yamchi
- Department of Biotechnology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Masoud Golalipour
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Gagan Deep Jhingan
- VProteomics, K-37A, Ground Floor Green Park Main, New Delhi 110016, India
| | - Majid Shahbazi
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; AryaTinaGene Biopharmaceutical Company, Gorgan, Iran.
| |
Collapse
|
8
|
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
|
9
|
Demin DE, Bogolyubova AV, Zlenko DV, Uvarova AN, Deikin AV, Putlyaeva LV, Belousov PV, Mitkin NA, Korneev KV, Sviryaeva EN, Kulakovskiy IV, Tatosyan KA, Kuprash DV, Schwartz AM. The Novel Short Isoform of Securin Stimulates the Expression of Cyclin D3 and Angiogenesis Factors VEGFA and FGF2, but Does Not Affect the Expression of MYC Transcription Factor. Mol Biol 2018. [DOI: 10.1134/s0026893318030032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Xu X, Cao L, Zhang Y, Yin Y, Hu X, Cui Y. Network analysis of DEGs and verification experiments reveal the notable roles of PTTG1 and MMP9 in lung cancer. Oncol Lett 2018; 15:257-263. [PMID: 29387220 PMCID: PMC5768071 DOI: 10.3892/ol.2017.7329] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/02/2017] [Indexed: 01/05/2023] Open
Abstract
Lung cancer, a malignant tumor, is the most frequently fatal cancer, with poor survival rates in the advanced stages. In order to improve the understanding of this disease, and to improve the outcomes of patients, additional studies are required. In the present study, differentially expressed genes (DEGs) in patients with lung cancer compared with controls were identified. To understand how these DEGs act together to account for the initiation of lung cancer, a protein interaction network and a transcriptional regulatory network were constructed to explore the clusters and pathways in lung cancer, and the results indicated that PTTG1 and MMP9 served major roles in the development of lung cancer in the regulatory system. Consistent with this, mRNA and protein expression levels of PTTG1 and MMP9 were significantly upregulated in lung cancer tissues compared with normal lung tissues. The overexpression of PTTG1 or MMP9 was induced in the human bronchial epithelial BEAS-2B cell line, indicating that increased PTTG1 or MMP9 alone may not only facilitate cell migration, proliferation and induce colony formation, but also suppress cell apoptosis. In summary, PTTG1 and MMP9 were identified as potential targets for therapeutic intervention through gene therapy in lung cancer.
Collapse
Affiliation(s)
- Xiaohui Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yan Yin
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Xue Hu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yushang Cui
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
11
|
Elevated PTTG and PBF predicts poor patient outcome and modulates DNA damage response genes in thyroid cancer. Oncogene 2017; 36:5296-5308. [PMID: 28504713 PMCID: PMC5563453 DOI: 10.1038/onc.2017.154] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 04/06/2017] [Accepted: 04/17/2017] [Indexed: 01/15/2023]
Abstract
The proto-oncogene PTTG and its binding partner PBF have been widely studied in multiple cancer types, particularly thyroid and colorectal, but their combined role in tumourigenesis is uncharacterised. Here, we show for the first time that together PTTG and PBF significantly modulate DNA damage response (DDR) genes, including p53 target genes, required to maintain genomic integrity in thyroid cells. Critically, DDR genes were extensively repressed in primary thyrocytes from a bitransgenic murine model (Bi-Tg) of thyroid-specific PBF and PTTG overexpression. Irradiation exposure to amplify p53 levels further induced significant repression of DDR genes in Bi-Tg thyrocytes (P=2.4 × 10-4) compared with either PBF- (P=1.5 × 10-3) or PTTG-expressing thyrocytes (P=NS). Consistent with this, genetic instability was greatest in Bi-Tg thyrocytes with a mean genetic instability (GI) index of 35.8±2.6%, as well as significant induction of gross chromosomal aberrations in thyroidal TPC-1 cells following overexpression of PBF and PTTG. We extended our findings to human thyroid cancer using TCGA data sets (n=322) and found striking correlations with PBF and PTTG expression in well-characterised DDR gene panel RNA-seq data. In addition, genetic associations and transient transfection identified PBF as a downstream target of the receptor tyrosine kinase-BRAF signalling pathway, emphasising a role for PBF as a novel component in a pathway well described to drive neoplastic growth. We also showed that overall survival (P=1.91 × 10-5) and disease-free survival (P=4.9 × 10-5) was poorer for TCGA patients with elevated tumoural PBF/PTTG expression and mutationally activated BRAF. Together our findings indicate that PBF and PTTG have a critical role in promoting thyroid cancer that is predictive of poorer patient outcome.
Collapse
|
12
|
Zhang N, Pati D. Biology and insights into the role of cohesin protease separase in human malignancies. Biol Rev Camb Philos Soc 2017; 92:2070-2083. [PMID: 28177203 DOI: 10.1111/brv.12321] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/05/2017] [Accepted: 01/12/2017] [Indexed: 12/11/2022]
Abstract
Separase, an enzyme that resolves sister chromatid cohesion during the metaphase-to-anaphase transition, plays a pivotal role in chromosomal segregation and cell division. Separase protein, encoded by the extra spindle pole bodies like 1 (ESPL1) gene, is overexpressed in numerous human cancers including breast, bone, brain, and prostate. Separase is oncogenic, and its overexpression is sufficient to induce mammary tumours in mice. Either acute or chronic overexpression of separase in mouse mammary glands leads to aneuploidy and tumorigenesis, and inhibition of separase enzymatic activity decreases the growth of human breast tumour xenografts in mice. This review focuses on the biology of and insights into the molecular mechanisms of separase as an oncogene, and its significance and implications for human cancers.
Collapse
Affiliation(s)
- Nenggang Zhang
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| | - Debananda Pati
- Departments of Pediatrics and Molecular and Cellular Biology, Texas Children's Cancer Center, Baylor College of Medicine, 1102 Bates St., FC1220, Houston, TX 77030, U.S.A
| |
Collapse
|
13
|
Bussey KJ, Bapat A, Linnehan C, Wandoloski M, Dastrup E, Rogers E, Gonzales P, Demeure MJ. Targeting polo-like kinase 1, a regulator of p53, in the treatment of adrenocortical carcinoma. Clin Transl Med 2016; 5:1. [PMID: 26754547 PMCID: PMC4709336 DOI: 10.1186/s40169-015-0080-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is an aggressive cancer with a 5 year survival rate of 20-30 %. Various factors have been implicated in the pathogenesis of ACC including dysregulation of the G2/M transition and aberrant activity of p53 and MDM2. Polo-like kinase 1 (PLK-1) negatively modulates p53 functioning, promotes MDM2 activity through its phosphorylation, and is involved in the G2/M transition. Gene expression profiling of 44 ACC samples showed that increased expression of PLK-1 in 29 % of ACC. Consequently, we examined PLK-1's role in the modulation of the p53 signaling pathway in adrenocortical cancer. METHODS We used siRNA knock down PLK-1 and pharmacological inhibition of PLK-1 and MDM2 ACC cell lines SW-13 and H295R. We examined viability, protein expression, p53 transactivation, and induction of apoptosis. RESULTS Knocking down expression of PLK-1 with siRNA or inhibition of PLK-1 by a small molecule inhibitor, BI-2536, resulted in a loss of viability of up to 70 % in the ACC cell lines H295R and SW-13. In xenograft models, BI-2536 demonstrated marked inhibition of growth of SW-13 with less inhibition of H295R. BI-2536 treatment resulted in a decrease in mutant p53 protein in SW-13 cells but had no effect on wild-type p53 protein levels in H295R cells. Additionally, inhibition of PLK-1 restored wild-type p53's transactivation and apoptotic functions in H295R cells, while these functions of mutant p53 were restored only to a smaller extent. Furthermore, inhibition of MDM2 with nutlin-3 reduced the viability of both the ACC cells and also reactivated wild-type p53's apoptotic function. Inhibition of PLK-1 sensitized the ACC cell lines to MDM2 inhibition and this dual inhibition resulted in an additive apoptotic response in H295R cells with wild-type p53. CONCLUSIONS These preclinical studies suggest that targeting p53 through PLK-1 is an attractive chemotherapy strategy warranting further investigation in adrenocortical cancer.
Collapse
Affiliation(s)
- Kimberly J Bussey
- NantOmics, LLC, The Biodesign Institute, Arizona State University, PO Box 875001, Tempe, AZ, 85287-5001, USA.
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Aditi Bapat
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Claire Linnehan
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | | | - Erica Dastrup
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Erik Rogers
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
| | - Paul Gonzales
- Translational Drug Development (TD2), Scottsdale, AZ, USA.
| | | |
Collapse
|
14
|
Zhang H, Xu C, Sun N, Zhou Y, Yu X, Yan X, Zhang Q. Gene expression profiling analysis of MENX-associated rat pituitary adenomas contributes to understand molecular mechanisms of human pituitary adenomas. Oncol Lett 2015; 11:125-133. [PMID: 26870179 PMCID: PMC4727179 DOI: 10.3892/ol.2015.3904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 07/10/2015] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to screen potential genes associated with pituitary adenomas to obtain further understanding with regard to the pathogenesis of pituitary adenomas. The microarray GSE23207 dataset, containing 16 pituitary adenoma samples from multiple endocrine neoplasia syndrome-associated rats and 5 normal pituitary tissue samples, was downloaded from Gene Expression Omnibus. The Linear Models for Microarray Data package was used to identify the differentially-expressed genes (DEGs) with the cut-off criteria of a |log2fold change (FC)|>1 and adjusted P-values of <0.05. The potential functions of the DEGs were predicted by functional and pathway enrichment analysis with the Database for Annotation, Visualization and Integrated Discovery. Furthermore, the interaction associations of the up- and downregulated DEGs obtained from the Search Tool for the Retrieval of Interacting Genes database were respectively revealed by the protein-protein interaction networks visualized with Cytoscape. A total of 391 upregulated and 238 downregulated DEGs in were screened in the pituitary adenoma samples. The upregulated DEGs with a higher degree in the protein-protein interaction network (e.g., CCNA2, CCNB1 and CDC20) were significantly involved in cell cycle and cell division. Notably, PTTG1 was enriched in every functional term. These DEGs interacted with each other. The downregulated DEGs (e.g., GABRA1, GABRA4 and GABRB1) also interacted with each other, and were relevant to neuroactive ligand-receptor interaction; the DEG POU1F1, interacting with POMC, was correlated with the development of the pituitary gland, adenohypophysis and endocrine system. Certain DEGs, including CCNB1, CCNA2, CDC20, GABRA1, GABRA4, GABRB1, POU1F1 and POMC, and particularly PTTG1, were shown to be closely involved in the pathogenesis of pituitary adenomas.
Collapse
Affiliation(s)
- Hongzhi Zhang
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Chuan Xu
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Ningyang Sun
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Yinting Zhou
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Xiaofei Yu
- Department of Neurology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Xue Yan
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| | - Qiujuan Zhang
- Department of Neurology, Yueyang Hospital of Integrated Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200437, P.R. China
| |
Collapse
|
15
|
Noll JE, Vandyke K, Hewett DR, Mrozik KM, Bala RJ, Williams SA, Kok CH, Zannettino AC. PTTG1 expression is associated with hyperproliferative disease and poor prognosis in multiple myeloma. J Hematol Oncol 2015; 8:106. [PMID: 26445238 PMCID: PMC4595141 DOI: 10.1186/s13045-015-0209-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/28/2015] [Indexed: 01/08/2023] Open
Abstract
Background Multiple myeloma (MM) is an incurable haematological malignancy characterised by the clonal proliferation of malignant plasma cells within the bone marrow. We have previously identified pituitary tumour transforming gene 1 (Pttg1) as a gene that is significantly upregulated in the haematopoietic compartment of the myeloma-susceptible C57BL/KaLwRij mouse strain, when compared with the myeloma-resistant C57BL/6 mouse. Over-expression of PTTG1 has previously been associated with malignant progression and an enhanced proliferative capacity in solid tumours. Methods In this study, we investigated PTTG1 gene and protein expression in MM plasma cells from newly diagnosed MM patients. Gene expression profiling was used to identify gene signatures associated with high PTTG1 expression in MM patients. Additionally, we investigated the effect of short hairpin ribonucleic acid (shRNA)-mediated PTTG1 knockdown on the proliferation of the murine myeloma plasma cell line 5TGM1 in vitro and in vivo. Results PTTG1 was found to be over-expressed in 36–70 % of MM patients, relative to normal controls, with high PTTG1 expression being associated with poor patient outcomes (hazard ratio 2.49; 95 % CI 1.28 to 4.86; p = 0.0075; log-rank test). In addition, patients with high PTTG1 expression exhibited increased expression of cell proliferation-associated genes including CCNB1, CCNB2, CDK1, AURKA, BIRC5 and DEPDC1. Knockdown of Pttg1 in 5TGM1 cells decreased cellular proliferation, without affecting cell cycle distribution or viability, and decreased expression of Ccnb1, Birc5 and Depdc1 in vitro. Notably, Pttg1 knockdown significantly reduced MM tumour development in vivo, with an 83.2 % reduction in tumour burden at 4 weeks (p < 0.0001, two-way ANOVA). Conclusions This study supports a role for increased PTTG1 expression in augmenting tumour development in a subset of MM patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0209-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jacqueline E Noll
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Kate Vandyke
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,SA Pathology, Adelaide, Australia.
| | - Duncan R Hewett
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Krzysztof M Mrozik
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Rachel J Bala
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Sharon A Williams
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia.
| | - Chung H Kok
- Leukaemia Research Group, Cancer Theme, SAHMRI, Adelaide, Australia.
| | - Andrew Cw Zannettino
- Myeloma Research Laboratory, Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide and Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia. .,Discipline of Physiology, School of Medicine, Faculty of Health Sciences, University of Adelaide, Cancer Theme, Level 5 South, SAHMRI, PO Box 11060, Adelaide, SA, 5001, Australia.
| |
Collapse
|
16
|
Zheng Y, Guo J, Zhou J, Lu J, Chen Q, Zhang C, Qing C, Koeffler HP, Tong Y. FoxM1 transactivates PTTG1 and promotes colorectal cancer cell migration and invasion. BMC Med Genomics 2015; 8:49. [PMID: 26264222 PMCID: PMC4534164 DOI: 10.1186/s12920-015-0126-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 07/31/2015] [Indexed: 12/13/2022] Open
Abstract
Background Metastasis is the major cause of cancer-related death. Forkhead Box M1 (FoxM1) is a master regulator of tumor metastasis. This study aims to identify new FoxM1 targets in regulating tumor metastasis using bioinformatics tools as well as biological experiments. Methods Illumina microarray was used to profile WT and PTTG1 knockout HCT116 cells. R2 Genomics Analysis was used to identify PTTG1 as a potential FoxM1 targeted gene. Luciferase reporter array, EMSA and Chromatin Immunoprecipitation (ChIP) were used to determine the binding of FoxM1 to PTTG1 promoter. Boyden chamber assay was used to evaluate the effects of FoxM1-PTTG1 on cell migration and invasion. Splenic-injection induced liver metastasis model was used to evaluate the effects of FoxM1-PTTG1 on liver metastasis of colorectal cancer. Results Analyses of multiple microarray datasets derived from human colorectal cancer indicated that correlation levels of FoxM1 and pituitary tumor transforming gene (PTTG1) are highly concordant (R = 0.68 ~ 0.89, p = 2.1E-226 ~ 9.6E-86). FoxM1 over-expression increased and knock-down decreased PTTG1 expression. Luciferase reporter assay identified that the −600 to −300 bp region of PTTG1 promoter is important for FoxM1 to enhance PTTG1 promoter activity. EMSA and ChIP assays confirmed that FoxM1 directly binds to PTTG1 promoter at the −391 to −385 bp region in colorectal cancer cells. Boyden chamber assay indicated that both FoxM1 and PTTG1 regulate migration and invasion of HCT116 and SW620 colorectal cancer cells. Further in vivo assays indicated that PTTG1 knock out decreased the liver metastasis of FoxM1 over-expressing HCT116 cells. Microarray analyses identified 662 genes (FDR < 0.05) differentially expressed between WT and PTTG1−/− HCT116 cells. Among them, dickkopf homolog 1 (DKK1), a known WNT pathway inhibitor, was suppressed by PTTG1 and FoxM1. Conclusions PTTG1 is a FoxM1 targeted gene. FoxM1 binds to PTTG1 promoter to enhance PTTG1 transcription, and FoxM1-PTTG1 pathway promotes colorectal cancer migration and invasion. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0126-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yun Zheng
- Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Room 3021, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA. .,Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Jinjun Guo
- Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Room 3021, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA. .,Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jin Zhou
- Division of Epidemiology and Biostatistics, College of Public Health, University of Arizona, Tucson, AZ, USA.
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Qi Chen
- Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Room 3021, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.
| | - Cui Zhang
- Department of Pathology, Xinxiang Medical University, 601 East Jinsui Ave, Xinxiang, Henan, China.
| | - Chen Qing
- School of Pharmaceutical Science, Kunming Medical University, 1168 Western Chunrong Road,Yuhua Street, Chenggong New City, Kunming, China.
| | - H Philip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Room 3021, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.
| | - Yunguang Tong
- Department of Medicine, Cedars-Sinai Medical Center, UCLA School of Medicine, Room 3021, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA. .,Department of Pathology, Xinxiang Medical University, 601 East Jinsui Ave, Xinxiang, Henan, China.
| |
Collapse
|
17
|
Hellmuth S, Pöhlmann C, Brown A, Böttger F, Sprinzl M, Stemmann O. Positive and negative regulation of vertebrate separase by Cdk1-cyclin B1 may explain why securin is dispensable. J Biol Chem 2015; 290:8002-10. [PMID: 25659430 DOI: 10.1074/jbc.m114.615310] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sister chromatid cohesion is established during replication by entrapment of both dsDNAs within the cohesin ring complex. It is dissolved in anaphase when separase, a giant cysteine endopeptidase, cleaves the Scc1/Rad21 subunit of cohesin, thereby triggering chromosome segregation. Separase is held inactive by association with securin until this anaphase inhibitor is destroyed at the metaphase-to-anaphase transition by ubiquitin-dependent degradation. The relevant ubiquitin ligase, the anaphase-promoting complex/cyclosome, also targets cyclin B1, thereby causing inactivation of Cdk1 and mitotic exit. Although separase is essential, securin knock-out mice are surprisingly viable and fertile. Capitalizing on our previous finding that Cdk1-cyclin B1 can also bind and inhibit separase, we investigated whether this kinase might be suitable to maintain faithful timing and execution of anaphase in the absence of securin. We found that, similar to securin, Cdk1-cyclin B1 regulates separase in both a positive and negative manner. Although securin associates with nascent separase to co-translationally assist proper folding, Cdk1-cyclin B1 acts on native state separase. Upon entry into mitosis, Cdk1-cyclin B1-dependent phosphorylation of Ser-1126 renders separase prone to inactivation by aggregation/precipitation. Stable association of Cdk1-cyclin B1 with phosphorylated separase counteracts this tendency and stabilizes separase in an inhibited yet activatable state. These opposing effects are suited to prevent premature cleavage of cohesin in early mitosis while ensuring timely activation of separase by anaphase-promoting complex/cyclosome-dependent degradation of cyclin B1. Coupling sister chromatid separation with subsequent exit from mitosis by this simplified mode might have been the common scheme of mitotic control prior to the evolution of securin.
Collapse
Affiliation(s)
| | | | | | | | - Mathias Sprinzl
- Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany
| | | |
Collapse
|
18
|
Castilla C, Flores ML, Medina R, Pérez-Valderrama B, Romero F, Tortolero M, Japón MA, Sáez C. Prostate cancer cell response to paclitaxel is affected by abnormally expressed securin PTTG1. Mol Cancer Ther 2014; 13:2372-83. [PMID: 25122070 DOI: 10.1158/1535-7163.mct-13-0405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PTTG1 protein, the human securin, has a central role in sister chromatid separation during mitosis, and its altered expression has been reported in many tumor types. Paclitaxel is a widely used chemotherapeutic drug, whose mechanism of action is related to its ability to arrest cells in mitosis and the subsequent induction of the intrinsic apoptotic pathway. By using two prostate cancer cell lines with different responses to paclitaxel treatment, we have identified two situations in which PTTG1 influences cell fate differentially. In slippage-prone PC3 cells, both PTTG1 downregulation and overexpression induce an increase in mitotic cells that is associated with diminished apoptosis after paclitaxel treatment. In LNCaP cells, however, PTTG1 downregulation prevents mitotic entry and, subsequently, inhibits mitosis-associated, paclitaxel-induced apoptosis. In contrast, PTTG1 overexpression induces an increase in mitotic cells and apoptosis after paclitaxel treatment. We have also identified a role for Mcl-1 protein in preventing apoptosis during mitosis in PC3 cells, as simultaneous PTTG1 and Mcl-1 silencing enhances mitosis-associated apoptosis after paclitaxel treatment. The finding that a more efficient mitotic arrest alone in PC3 cells is not enough to increase apoptosis was also confirmed with the observation that a selected paclitaxel-resistant PC3 cell line showed an apoptosis-resistant phenotype associated with increased mitosis upon paclitaxel treatment. These findings could contribute to identify putative responsive and nonresponsive cells and help us to approach incomplete responses to paclitaxel in the clinical setting.
Collapse
Affiliation(s)
- Carolina Castilla
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - M Luz Flores
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Rafael Medina
- Department of Urology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Francisco Romero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - María Tortolero
- Department of Microbiology, Faculty of Biology, Universidad de Sevilla, Seville, Spain
| | - Miguel A Japón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| | - Carmen Sáez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain. Department of Pathology, Hospital Universitario Virgen del Rocío, Seville, Spain.
| |
Collapse
|
19
|
Gene expression profiling and pathway analysis of hepatotoxicity induced by triptolide in Wistar rats. Food Chem Toxicol 2013; 58:495-505. [DOI: 10.1016/j.fct.2013.04.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/19/2013] [Accepted: 04/22/2013] [Indexed: 12/30/2022]
|
20
|
TFE2 and GATA3 enhance induction of POU4F3 and myosin VIIa positive cells in nonsensory cochlear epithelium by ATOH1. Dev Biol 2012; 372:68-80. [PMID: 22985730 DOI: 10.1016/j.ydbio.2012.09.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 08/01/2012] [Accepted: 09/08/2012] [Indexed: 11/21/2022]
Abstract
Transcription factors (TFs) can regulate different sets of genes to determine specific cell types by means of combinatorial codes. We previously identified closely-spaced TF binding motifs located 8.2-8.5 kb 5' to the ATG of the murine Pou4f3 gene, a gene required for late hair cell (HC) differentiation and survival. These motifs, 100% conserved among four mammalian species, include a cluster of E-boxes preferred by TCF3/ATOH1 heterodimers as well as motifs for GATA factors and SP1. We hypothesized that these factors might interact to regulate the Pou4f3 gene and possibly induce a HC phenotype in non-sensory cells of the cochlea. Cochlear sensory epithelium explants were prepared from postnatal day 1.5 transgenic mice in which expression of GFP is driven by 8.5 kb of Pou4f3 5' genomic DNA (Pou4f3/GFP). Electroporation was used to transfect cells of the greater epithelial ridge with multiple plasmids encoding human ATOH1 (hATOH1), hTCF3 (also known as E2A or TEF2), hGATA3, and hSP1. hATOH1 or hTCF3 alone induced Pou4f3/GFP cells but hGATA3 and hSP1 did not. hATOH1 but not hTCF3 induced conversion of greater epithelial ridge cells into Pou4f3/GFP and myosin VIIa double-positive cells. Transfection of hATOH1 in combination with hTCF3 or hGATA3 induced 2-3X more Pou4f3/GFP cells, and similarly enhanced Pou4f3/GFP and myosin VIIa double-positive cells, when compared to hATOH1 alone. Triple or quadruple TF combinations were generally not more effective than double TF combinations except in the middle turn, where co-transfection of hATOH1, hE2A, and hGATA3 was more effective than hATOH1 plus either hTCF3 or hGATA3. The results demonstrate that TFs can cooperate in regulation of the Pou4f3 gene and in the induction of at least one other element of a HC phenotype. Our data further indicate that combinations of TFs can be more effective than individual TFs in the inner ear.
Collapse
|
21
|
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
|
22
|
Karra H, Pitkänen R, Nykänen M, Talvinen K, Kuopio T, Söderström M, Kronqvist P. Securin predicts aneuploidy and survival in breast cancer. Histopathology 2012; 60:586-96. [DOI: 10.1111/j.1365-2559.2011.04107.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Association of Vpu with hepatitis C virus NS3/4A stimulates transcription of type 1 human immunodeficiency virus. Virus Res 2012; 163:74-81. [DOI: 10.1016/j.virusres.2011.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/07/2011] [Accepted: 08/19/2011] [Indexed: 01/27/2023]
|
24
|
Regulation of POU4F3 gene expression in hair cells by 5' DNA in mice. Neuroscience 2011; 197:48-64. [PMID: 21958861 DOI: 10.1016/j.neuroscience.2011.09.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 09/12/2011] [Accepted: 09/13/2011] [Indexed: 01/21/2023]
Abstract
The POU-domain transcription POU4F3 is expressed in the sensory cells of the inner ear. Expression begins shortly after commitment to the hair cell (HC) fate, and continues throughout life. It is required for terminal HC differentiation and survival. To explore regulation of the murine Pou4f3 gene, we linked enhanced green fluorescent protein (eGFP) to 8.5 kb of genomic sequence 5' to the start codon in transgenic mice. eGFP was uniformly present in all embryonic and neonatal HCs. Expression of eGFP was also observed in developing Merkel cells and olfactory neurons as well as adult inner and vestibular HCs, mimicking the normal expression pattern of POU4F3 protein, with the exception of adult outer HCs. Apparently ectopic expression was observed in developing inner ear neurons. On a Pou4f3 null background, the transgene produced expression in embryonic HCs which faded soon after birth both in vivo and in vitro. Pou4f3 null HCs treated with caspase 3 and 9 inhibitors survived longer than untreated HCs, but still showed reduced expression of eGFP. The results suggest the existence of separate enhancers for different HC types, as well as strong autoregulation of the Pou4f3 gene. Bioinformatic analysis of four divergent mammalian species revealed three highly conserved regions within the transgene: 400 bp immediately 5' to the Pou4f3 ATG, a short sequence at -1.3 kb, and a longer region at -8.2 to -8.5 kb. The latter contained E-box motifs that bind basic helix-loop-helix (bHLH) transcription factors, including motifs activated by ATOH1. Cotransfection of HEK293 or VOT-E36 cells with ATOH1 and the transgene as a reporter enhanced eGFP expression when compared with the transgene alone. Chromatin immunoprecipitation of the three highly conserved regions revealed binding of ATOH1 to the distal-most conserved region. The results are consistent with regulation of Pou4f3 in HCs by ATOH1 at a distal enhancer.
Collapse
|
25
|
Chen S, Xiao L, Liu Z, Liu J, Liu Y. Pituitary tumor transforming gene-1 haplotypes and risk of pituitary adenoma: a case-control study. BMC MEDICAL GENETICS 2011; 12:44. [PMID: 21439054 PMCID: PMC3078851 DOI: 10.1186/1471-2350-12-44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 03/25/2011] [Indexed: 11/10/2022]
Abstract
Background It has been suggested that pituitary adenoma results from accumulation of multiple genetic and/or epigenetic aberrations, which may be identified through association studies. As pituitary tumor transforming gene-1 (PTTG1)/securin plays a critical role in promoting genomic instability in pituitary neoplasia, the present study explored the association of PTTG1 haplotypes with the risk of pituitary adenoma. Methods We genotyped five PTTG1 haplotype-tagging SNPs (htSNP) by PCR-RFLP assays in a case-control study, which included 280 Han Chinese patients diagnosed with pituitary adenoma and 280 age-, gender- and geographically matched Han Chinese controls. Haplotypes were reconstructed according to the genotyping data and linkage disequilibrium status of the htSNPs. Results No significant differences in allele and genotype frequencies of the htSNPs were observed between pituitary adenoma patients and controls, indicating that none of the individual PTTG1 SNPs examined in this study is associated with the risk of pituitary adenoma. In addition, no significant association was detected between the reconstructed PTTG1 haplotypes and pituitary adenoma cases or the controls. Conclusions Though no significant association was found between PTTG1 haplotypes and the risk of pituitary adenoma, this is the first report on the association of individual PTTG1 SNPs or PTTG1 haplotypes with the risk of pituitary adenoma based on a solid study; it will provide an important reference for future studies on the association between genetic alterations in PTTG1 and the risk of pituitary adenoma or other tumors.
Collapse
Affiliation(s)
- Shuai Chen
- Gamma Knife Treatment and Research Center, Xiangya Hospital, Central South University, Changsha, 410008 PR China
| | | | | | | | | |
Collapse
|
26
|
Niemantsverdriet M, de Jong E, Langendijk JA, Kampinga HH, Coppes RP. Synergistic induction of profibrotic PAI-1 by TGF-β and radiation depends on p53. Radiother Oncol 2011; 97:33-5. [PMID: 20435362 DOI: 10.1016/j.radonc.2010.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 03/30/2010] [Accepted: 04/05/2010] [Indexed: 11/16/2022]
Abstract
Radiation-induced fibrosis is a severe side effect of radiotherapy. TGF-β and radiation synergistically induce expression of the profibrotic PAI-1 gene and this cooperation potentially involves p53. Here, we demonstrate that p53 is both indispensable and sufficient for the radiation effect inducing synergistic activation of PAI-1 by radiation and TGF-β.
Collapse
Affiliation(s)
- Maarten Niemantsverdriet
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
27
|
Jiang RH, Su WC, Liu HF, Huang HS, Chao JI. Opposite expression of securin and γ-H2AX regulates baicalein-induced cancer cell death. J Cell Biochem 2011; 111:274-83. [PMID: 20506293 DOI: 10.1002/jcb.22697] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Securin and γ-H2AX have been shown to regulate cell survival and genomic stability. However, it is still unknown how the expression and regulation of these proteins is altered following treatment with baicalein, a natural flavonoid extracted from the Scutellaria baicalensis root. In the present study, we investigate the possible roles of securin and γ-H2AX in baicalein-induced cancer cell death. Baicalein reduced cell viability in a variety of human cancer cell lines, including bladder, cervical, colon, and lung cancer cells. Interestingly, baicalein treatment (40-80 µM for 24 h) markedly inhibited securin expression, while the levels of γ-H2AX were elevated. Abnormal spindle formation and chromosomal segregation were induced by baicalein. Furthermore, wild type HCT116 cancer cells had a higher incidence of cytotoxicity and apoptosis than securin-null HCT116 cells following treatment with baicalein. In contrast, baicalein increased the levels of γ-H2AX to a similar extent in both cell types. Transfection with H2AX siRNA further increased baicalein-induced cell death. Additionally, blockade of the AKT pathway by treatment with wortmannin or AKT shRNA lowered the levels of γ-H2AX and enhanced cytotoxicity in baicalein-treated cells. Taken together, our findings suggest that the opposing effects of baicalein on securin and γ-H2AX levels may be involved in the regulation of cell viability and genomic stability by this compound.
Collapse
Affiliation(s)
- Ren-Huei Jiang
- Molecular Anticancer Laboratory, Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30068, Taiwan
| | | | | | | | | |
Collapse
|
28
|
Abstract
The pituitary tumor-transforming gene (PTTG1) encodes a multifunctional protein (PTTG) that is overexpressed in numerous tumours, including pituitary, thyroid, breast and ovarian carcinomas. PTTG induces cellular transformation in vitro and tumourigenesis in vivo, and several mechanisms by which PTTG contributes to tumourigenesis have been investigated. Also known as the human securin, PTTG is involved in cell cycle regulation, controlling the segregation of sister chromatids during mitosis. This review outlines current information regarding PTTG structure, expression, regulation and function in the pathogenesis of neoplasia. Recent progress concerning the use of PTTG as a prognostic marker or therapeutic target will be considered. In addition, the PTTG binding factor (PBF), identified through its interaction with PTTG, has also been established as a proto-oncogene that is upregulated in several cancers. Current knowledge regarding PBF is outlined and its role both independently and alongside PTTG in endocrine and related cancers is discussed.
Collapse
|
29
|
Tesfaye D, Regassa A, Rings F, Ghanem N, Phatsara C, Tholen E, Herwig R, Un C, Schellander K, Hoelker M. Suppression of the transcription factor MSX1 gene delays bovine preimplantation embryo development in vitro. Reproduction 2010; 139:857-70. [DOI: 10.1530/rep-09-0312] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study was conducted to investigate the effect of suppressing transcription factor geneMSX1on the development ofin vitroproduced bovine oocytes and embryos, and identify its potential target genes regulated by this gene. Injection of long double-stranded RNA (LdsRNA) and small interfering RNA (siRNA) at germinal vesicle stage oocyte reducedMSX1mRNA expression by 73 and 37% respectively at metaphase II stage compared with non-injected controls. Similarly, injection of the same anti-sense oligomers at zygote stage reducedMSX1mRNA expression by 52 and 33% at 8-cell stage compared with non-injected controls. Protein expression was also reduced in LdsRNA- and siRNA-injected groups compared with non-injected controls at both stages. Blastocysts rates were 33, 28, 20 and 18% in non-injected control, scrambled RNA (scRNA), LdsRNA- and siRNA-injected groups respectively. Cleavage rates were also significantly reduced in Smartpool siRNA (SpsiRNA)-injected group (53.76%) compared with scRNA-injected group (57.76%) and non-injected control group (61%). Large-scale gene expression analysis showed that 135 genes were differentially regulated in SpsiRNA-injected group compared with non-injected controls, of which 54 and 81 were down- and up-regulated respectively due to suppression ofMSX1. Additionally, sequence homology mapping and gene enrichment analysis with known human pathway information identified several functional modules that were affected due to suppression ofMSX1. In conclusion, suppression ofMSX1affects oocyte maturation, embryo cleavage rate and the expression of several genes, suggesting its potential role in the development of bovine preimplantation embryos.
Collapse
|
30
|
Salehi F, Kovacs K, Scheithauer BW, Cantelmi D, Horvath E, Lloyd RV, Cusimano M. Immunohistochemical expression of pituitary tumor transforming gene (PTTG) in pituitary adenomas: a correlative study of tumor subtypes. Int J Surg Pathol 2010; 18:5-13. [PMID: 20106827 DOI: 10.1177/1066896909356105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE We investigated the correlation between immunohistochemical expression of the pituitary tumor transforming gene (PTTG) and pituitary adenoma subtype. METHODS Pituitary adenomas (n = 89) were stained for PTTG using the streptavidin-biotin-peroxidase complex method and a monoclonal PTTG antibody. RESULTS PTTG staining was found to be cytoplasmic with a pronounced paranuclear expression pattern. Reactivity was highest in growth hormone (GH) adenomas as compared with other tumors, including prolactin (PRL), follicle-stimulating hormone/luteinizing hormone/alpha subunit, as well as adrenocorticotrophic hormone-secreting adenomas. PRL adenomas exhibited the lowest expression levels. Among GH adenomas, untreated tumors demonstrated significantly higher PTTG levels than octreotide-treated examples. Although dopamine agonist-treated PRL adenomas tended to show lower expression levels, statistical significance was not reached. CONCLUSIONS Our finding that PTTG was differentially expressed in pituitary adenoma subtypes suggests a cell-specific function for PTTG. Moreover, treatment of GH adenomas with somatostatin analogues lowered PTTG expression. Further investigation into mechanisms mediating cell-specific expression of PTTG is warranted.
Collapse
Affiliation(s)
- Fateme Salehi
- St Michael's Hospital, University of Toronto, Toronto, ON, Canada.
| | | | | | | | | | | | | |
Collapse
|
31
|
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]
|
32
|
Panguluri SK, Kakar SS. Effect of PTTG on endogenous gene expression in HEK 293 cells. BMC Genomics 2009; 10:577. [PMID: 19958546 PMCID: PMC2793268 DOI: 10.1186/1471-2164-10-577] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 12/03/2009] [Indexed: 11/27/2022] Open
Abstract
Background Pituitary tumor transforming gene (PTTG), also known as securin, is highly expressed in various tumors including pituitary, thyroid, colon, ovary, testis, lung, and breast. An overexpression of PTTG enhances cell proliferation, induces cellular transformation in vitro, and promotes tumor development in nude mice. PTTG also inhibits separation of sister chromatids leading to aneuploidy and genetic instability. A great amount of work has been undertaken to understand the biology of PTTG and its expression in various tumors. However, mechanisms by which PTTG mediates its tumorigenic function are not fully understood. To utilize this gene for cancer therapy, identification of the downstream signaling genes regulated by PTTG in mediation of its tumorigenic function is necessary. For this purpose, we expressed PTTG in human embryonic kidney (HEK293) cells that do not express PTTG and analyzed the downstream genes using microarray analysis. Results A total of 22,277 genes printed on an Affymetrix HG-U133A 2.0 GeneChip™ array were screened with labeled cRNA prepared from HEK293 cells infected with adenovirus vector expressing PTTG cDNA (AdPTTG cDNA) and compared with labeled cRNA prepared from HEK293 cells infected with control adenovirus (control Ad) or adenovirus vector expressing GFP (AdGFP). Out of 22,277 genes, 71 genes were down-regulated and 35 genes were up-regulated with an FDR corrected p-value of ≤ 0.05 and a fold change of ≥2. Most of the altered genes identified are involved in the cell cycle and cell apoptosis; a few are involved in mRNA processing and nitrogen metabolism. Most of the up-regulated genes belong to the histone protein family. Conclusion PTTG is a well-studied oncogene for its role in tumorigenesis. In addition to its importance in regulation of the cell cycle, this gene has also been recently shown to play a role in the induction of cell apoptosis. The microarray analysis in the present study demonstrated that PTTG may induce apoptosis by down-regulation of oncogenes such as v-Jun and v-maf and up-regulation of the histone family of genes.
Collapse
Affiliation(s)
- Siva K Panguluri
- Department of Physiology and Biophysics, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| | | |
Collapse
|
33
|
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
|
34
|
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]
|
35
|
Tena-Suck ML, Ortiz-Plata A, Galán F, Sánchez A. Expression of epithelial cell adhesion molecule and pituitary tumor transforming gene in adamantinomatous craniopharyngioma and its correlation with recurrence of the tumor. Ann Diagn Pathol 2009; 13:82-8. [DOI: 10.1016/j.anndiagpath.2008.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Csizmok V, Felli IC, Tompa P, Banci L, Bertini I. Structural and dynamic characterization of intrinsically disordered human securin by NMR spectroscopy. J Am Chem Soc 2009; 130:16873-9. [PMID: 19053469 DOI: 10.1021/ja805510b] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Understanding the molecular action of securin, the inhibitor of separase in mitosis, is of immense theoretical and biomedical importance. The residue-level structural description of an intrinsically disordered protein of this length (202 amino acids, containing 24 prolines), however, represents a particular challenge. Here we combined (1)H-detected and (13)C-detected protonless NMR experiments to achieve full assignment of securin's backbone amide resonances. Chemical shifts, (15)N relaxation rates (R(1), R(2), (1)H-(15)N NOEs), (1)H exchange rates with the solvent (CLEANEX-PM), and (1)H-(15)N residual dipolar couplings were determined along the entire length of the protein. This analysis showed that securin is not entirely disordered, but segregates into a largely disordered N-terminal half and a C-terminal half with transient segmental order, within which the segment D(150)-F(159) has a significant helical tendency and segments E(113)-S(127) and W(174)-L(178) also show a significant deviation from random-coil behavior. These results, in combination with bioinformatic and biochemical data on the securin/separase interaction, shed light on the inhibitory action of securin on separase.
Collapse
Affiliation(s)
- Veronika Csizmok
- Institute of Enzymology, Biological Research Center, Hungarian Academy of Sciences, Budapest, Karolina ut 29, H-1113, Hungary
| | | | | | | | | |
Collapse
|
37
|
Panigrahi AK, Pati D. Road to the crossroads of life and death: linking sister chromatid cohesion and separation to aneuploidy, apoptosis and cancer. Crit Rev Oncol Hematol 2009; 72:181-93. [PMID: 19162508 DOI: 10.1016/j.critrevonc.2008.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 12/02/2008] [Accepted: 12/11/2008] [Indexed: 01/22/2023] Open
Abstract
Genomic instability, aberrant cell proliferation and defects in apoptotic cell death are critical issues in cancer. The two most prominent hallmarks of cancer cells are multiple mutations in key genes encoding proteins that regulate important cell-survival pathways, and marked restructuring or redistribution of the chromosomes (aneuploidy) indicative of genomic instability. Both these aspects have been suggested to cause cancer, though a causal role for chromosomal restructuring in tumorigenesis has not been experimentally fully substantiated. This review is aimed at understanding the mechanisms of cell cycle (proliferation) and programmed cell death (apoptosis) and chromosomal instability governed by cohesin and other aneuploidy promoters, which will provide new insights into the process of carcinogenesis and new avenues for targeted treatment.
Collapse
Affiliation(s)
- Anil K Panigrahi
- Department of Pediatric Hematology/Oncology, Texas Children's Cancer Center, Baylor College of Medicine, 6621 Fannin St., MC3-3320, Houston, TX 77030, USA
| | | |
Collapse
|
38
|
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. [PMID: 18217976 DOI: 10.1111/j.1525-1438.2007.01168.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
Affiliation(s)
- J W Kim
- Department of Obstetrics and Gynecology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Panguluri SK, Yeakel C, Kakar SS. PTTG: an important target gene for ovarian cancer therapy. J Ovarian Res 2008; 1:6. [PMID: 19014669 PMCID: PMC2584053 DOI: 10.1186/1757-2215-1-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 10/20/2008] [Indexed: 12/13/2022] Open
Abstract
Pituitary tumor transforming gene (PTTG), also known as securin is an important gene involved in many biological functions including inhibition of sister chromatid separation, DNA repair, organ development, and expression and secretion of angiogenic and metastatic factors. Proliferating cancer cells and most tumors express high levels of PTTG. Overexpression of PTTG in vitro induces cellular transformation and development of tumors in nude mice. The PTTG expression levels have been correlated with tumor progression, invasion, and metastasis. Recent studies show that down regulation of PTTG in tumor cell lines and tumors in vivo results in suppression of tumor growth, suggesting its important role in tumorigenesis. In this review, we focus on PTTG structure, sub-cellular distribution, cellular functions, and role in tumor progression with suggestions on possible exploration of this gene for cancer therapy.
Collapse
Affiliation(s)
- Siva Kumar Panguluri
- Department of Physiology and Biophysics, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | |
Collapse
|
40
|
Sak A, Fegers I, Groneberg M, Stuschke M. Effect of separase depletion on ionizing radiation-induced cell cycle checkpoints and survival in human lung cancer cell lines. Cell Prolif 2008; 41:660-70. [PMID: 18616699 PMCID: PMC6496864 DOI: 10.1111/j.1365-2184.2008.00540.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2007] [Accepted: 11/26/2007] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES This study is to evaluate the effect of separase depletion on cell cycle progression of irradiated and non-irradiated cells through the G(2)/M phases and consecutive cell survival. MATERIALS AND METHODS Separase was depleted with siRNA in two human non-small cell lung carcinoma (NSCLC) cell lines. Cell cycle progression, mitotic fraction, DNA repair, apoptotic and clonogenic cell death were determined. RESULTS By depletion of endogenous separase with siRNA in NSCLCs, we showed that separase affects progression through the G(2) phase. In non-irradiated exponentially growing cells, separase depletion led to an increased G(2) accumulation from 17.2% to 29.1% in H460 and from 15.7% to 30.9% in A549 cells and a decrease in mitotic cells. Depletion of separase significantly (P < 0.01) increased the fraction of radiation-induced G(2) arrested cells 30-56 h after irradiation and led to decrease in the mitotic fraction. This was associated with increased double-strand break repair as measured by gamma-H2AX foci kinetics in H460 cells and to a lesser extent in A549 cells. In addition, a decrease in the expression of mitotic linked cell death after irradiation was found. CONCLUSIONS These results indicate that separase has additional targets involved in regulation of G(2) to M progression after DNA damage. Prolonged G(2) phase arrest in the absence of separase has consequences on repair of damaged DNA and cell death.
Collapse
Affiliation(s)
- A Sak
- Department of Radiotherapy, University Hospital Essen, Essen, Germany.
| | | | | | | |
Collapse
|
41
|
Wu BW, Ma D, Li YJ, Li DF, Deng G, Zheng ZG, Zhang KJ, Zhang YH, Geng QS. Expression of human pituitary tumor transforming gene 1 in colorectal cancer and its clinical significance. Shijie Huaren Xiaohua Zazhi 2008; 16:2248-2252. [DOI: 10.11569/wcjd.v16.i20.2248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigated the expression of human pituitary tumor transforming gene 1 (hPTTG1) protein in colorectal cancerous tissues and to analyze its relationship with clinicopathological parameters so as to gain more experimental evidence for hPTTG1's clinical value in colorectal cancer diagnosis and relapse.
METHODS: Sixty colorectal cancer samples and their corresponding noncancerous tissues were examined for hPTTG1 protein expression using immunohistochemical staining and Western blotting analysis. Subsequently, we also studied the relationship between seven clinicopathological parameters and hPTTG1 protein expression.
RESULTS: Of 60 colorectal tissue samples, 50 samples of hPTTG1 protein expression were positive (93.3%), but only 8 corresponding noncancerous tissues were positive and all weakly positive (13.3%) (c2 = 77.13, P < 0.001). There was a significant correlation between hPTTG1 mRNA expression and serum CEA level, and CEA ≥ 5 mg/L was significantly higher than CEA < 5 mg/L (c2 = 30.886, P < 0.001); hPTTG1 gene was expressed significantly higher in Dukes C, D stages than in Dukes A, B stages (c2 = 9.87, P < 0.001), and significantly higher in colorectal cancer with the lymph node invasion, liver or other organs metastasis than no invasion (c2 = 9.87, P < 0.001). The hPTTG1 expression was not correlated with age, sex, tumor size or histologic types.
CONCLUSION: The hPTTG1 is overexpressed in and closely correlated with the progress of colorectal cancer, therefore hPTTG1 expression may be helpful for the judgement of the prognosis for colorectal cancer patients.
Collapse
|
42
|
Chiu SJ, Chao JI, Lee YJ, Hsu TS. Regulation of gamma-H2AX and securin contribute to apoptosis by oxaliplatin via a p38 mitogen-activated protein kinase-dependent pathway in human colorectal cancer cells. Toxicol Lett 2008; 179:63-70. [PMID: 18499365 DOI: 10.1016/j.toxlet.2008.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 03/15/2008] [Accepted: 04/03/2008] [Indexed: 12/27/2022]
Abstract
Oxaliplatin, a chemotherapeutic drug, induces DNA strand breaks leading to apoptosis in colorectal cancer cells. gamma-H2AX is a phosphorylated histone H2AX that can act as a marker of DNA double-strand breaks (DSBs). It has been shown that securin proteins were over-expressed in a variety of cancer cells. However, the roles of gamma-H2AX and securin on the oxaliplatin-induced apoptosis in human colorectal cancer cells remain unclear. Treatment of oxaliplatin (1-10 microM for 6-24h) persistently induced gamma-H2AX formation and inhibited securin protein expression via a time- and concentration-dependent manner in HCT116 securin-wild type colorectal cancer cells. Compared with HCT116 securin-wild type cells, the induction of apoptosis and persistent gamma-H2AX formation by oxaliplatin was reduced in the HCT116 securin-null colorectal cancer cells. Furthermore, the blockage of caspases by specific caspase inhibitors reduced the levels of gamma-H2AX proteins and cytotoxicity but increased securin protein expression in the oxaliplatin-exposed cells. The gene knockdown of H2AX by transfection with a short interfering RNA of H2AX enhanced the oxaliplatin-induced cell death. Interestingly, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was markedly increased by oxaliplatin. Pre-treatment of a specific p38 MAPK inhibitor SB202190 reduced gamma-H2AX proteins and increased securin protein expression in the oxaliplatin-treated cells. Our findings suggest that p38 MAPK may oppositely regulate securin protein expression and gamma-H2AX formation in the oxaliplatin-induced apoptosis of human colorectal cancer cells.
Collapse
Affiliation(s)
- Shu-Jun Chiu
- Department of Life Science, Tzu Chi University, Hualien 970, Taiwan.
| | | | | | | |
Collapse
|
43
|
Morris SM, Akerman GS, Desai VG, Tsai CA, Tolleson WH, Melchior WB, Lin CJ, Fuscoe JC, Casciano DA, Chen JJ. Effect of p53 genotype on gene expression profiles in murine liver. Mutat Res 2008; 640:54-73. [PMID: 18206960 DOI: 10.1016/j.mrfmmm.2007.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 11/30/2007] [Accepted: 12/11/2007] [Indexed: 05/25/2023]
Abstract
The tumor suppressor protein p53 is a key regulatory element in the cell and is regarded as the "guardian of the genome". Much of the present knowledge of p53 function has come from studies of transgenic mice in which the p53 gene has undergone a targeted deletion. In order to provide additional insight into the impact on the cellular regulatory networks associated with the loss of this gene, microarray technology was utilized to assess gene expression in tissues from both the p53(-/-) and p53(+/-) mice. Six male mice from each genotype (p53(+/+), p53(+/-), and p53(-/-)) were humanely killed and the tissues processed for microarray analysis. The initial studies have been performed in the liver for which the Dunnett test revealed 1406 genes to be differentially expressed between p53(+/+) and p53(+/-) or between p53(+/+) and p53(-/-) at the level of p < or = 0.05. Both genes with increased expression and decreased expression were identified in p53(+/-) and in p53(-/-) mice. Most notable in the gene list derived from the p53(+/-) mice was the significant reduction in p53 mRNA. In the p53(-/-) mice, not only was there reduced expression of the p53 genes on the array, but genes associated with DNA repair, apoptosis, and cell proliferation were differentially expressed, as expected. However, altered expression was noted for many genes in the Cdc42-GTPase pathways that influence cell proliferation. This may indicate that alternate pathways are brought into play in the unperturbed liver when loss or reduction in p53 levels occurs.
Collapse
Affiliation(s)
- Suzanne M Morris
- Division of Genetic and Reproductive Toxicology, National Center for Toxicological Research, US Food and Drug Administration, 3900 NCTR Road, Jefferson, AR 72079, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Chiu SJ, Hsu TS, Chao JI. Expression of securin promotes colorectal cancer cell death via a p53-independent pathway after radiation. Chem Biol Interact 2007; 170:153-61. [PMID: 17854787 DOI: 10.1016/j.cbi.2007.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Revised: 07/22/2007] [Accepted: 07/25/2007] [Indexed: 11/16/2022]
Abstract
Securin has been shown to regulate genomic stability; nevertheless, the role of securin on the cytotoxicity after radiation is still unclear. Exposure to 1-10 Gy X-ray radiation induced cell death in RKO colorectal cancer cells. The protein levels of securin, p53, and p21 were elevated by radiation. The proteins of phosphorylation of p53 at serine-15, which located on the nuclei of cancer cells, were highly induced by radiation. However, radiation increased securin proteins, which located on both of nuclei and cytoplasma in RKO cells. The p53-wild type colorectal cancer cells were more susceptible on cytotoxicity than the p53-mutant cells following exposure to radiation. Besides, the existence of securin in colorectal cancer cells induced higher apoptosis than the securin-null after radiation. Securin proteins were elevated by radiation in the p53-wild type and -mutant cells; furthermore, radiation raised the p53 protein expression in both the securin-wild type and -null cells. As a whole, these findings suggest that the existence of securin promotes apoptosis via a p53-indpendent pathway after radiation in human colorectal cancer cells.
Collapse
Affiliation(s)
- Shu-Jun Chiu
- Department of Life Science, College of Life Sciences, Tzu Chi University, Hualien 970, Taiwan
| | | | | |
Collapse
|
45
|
Kim DS, Fong J, Read ML, McCabe CJ. The emerging role of pituitary tumour transforming gene (PTTG) in endocrine tumourigenesis. Mol Cell Endocrinol 2007; 278:1-6. [PMID: 17928133 DOI: 10.1016/j.mce.2007.08.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 08/17/2007] [Indexed: 11/20/2022]
Abstract
It is now 10 years since PTTG was first cloned and isolated. Perhaps the major story of the intervening decade of work performed by numerous groups around the world is the sheer multifunctionality ascribed to this gene. PTTG has been implicated in mechanisms of gene transactivation, cell transformation, angiogenesis, metabolism, apoptosis, DNA repair, genetic instability and mitotic control, both in endocrine and non-endocrine settings. In the current review, we cast a critical eye over a decade of PTTG research within the field of endocrine neoplasia.
Collapse
Affiliation(s)
- D S Kim
- Institute of Biomedical Research, Division of Medical Sciences, University of Birmingham, Birmingham B15 2TH, UK
| | | | | | | |
Collapse
|
46
|
Boikos SA, Stratakis CA. Molecular genetics of the cAMP-dependent protein kinase pathway and of sporadic pituitary tumorigenesis. Hum Mol Genet 2007; 16 Spec No 1:R80-7. [PMID: 17613552 DOI: 10.1093/hmg/ddm019] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pituitary tumors are among the most common human neoplasms. Although these common lesions rarely become clinically manifest and they are almost never malignant, they are the cause of significant morbidity in affected patients. The genetic causes of common pituitary tumors remain for the most part unknown; progress has been limited to the elucidation of the molecular etiology of four genetic syndromes predisposing to pituitary neoplasias: McCune-Albright syndrome, multiple endocrine neoplasia type 1, Carney complex and, most recently, familial acromegaly and prolactinomas and other tumors caused by mutations in the GNAS, menin, PRKAR1A, AIP, and p27 (CDKN1B) genes, respectively. Intense molecular studies of sporadic pituitary tumors from patients with negative family histories and no other neoplasms have yielded interesting findings with abnormalities in growth factor expression and cell cycle control dysregulation. To add to the difficulties in understanding pituitary tumorigenesis in man, good murine models of these neoplasms simply do not exist: pituitary tumors are common in rodents, but their histologic origin (mostly from the intermediate lobe), age of presentation (late in murine life) and clinical course make them hardly models of their human counterparts. The present report reviews the clinical and molecular genetics of the cAMP-dependent protein kinase pathway in human pituitary tumors; it also reviews briefly other pathways that have been involved in sporadic pituitary neoplasms. At the end, we attempt a unifying hypothesis for pituitary tumorigenesis, taking into account data that are also discussed elsewhere in this issue.
Collapse
Affiliation(s)
- Sosipatros A Boikos
- Section on Endocrinology and Genetics (SEGEN), Developmental Endocrinology Branch (DEB), National Institute of Child Health and Human Development (NICHD), National Institues of Health, Bethesda, MD 20892-1103, USA
| | | |
Collapse
|
47
|
Lu Q, Shen N, Li XM, Chen SL. Genomic view of IFN-α response in pre-autoimmune NZB/W and MRL/lpr mice. Genes Immun 2007; 8:590-603. [PMID: 17728792 DOI: 10.1038/sj.gene.6364421] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interferon (IFN)-alpha is involved in the pathogenesis of systemic lupus erythematosus. Studies in murine lupus models have revealed that type I IFN exerts either a protective effect in MRL/lpr, or can detrimentally impact disease progression, as in NZB/W mice. To understand this paradox, we examined the kinetic global gene expression in pre-autoimmune NZB/W-, MRL/lpr- and normal BALB/c-derived splenic mononuclear cells following ex vivo IFN-alpha treatment. Analysis of IFN-alpha-induced gene expression patterns revealed genes associated with antiproliferative activity of IFN-alpha including CDKN1A, GADD45B, pituitary tumor-transforming 1, SCOTIN, ataxia telangiectasia-mutated homolog and calcyclin-binding protein were upregulated in MRL/lpr and/or BALB/c mice. Of IFN-alpha-induced genes differentially expressed in NZB/W vs BALB/c and MRL/lpr mice at 3 h time point, enhanced expression of CCND1, cyclin D2, matrix metalloproteinase 13 and a panel of cytokines and chemokines and impaired expression of negative inflammatory regulators CD69 and an Src family kinase hemopoietic cell kinase were notable. Interestingly, the splenic mononuclear cells from the NZB/W not MRL/lpr lupus-prone mice at the pre-autoimmune stage before ex vivo IFN-alpha treatment, have increased expression of many known IFN-regulated genes. These results provide a unique genomic view of ex vivo IFN-alpha response in two lupus-prone models, and help to have an insight into the role of IFN-alpha in lupus pathogenesis.
Collapse
Affiliation(s)
- Q Lu
- Joint Molecular Rheumatology Laboratory of Institute of Health Sciences and Shanghai RenJi Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai JiaoTong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
48
|
Ghanem N, Hölker M, Rings F, Jennen D, Tholen E, Sirard MA, Torner H, Kanitz W, Schellander K, Tesfaye D. Alterations in transcript abundance of bovine oocytes recovered at growth and dominance phases of the first follicular wave. BMC DEVELOPMENTAL BIOLOGY 2007; 7:90. [PMID: 17662127 PMCID: PMC1976425 DOI: 10.1186/1471-213x-7-90] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2007] [Accepted: 07/27/2007] [Indexed: 11/10/2022]
Abstract
Background Oocyte developmental competence is highly affected by the phase of ovarian follicular wave. Previous studies have shown that oocytes from subordinate follicles recovered at growth phase (day 3 after estrus) are developmentally more competent than those recovered at dominance phase (day 7 after estrus). However, the molecular mechanisms associated with these differences are not well elucidated. Therefore, the objective of this study was to investigate transcript abundance of bovine oocytes retrieved from small follicles at growth and dominance phases of the first follicular wave and to identify candidate genes related to oocyte developmental competence using cDNA microarray. Results Comparative gene expression analysis of oocytes from growth and dominance phases and subsequent data analysis using Significant Analysis of Microarray (SAM) revealed a total of 51 differentially regulated genes, including 36 with known function, 6 with unknown function and 9 novel transcripts. Real-time PCR has validated 10 transcripts revealed by microarray analysis and quantified 5 genes in cumulus cells derived from oocytes of both phases. The expression profile of 8 (80%) transcripts (ANAXA2, FL396, S100A10, RPL24, PP, PTTG1, MSX1 and BMP15) was in agreement with microarray data. Transcript abundance of five candidate genes in relation to oocyte developmental competence was validated using Brilliant Cresyl Blue (BCB) staining as an independent model. Furthermore, localization of mRNA and protein product of the candidate gene MSX1 in sections of ovarian follicles at days 0, 1, 3 and 7 of estrous cycle showed a clear fluorescent signal in both oocytes and cumulus cells with higher intensity in the former. Moreover, the protein product was detected in bovine oocytes and early cleavage embryos after fertilization with higher intensity around the nucleus. Conclusion This study has identified distinct sets of differentially regulated transcripts between bovine oocytes recovered from small follicles at growth and dominance phases of the first follicular wave. The validation with independent model supports our notion that many of the transcripts identified here may represent candidate genes associated with oocyte developmental competence. Further specific functional analysis will provide insights into the exact role of these transcripts in oocyte competence and early embryonic development.
Collapse
Affiliation(s)
- Nasser Ghanem
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Michael Hölker
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Franca Rings
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Danyel Jennen
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Ernst Tholen
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Marc-André Sirard
- Centre de Recherche en Biologie de la Reproduction, Université Laval, Département des Sciences Animales, Pav. Comtois, Laval, Sainte-Foy, Québec, G1K 7P4, Canada
| | - Helmut Torner
- Research Institute for Biology of Farm Animals, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Wilhelm Kanitz
- Research Institute for Biology of Farm Animals, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Karl Schellander
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| | - Dawit Tesfaye
- Institute of Animal Science, Animal Breeding and Husbandry Group, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
49
|
Di Pietro C, Piro S, Tabbì G, Ragusa M, Di Pietro V, Zimmitti V, Cuda F, Anello M, Consoli U, Salinaro ET, Caruso M, Vancheri C, Crimi N, Sabini MG, Cirrone GAP, Raffaele L, Privitera G, Pulvirenti A, Giugno R, Ferro A, Cuttone G, Lo Nigro S, Purrello R, Purrello F, Purrello M. Cellular and molecular effects of protons: apoptosis induction and potential implications for cancer therapy. Apoptosis 2007; 11:57-66. [PMID: 16374542 DOI: 10.1007/s10495-005-3346-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Due to their ballistic precision, apoptosis induction by protons could be a strategy to specifically eliminate neoplastic cells. To characterize the cellular and molecular effects of these hadrons, we performed dose-response and time-course experiments by exposing different cell lines (PC3, Ca301D, MCF7) to increasing doses of protons and examining them with FACS, RT-PCR, and electron spin resonance (ESR). Irradiation with a dose of 10 Gy of a 26,7 Mev proton beam altered cell structures such as membranes, caused DNA double strand breaks, and significantly increased intracellular levels of hydroxyl ions, are active oxygen species (ROS). This modified the transcriptome of irradiated cells, activated the mitochondrial (intrinsic) pathway of apoptosis, and resulted in cycle arrest at the G2/M boundary. The number of necrotic cells within the irradiated cell population did not significantly increase with respect to the controls. The effects of irradiation with 20 Gy were qualitatively as well as quantitatively similar, but exposure to 40 Gy caused massive necrosis. Similar experiments with photons demonstrated that they induce apoptosis in a significantly lower number of cells and in a temporally delayed manner. These data advance our knowledge on the cellular and molecular effects of proton irradiation and could be useful for improving current hadrontherapy protocols.
Collapse
Affiliation(s)
- C Di Pietro
- Dipartimento di Scienze Biomediche, Sezione di Biologia, Genetica e Bioinformatica, Università di Catania, Catania, 95123, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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
|