1
|
You D, Tong K, Li Y, Zhang T, Wu Y, Wang L, Chen G, Zhang X. PinX1 plays multifaceted roles in human cancers: a review and perspectives. Mol Biol Rep 2024; 51:1163. [PMID: 39550726 PMCID: PMC11570563 DOI: 10.1007/s11033-024-10082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Pin2/TRF1 interacting protein X1 (PinX1), a telomerase inhibitor, is located at human chromosome 8p23. This region is important for telomere length maintenance and chromosome stability, both of which are essential for regulating human ageing and associated diseases. METHODS AND RESULTS We investigated the research progress of PinX1 in human cancers. In cancers, the expression levels of PinX1 mRNA and protein vary according to cancer cell types, and PinX1 plays a critical role in the regulation of cancer development and progression. Additionally, a review of the literature indicates that PinX1 is involved in mitosis and affects the sensitivity of cancer cells to radiation-induced DNA damage. Therefore, PinX1 has therapeutic potential for cancer, and understanding the function of PinX1 in the regulation of cancers is crucial for improving treatment. In this review, we discuss the expression level of PinX1 in a variety of cancers and how it affects the implicated pathways. Additionally, we outline the function of PinX1 in cancer cells and provide a theoretical basis for PinX1-related cancer therapy. CONCLUSIONS PinX1 has promising prospects in future cancer therapeutics. This review may provide theoretical support for researchers in this field.
Collapse
Affiliation(s)
- Dian You
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China
| | - Kaiwen Tong
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China
| | - Yuan Li
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China
| | - Ting Zhang
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China
| | | | - Ling Wang
- Botuvac Biotechnology Co., Ltd, Beijing, China
| | - Guangming Chen
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China
| | - Xiaoying Zhang
- Huzhou Key Laboratory of Precise Prevention and Control of Major Chronic Diseases, School of Medicine, Huzhou University, 759 East 2nd Rd., Huzhou, 313000, China.
| |
Collapse
|
2
|
Liu Y, Gong P, Zhou N, Zhang J, He C, Wang S, Peng H. Insufficient PINX1 expression stimulates telomerase activation by direct inhibition of EBV LMP1-NF-κB axis during nasopharyngeal carcinoma development. Biochem Biophys Res Commun 2019; 514:127-133. [PMID: 31027734 DOI: 10.1016/j.bbrc.2019.04.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Early malignant transformation of nasopharyngeal carcinoma(NPC) is associated with Epstein-Barr virus(EBV) infection and telomerase activation. The EBV latent membrane protein 1(LMP1) regulates expression of various genes by triggering NF-κB signaling pathway. PINX1 is a well-identified tumor suppressor gene by inhibiting telomerase activity and cancer cell growth. However, whether and how EBV inhibit PINX1 expression and activate telomerase in NPC is still incompletely elucidated. METHODS Immunohistochemistry, real-time PCR and Western blotting were utilized to explore the expression of PINX1. Chromatin immunoprecipitation(ChIP) and Dual-luciferase reporter assay were used to elucidate the regulatory mechanism between NF-κB and PINX1. TRAP-SYBR Green assay and Southern blotting were utilized to detect telomerase activity and telomere length. CCK8 and EdU tests were conducted to measure proliferation ability. RESULTS We demonstrated that PINX1 is down-regulated in NPC for the first time. Mechanistically, we found that LMP1 could inhibit the transcriptional activity of PINX1 by promoting the binding of p65 to three specific sites in PINX1 promoter, significantly, two(-1698/-1689, tgcaatttcc; -206/-197, cgggctttac) of which have not been reported. In addition, we also observed that LMP1 overexpression resulted in increased telomerase activity, prolonged telomere length and enhanced proliferation. CONCLUSION We first discovered EBV led to reduced PINX1 expression through LMP1-NF-κB-PINX1 axis, which up-regulated telomerase activity in NPC. And hence, the tumor cells acquired the ability to proliferate more exuberantly. This signaling pathway illustrates the relationship between EBV latent infection and telomerase activation, and further provides new thinking for early diagnosis and treatment in NPC.
Collapse
Affiliation(s)
- Yunyi Liu
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Pinggui Gong
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China
| | - Ni Zhou
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Junjun Zhang
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Cui He
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China
| | - Shuilian Wang
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Hong Peng
- Guangdong Provincial Second People's Hospital, Southern Medical University, Guangzhou, 510317, Guangdong, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
3
|
Prognostic and Clinicopathological Value of PINX1 in Various Human Tumors: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4621015. [PMID: 30079348 PMCID: PMC6069698 DOI: 10.1155/2018/4621015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/30/2018] [Accepted: 06/27/2018] [Indexed: 01/11/2023]
Abstract
PINX1 (Pin2/TRF1 interacting protein X1, an intrinsic telomerase inhibitor and putative tumor suppressor gene) may represent a novel prognostic tumor biomarker. However, the results of previous studies are inconsistent and the prognostic value of PINX1 remains controversial. Therefore, we conducted a meta-analysis to determine whether PINX1 expression is associated with overall survival (OS), disease-specific survival (DSS), disease-free survival (DFS), recurrence-free survival (RFS), and clinicopathological characteristics in patients with malignant tumors. A systematic search was performed in the PubMed, Web of Science, and Embase databases in April 2018. Quality assessment was performed according to the modified Newcastle-Ottawa Scale. Pooled odds ratios (ORs) and hazard ratios (HRs) with 95.0% confidence intervals (CIs) were calculated to determine the relationship between PINX1 expression and OS, DSS, DFS/RFS, and clinicopathological characteristics. Due to the heterogeneity across the included studies, subgroup and sensitivity analyses were performed. Fixed-effects models were used when the heterogeneity was not significant and random-effects models were used when the heterogeneity was significant. Fourteen studies of 16 cohorts including 2,624 patients were enrolled. Low PINX1 expression was associated with poor OS (HR: 1.51, 95.0% CI: 1.03-2.20; P = 0.035) and DFS/RFS (HR: 1.78, 95.0% CI: 1.28-2.47; P = 0.001) but not DSS (HR: 0.80, 95.0% CI: 0.38-1.67; P = 0.548). Low PINX1 expression was also associated with lymphatic invasion (OR: 2.23, 95.0% CI: 1.35-3.70; P = 0.002) and advanced tumor-node-metastasis stage (OR: 2.43, 95.0% CI: 1.29-4.57; P = 0.006). No significant associations were observed between low PINX1 expression and sex, depth of invasion, grade of differentiation, and distant metastasis. Low PINX1 expression was associated with poor OS and DFS/RFS and lymphatic invasion and advanced tumor-node-metastasis stage, suggesting that PINX1 expression may be a useful predictor of prognosis in patients with malignant tumors.
Collapse
|
4
|
Li HL, Song J, Yong HM, Hou PF, Chen YS, Song WB, Bai J, Zheng JN. PinX1: structure, regulation and its functions in cancer. Oncotarget 2018; 7:66267-66275. [PMID: 27556185 PMCID: PMC5323232 DOI: 10.18632/oncotarget.11411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/09/2016] [Indexed: 02/07/2023] Open
Abstract
PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) is a novel cloned gene located at human chromosome 8p23, playing a vital role in maintaining telomeres length and chromosome stability. It has been demonstrated to be involved in tumor genesis and progression in most malignancies. However, some researches showed opposing molecular status of PinX1 gene and its expression patterns in several other types of tumors. The pathogenic mechanism of PinX1 expression in human malignancy is not yet clear. Moreover, emerging evidence suggest that PinX1 (especially its TID domain) might be a potential new target cancer treatment. Therefore, PinX1 may be a new potential diagnostic biomarker and therapeutic target for human cancers, and may play different roles in different human cancers. The functions and the mechanisms of PinX1 in various human cancers remain unclear, suggesting the necessity of further extensive works of its role in tumor genesis and progression.
Collapse
Affiliation(s)
- Hai-Long Li
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Department of Urology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jun Song
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Department of General Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hong-Mei Yong
- Department of Medical Oncology, Huai'an Hospital to Xuzhou Medical College, Huai'an, Jiangsu, China
| | - Ping-Fu Hou
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yan-Su Chen
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Wen-Bo Song
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jin Bai
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jun-Nian Zheng
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu, China
| |
Collapse
|
5
|
Liu D, Miao H, Zhao Y, Kang X, Shang S, Xiang W, Shi R, Hou A, Wang R, Zhao K, Liu Y, Ma Y, Luo H, Miao H, He F. NF-κB potentiates tumor growth by suppressing a novel target LPTS. Cell Commun Signal 2017; 15:39. [PMID: 29017500 PMCID: PMC5634951 DOI: 10.1186/s12964-017-0196-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023] Open
Abstract
Background Chronic inflammation is causally linked to the carcinogenesis and progression of most solid tumors. LPTS is a well-identified tumor suppressor by inhibiting telomerase activity and cancer cell growth. However, whether and how LPTS is regulated by inflammation signaling is still incompletely elucidated. Methods Real-time PCR and western blotting were used to determine the expression of p65 and LPTS. Reporter gene assay, electrophoretic mobility shift assay and chromatin immunoprecipitation were performed to decipher the regulatory mechanism between p65 and LPTS. Cell counting kit-8 assays and xenograt models were used to detect p65-LPTS-regulated cancer cell growth in vitro and in vivo, respectively. Results Here we for the first time demonstrated that NF-κB could inhibit LPTS expression in the mRNA and protein levels in multiple cancer cells (e.g. cervical cancer and colon cancer cells). Mechanistically, NF-κB p65 could bind to two consensus response elements locating at −1143/−1136 and −888/−881 in the promoter region of human LPTS gene according to EMSA and ChIP assays. Mutation of those two binding sites rescued p65-suppressed LPTS promoter activity. Functionally, NF-κB regulated LPTS-dependent cell growth of cervical and colon cancers in vitro and in xenograft models. In translation studies, we verified that increased p65 expression was associated with decreased LPTS level in multiple solid cancers. Conclusions Taken together, we revealed that NF-κB p65 potentiated tumor growth via suppressing a novel target LPTS. Modulation of NF-κB-LPTS axis represented a potential strategy for treatment of those inflammation-associated malignancies.
Collapse
Affiliation(s)
- Dongbo Liu
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Hongping Miao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuanyin Zhao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Xia Kang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Shenglan Shang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Wei Xiang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Rongchen Shi
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Along Hou
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Kun Zhao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yingzhe Liu
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yue Ma
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Huan Luo
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China.
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
6
|
Huang WJ, Li M, Jin XH, Huang XJ, Zhao W, Tian XP. Genetic profile and biological implication of PIN2/TRF1-interacting telomerase inhibitor 1 (PinX1) in human cancers: an analysis using The Cancer Genome Atlas. Oncotarget 2017; 8:67241-67253. [PMID: 28978030 PMCID: PMC5620170 DOI: 10.18632/oncotarget.18589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/03/2017] [Indexed: 12/15/2022] Open
Abstract
Pin2/TRF1-interacting telomere inhibitor 1 (PinX1) was originally identified as a telomerase inhibitor, involved in maintaining telomerase activity, telomere length, and chromosomal stability. However, research has shown that PinX1 can have opposing molecular status in its expression patterns in several other tumor types. We thus investigated the genetic profile and biological implication of PinX1 in several human cancers using the cBioportal database. Our results showed that PinX1 deletion accounted for the most alterations, with the frequency of its deletion regularly occurring in pathological types of carcinosarcoma and adenocarcinoma. We found few instances of PinX1 gene mutations and 3D structural analysis demonstrated that these mutation sites were always located within telomerase inhibitor domains. Furthermore, our analysis of several human cancers from the cBioportal database revealed more frequent PinX1 homozygous depletion and PinX1 heterozygous deficiency, but both more infrequent PinX1 gain and rare instances of PinX1 amplification. The status of PinX1 genetic alterations was correlated with prognosis and may be tumor-type specific. As such, its biological function in tumorigenesis and later prognosis is complicated and may involve co-worked with NEIL2, R3HCC1, POLR3D, GTF2E2, and INTS10. In addition, we observed that PinX1 interacts with TERT, DKC1, PTGES3, and HSP90AA1. PinX1 mRNA expression was decreased in most selected cancer tissues, which could promote tumor growth and enhance tumorigenicity. Collectively, our data reveal PinX1 expression patterns and potential mechanisms in various human cancers. Further work will be needed to comprehensively examine its role in tumor genesis and progression.
Collapse
Affiliation(s)
- Wei-Juan Huang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Pharmacology, Jinan University, Guangzhou, China
| | - Mei Li
- Department of Pathology, Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Han Jin
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiao-Jia Huang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wei Zhao
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Peng Tian
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
7
|
Feng YZ, Zhang QY, Fu MT, Zhang ZF, Wei M, Zhou JY, Shi R. Low expression of PinX1 is associated with malignant behavior in basal-like breast cancer. Oncol Rep 2017; 38:109-119. [PMID: 28586040 PMCID: PMC5492774 DOI: 10.3892/or.2017.5696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/15/2017] [Indexed: 12/12/2022] Open
Abstract
Human Pinx1 protein, associated with shelterin proteins, is widely revealed as a haploinsufficient tumor suppressor. Growing evidence has manifested the deregulation of PinX1 in distinct cancers. Nonetheless, the loss status of PinX1 and its diagnostic, prognostic and clinicopathological significance in Basal-like breast cancer are still unclear. In the present study, the PinX1 expression levels of breast cancer tissues were investigated by qRT-PCR and immunoblotting assays. Then immunohistochemistry (IHC) was performed to detect PinX1 expression on a tissue microarray. The optimal threshold for PinX1 positivity was determined by receiver operating characteristic (ROC) curve analysis. To clarify the probable role of PinX1 in BLBC, the PinX1 knockout and stably over-expressed MDA-MB-231 cell lines were constructed by the CRISPR-Cas9 system and gene transfection. The association of PinX1 expression with cell proliferation, migration and apoptosis of MDA-MB-231 cells were observed by CCK-8 assay, wound healing assay, Transwell assay, flow cytometric analysis and immunoblotting of the cleaved caspase-3 protein level. Our results showed that both PinX1 mRNA and protein expression were downregulated in breast cancer tissues (P<0.05). In IHC analysis, the optimal cut-off parameter for PinX1 positive expression was 62.5% (the AUC was 0.749, P<0.01). PinX1 positivity was 76.9% (10/14) in luminal subtypes, 50% (5/10) in Her2-enriched breast cancer and 27.3% (9/33) in basal-like subtypes. Besides, in 59 invasive ductal breast carcinomas, PinX1 expression was inversely related to histology grade (P<0.05) while it was positively associated with PR status (P<0.05) and ER status (P<0.05). These results indicated that low expression of PinX1 correlated with aggressive clinicopathological significance of breast cancer, especially in the basal-like subtype. Besides, we identified that overexpression of PinX1 inhibited the proliferation rates and migration ability and increased the apoptosis rates of BLBC. Our findings demonstrated that low expression of PinX1 was associated with malignant behaviors in basal-like subtype of breast cancer. PinX1 is likely a feasible biomarker and molecular target of BLBC.
Collapse
Affiliation(s)
- Yu-Zhen Feng
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qing-Yan Zhang
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Mei-Ting Fu
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhen-Fei Zhang
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Min Wei
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jue-Yu Zhou
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Rong Shi
- Institute of Genetic Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
8
|
Noriega-Reyes MY, Rivas-Torres MA, Oñate-Ocaña LF, Vallés AJ, Baranda-Avila N, Langley E. Novel role for PINX1 as a coregulator of nuclear hormone receptors. Mol Cell Endocrinol 2015; 414:9-18. [PMID: 26187699 DOI: 10.1016/j.mce.2015.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 11/20/2022]
Abstract
Estrogen receptor alpha (ERα) has an established role in breast cancer biology. Transcriptional activation by ERα is a multistep process influenced by coactivator and corepressor proteins. This work shows that Pin2 interacting protein 1 (PINX1) interacts with the N-terminal domain of ERα and functions as a corepressor of ERα. Furthermore, it represses both AF-1 and AF-2 transcriptional activities. Chromatin immunoprecipitation assays verified that the interaction between ERα and PINX1 occurs on E2 regulated promoters and enhanced expression of PINX1 deregulates the expression of a number of genes that have a role in cell growth and proliferation in breast cancer. PINX1 overexpression decreases estrogen mediated proliferation of breast cancer cell lines, while its depletion shows the opposite effect. Taken together, these data show a novel molecular mechanism for PINX1 as an attenuator of estrogen receptor activity in breast cancer cell lines, furthering its role as a tumor suppressor gene in breast cancer.
Collapse
Affiliation(s)
- Maria Yamilet Noriega-Reyes
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de Mexico. D.F., Mexico
| | - Miguel Angel Rivas-Torres
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico; Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de Mexico. D.F., Mexico
| | - Luis Fernando Oñate-Ocaña
- Departamento de Investigación Clínica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico
| | - Albert Jordan Vallés
- Institut de Biología Molecular de Barcelona (IBMB-CSIC) Parc Científic de Barcelona, Barcelona, Cataluña, España
| | - Noemi Baranda-Avila
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico
| | - Elizabeth Langley
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, Tlalpan 14080, Mexico D.F., Mexico.
| |
Collapse
|
9
|
Shi R, Zhao Z, Zhou H, Wei M, Ma WL, Zhou JY, Tan WL. Reduced expression of PinX1 correlates to progressive features in patients with prostate cancer. Cancer Cell Int 2014; 14:46. [PMID: 24936151 PMCID: PMC4059453 DOI: 10.1186/1475-2867-14-46] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 06/04/2014] [Indexed: 11/24/2022] Open
Abstract
Background Pin2/TRF1 binding protein X1 (PinX1) has been identified as an endogenous telomerase inhibitor and a major haploinsufficient tumor suppressor gene. Increasing evidence suggests that reduced expression of PinX1 plays a key role in tumorigenesis. However, the PinX1 expression status and its correlation with the clinicopathological features in prostate cancer (PCa) have not been investigated. Methods PinX1 mRNA and protein expression in PCa and adjacent normal prostate tissues were evaluated by real-time quantitative RT-PCR (qRT-PCR) and western blotting. The clinicopathological significance of PinX1 was investigated by immunohistochemistry (IHC) analysis on a PCa tissue microarray (TMA). The cut-off score for positive expression of PinX1 was determined by the receiver operating characteristic (ROC) analysis. The correlation between PinX1 expression and clinicopathological features of PCa was analyzed by Chi-square test. Results Reduced expression of PinX1 mRNA and protein was observed in the majority of PCa, compared with their paired adjacent normal prostate tissues. When PinX1 positive expression percentage was determined to be above 60% (area under ROC curve = 0.833, P = 0.000), positive expression of PinX1 was observed in 100% (8/8) of normal prostate tissues and 32.5% (13/40) of PCa tissues by IHC. Reduced expression of PinX1 in patients was correlated with advanced clinical stage (χ2 = 10.230, p = 0.017), high Gleason score (χ2 = 4.019, p = 0.045), positive regional lymph node metastasis (χ2 = 10.852, p = 0.004) and distant metastasis (χ2 = 7.965, p = 0.005). Conclusions Our findings suggest that reduced expression of PinX1 is correlates to progressive features in patients with PCa and may serve as a potential marker for diagnosis.
Collapse
Affiliation(s)
- Rong Shi
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Zhen Zhao
- Department of Urinary Surgery, Nanfang Hosptial, Southern Medical University, Guangzhou 510515, China
| | - Hui Zhou
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Min Wei
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Wen-Li Ma
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Jue-Yu Zhou
- Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Wan-Long Tan
- Department of Urinary Surgery, Nanfang Hosptial, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
10
|
The role of PinX1 in growth control of breast cancer cells and its potential molecular mechanism by mRNA and lncRNA expression profiles screening. BIOMED RESEARCH INTERNATIONAL 2014; 2014:978984. [PMID: 24672800 PMCID: PMC3929369 DOI: 10.1155/2014/978984] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 11/12/2013] [Accepted: 12/03/2013] [Indexed: 01/07/2023]
Abstract
As a major tumor suppressor gene, the role of PinX1 in breast cancer and its molecular mechanism remain unclear. In this study, overexpression of PinX1 was generated in 3 breast cancer cell lines, and knockdown of PinX1 was performed in a nontumorigenic breast cell line. The regulation of PinX1 on cell proliferation and cell cycle was observed. A microarray-based lncRNA and mRNA expression profile screening was also performed. We found a lower growth rate, G0/G1 phase arrest, and S phase inhibition in the PinX1 overexpressed breast cancer cells, while a higher growth rate, decreased G0/G1 phase, and increased S phase rate in the PinX1 knocked-down nontumorigenic breast cell. A total of 977 mRNAs and 631 lncRNAs were identified as differentially expressed transcripts between PinX1 overexpressed and control MCF-7 cells. Further analysis identified the involvement of these mRNAs in 52 cancer related pathways and various other biological processes. 11 enhancer-like lncRNAs and 25 lincRNAs with their adjacent mRNA pairs were identified as coregulated transcripts. Our results confirmed the role of PinX1 as a major tumor suppressor gene in breast cancer cell lines and provided information for further research on the molecular mechanisms of PinX1 in tumorigenesis.
Collapse
|
11
|
PinX1, a novel target gene of p53, is suppressed by HPV16 E6 in cervical cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:88-96. [PMID: 24412852 DOI: 10.1016/j.bbagrm.2014.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 12/27/2013] [Accepted: 01/02/2014] [Indexed: 01/14/2023]
|
12
|
Zuo J, Wang DH, Zhang YJ, Liu L, Liu FL, Liu W. Expression and mechanism of PinX1 and telomerase activity in the carcinogenesis of esophageal epithelial cells. Oncol Rep 2013; 30:1823-31. [PMID: 23912465 DOI: 10.3892/or.2013.2649] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/19/2013] [Indexed: 11/05/2022] Open
Abstract
Esophageal tissues were collected from an esophageal carcinoma high-risk area of China and were used to detect the telomere length and the expression of human telomerase reverse transcriptase (hTERT) by immuhistochemistry and fluorescence in situ hybridization; esophageal carcinoma tissues, paired-adjacent mucosa and paired normal mucosa were obtained from resected surgical specimens of esophageal squamous cell carcinoma in order to determine telomerase activity and expression of hTERT and Pin2/TRF1 interacting protein X1 (PinX1) by telomeric repeat amplification protocol-silver staining, RT-PCR and flow cytometry (FCM). The cell proliferation and apoptosis of Eca109 cells were analyzed by FCM and MTT assay. We found that the length of telomere DNA decreased and hTERT protein expression increased in the carcinogenesis of esophageal epithelial cells; telomerase activity was significantly upregulated followed by a decrease of PinX1 expression in esophageal carcinoma compared with dysplasia and normal patients, which notably correlated with grade and lymph node metastasis. Overexpression of PinX1 inhibited cell growth, arrested cells at the G0/G1 stage and induced cell apoptosis in Eca109 cells. In addition, PinX1 overexpression significantly inhibited telomerase activity. In conclusion, the length shortening of telomere was an important characteristic in the carcinogenesis of esophageal epithelial cells, followed by increase of telomerase activity and downregulation of PinX1. Overexpression of PinX1 blocked Eca109 cell proliferation and induced cell apoptosis by downregulating telomerase activity.
Collapse
Affiliation(s)
- Jing Zuo
- Department of Oncology, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050012, P.R. China
| | | | | | | | | | | |
Collapse
|
13
|
Lai XF, Shen CX, Wen Z, Qian YH, Yu CS, Wang JQ, Zhong PN, Wang HL. PinX1 regulation of telomerase activity and apoptosis in nasopharyngeal carcinoma cells. J Exp Clin Cancer Res 2012; 31:12. [PMID: 22316341 PMCID: PMC3296635 DOI: 10.1186/1756-9966-31-12] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 02/08/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human interacting protein X1 (PinX1) has been identified as a critical telomerase inhibitor and proposed to be a putative tumor suppressor gene. Loss of PinX1 has been found in a large variety of malignancies, however, its function in inhibiting telomerase activity of tumor cells is not well documented. Here we show that PinX1 is essential for down-regulation telomerase activity of nasopharyngeal carcinoma. METHODS Expression vectors of human PinX1 (pEGFP-C3-PinX1) and its small interfering RNA (PinX1-FAM-siRNA) were constructed and transfected into NPC. Their effects on mRNA of telomerase catalytic subunit (hTERT), telomerase activity, cell proliferation, cell migration, wound healing, cell cycles and apoptosis were examined using semi-quantitative RT-PCR, stretch PCR, MTT assay, Transwell, scratch assay and flow cytometry, respectively. RESULTS Transfection of pEGFP-C3-PinX1 and PinX1-FAM-siRNA increased and reduced PinX1 mRNA by 1.6-fold and 70%, respectively. Over-expression of PinX1 decreased hTERT mRNA by 21%, reduced telomerase activity, inhibited cell growth, migration and wound healing ability, arrested cells in G0/G1 phase, and increased apoptotic index. In contrast, down-regulation of PinX1 did not alter the above characteristics. CONCLUSIONS PinX1 may play important roles in NPC proliferation, migration and apoptosis and has application potential in tumor-targeted gene therapy.
Collapse
Affiliation(s)
- Xiao-Fen Lai
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Cong-Xiang Shen
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zhong Wen
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, 253# GongYe Road, Guangzhou 510282, China
| | - Yu-Hong Qian
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Chao-Sheng Yu
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jun-Qi Wang
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Ping-Neng Zhong
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hai-Li Wang
- Department of Otolaryngology Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
14
|
Lüscher B. MAD1 and its life as a MYC antagonist: an update. Eur J Cell Biol 2011; 91:506-14. [PMID: 21917351 DOI: 10.1016/j.ejcb.2011.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 07/21/2011] [Accepted: 07/25/2011] [Indexed: 12/16/2022] Open
Abstract
The MYC/MAX/MAD network is of central importance for controlling cell physiology. The network is compiled of transcriptional regulators that form different heterodimers, which can either activate or repress the expression of target genes. Thus these proteins function as a molecular switch to control gene expression. MAD1, a member of this network, acts as a transcriptional repressor. It interacts with MAX to form the OFF position of the switch, antagonizing MYC/MAX complexes that define the ON position. MAD1 regulates cell proliferation and apoptosis through a number of target genes. In addition recent evidence indicates that the expression and activity of MAD1 are regulated at multiple levels. Here the recent developments are summarized, in comparison to MYC, of our understanding how the expression of the MAD1 gene and protein are controlled and what the functional consequences and downstream effectors of MAD1 are, which relay its activity as a transcriptional regulator.
Collapse
Affiliation(s)
- Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, 52057 Aachen, Germany.
| |
Collapse
|
15
|
Wan SM, Tie J, Zhang YF, Guo J, Yang LQ, Wang J, Xia SH, Yang SM, Wang RQ, Fang DC. Silencing of the hPOT1 gene by RNA inference promotes apoptosis and inhibits proliferation and aggressive phenotype of gastric cancer cells, likely through up-regulating PinX1 expression. J Clin Pathol 2011; 64:1051-7. [PMID: 21778296 DOI: 10.1136/jclinpath-2011-200211] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The human protection of telomeres 1 (hPOT1) protein, a single-strand telomeric DNA binding protein, plays an important role in telomere protection and telomere length regulation. However, its effect on invasion of gastric cancer remains unclear. AIMS To explore the role of hPOT1 in the proliferation and invasion of gastric cancer cells. METHODS The gastric expression of hPOT1 was examined in normal gastric mucosa (n=25), intestinal metaplasia (n=20), gastric dysplasia (n=20) and gastric cancer (n=150) by immunohistochemistry. The mean optical density (MOD) of the immunostaining was determined by semi-quantitative image analysis. The role of hPOT1 in the cell proliferation, apoptosis and invasion of gastric cancer 7901 cells was determined by means of the RNA interference (RNAi) of hPOT1 mRNA. The effects of hPOT1 RNAi on the expression of hPinX1 and hTERT were detected with western blotting. RESULTS The hPOT1 MOD was progressively increased from the normal mucosa to intestinal metaplasia, dysplasia, and gastric cancer. An increased hPOT1 expression significantly correlated with tumour serosal invasion, node metastasis and advanced stage. Transfection of hPOT1 siRNA into SGC-7901 cells led to a decrease in cell proliferation, colony formation and invasion, and also an increase of apoptosis. An up-regulation of hPinX1 and down-regulation of hTERT were found in gastric cancer cells with hPOT1 siRNA. CONCLUSIONS Increased hPOT1 expression is associated with an advanced tumour stage. hPOT1 RNAi inhibits proliferation and invasion, and induces apoptosis of gastric cancer cells. The effects of hPOT1 RNAi seem to be functionally linked to up-regulation of PinX1 and down-regulation of hTERT.
Collapse
Affiliation(s)
- Shun-Mei Wan
- Department of Gastroenterology, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|