1
|
Huang Z, Liu D, Zhang Y, Lu W, Hu L, Zhang J, Xie L, Chen S. PITX1 as a grading, prognostic and tumor-infiltrating immune cells marker for chondrosarcoma: a public database-based immunoassay and tissue sample analysis. Front Oncol 2025; 15:1477649. [PMID: 40342824 PMCID: PMC12060168 DOI: 10.3389/fonc.2025.1477649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/24/2025] [Indexed: 05/11/2025] Open
Abstract
Background Chondrosarcoma (CHS) is a rare bone cancer originating from chondrocytes, with high-grade cases associated with high mortality rates. However, the prognostic factors and therapeutic targets for CHS have not been studied. Methods Graded gene differential analysis was conducted on 97 CHS tissues to identify genes associated with CHS grading. Additionally, we performed GO and KEGG enrichment analyses of the differentially-expressed genes (DEGs), as well as GSEA analysis, differential expression analysis, survival analysis, and univariable and multifactorial COX analysis of paired-like homology structural domain transcription factor 1 (PITX1). Furthermore, our findings investigated the relationship between tumor-infiltrating immune cells (TICs) in CHS tumors using CIBERSORT to calculate proportions and differences. Our findings also explored the associations among gene expression patterns, survival prognosis, TICs, and immune checkpoints across various cancer types. Finally, immunohistochemical staining was carried out on self-collected clinical samples to assess PITX1 expression levels and correlate them with clinical information. Results Gene differential expression analysis revealed a strong correlation between PITX1 expression and tumor grade. GO, KEGG enrichment, and GSEA analysis demonstrated the association of PITX1 with cell proliferation-related processes, such as cell cycle regulation and mitosis, and differentiation-related processes, such as RNA processing. PITX1 expression was associated with tumor stage and survival outcomes. Immunoassay indicated a positive correlation between PITX1 levels and TICs, immune checkpoints, and graded TICs. Pan-cancer analysis confirmed the differential expression of the PITX1 gene across multiple cancers, impacting survival prognosis, TIC patterns, and immune checkpoint regulation. Lastly, our 75 collection of clinical patient tissue samples exhibited varying levels of PITX1 expression across different cancer grades while also demonstrating a significant association with tumor differentiation and metastasis. Conclusion PITX1 is a novel biomarker for distinguishing between high-grade and low-grade CHS, serving as a prognostic indicator for patients with this condition and presenting a promising target for immunotherapy. These findings offer innovative insights into the treatment of CHS.
Collapse
Affiliation(s)
- Zikun Huang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongchen Liu
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ying Zhang
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Weiqing Lu
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lan Hu
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jinghao Zhang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Lei Xie
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Sport Medicine Centre, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shubiao Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Sport Medicine Centre, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
2
|
Zhao J, Xu Y. PITX1 plays essential functions in cancer. Front Oncol 2023; 13:1253238. [PMID: 37841446 PMCID: PMC10570508 DOI: 10.3389/fonc.2023.1253238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
PITX1, also known as the pituitary homeobox 1 gene, has emerged as a key regulator in animal growth and development, attracting significant research attention. Recent investigations have revealed the implication of dysregulated PITX1 expression in tumorigenesis, highlighting its involvement in cancer development. Notably, PITX1 interacts with p53 and exerts control over crucial cellular processes including cell cycle progression, apoptosis, and chemotherapy resistance. Its influence extends to various tumors, such as esophageal, colorectal, gastric, and liver cancer, contributing to tumor progression and metastasis. Despite its significance, a comprehensive review examining PITX1's role in oncology remains lacking. This review aims to address this gap by providing a comprehensive overview of PITX1 in different cancer types, with a particular focus on its clinicopathological significance.
Collapse
Affiliation(s)
- Jingpu Zhao
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
PITX1 inhibits the growth and proliferation of melanoma cells through regulation of SOX family genes. Sci Rep 2021; 11:18405. [PMID: 34526609 PMCID: PMC8443576 DOI: 10.1038/s41598-021-97791-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/30/2021] [Indexed: 01/04/2023] Open
Abstract
Melanoma is one of the most aggressive types of cancer wherein resistance to treatment prevails. Therefore, it is important to discover novel molecular targets of melanoma progression as potential treatments. Here we show that paired-like homeodomain transcription factor 1 (PITX1) plays a crucial role in the inhibition of melanoma progression through regulation of SRY-box transcription factors (SOX) gene family mRNA transcription. Overexpression of PITX1 in melanoma cell lines resulted in a reduction in cell proliferation and an increase in apoptosis. Additionally, analysis of protein levels revealed an antagonistic cross-regulation between SOX9 and SOX10. Interestingly, PITX1 binds to the SOX9 promoter region as a positive regulatory transcription factor; PITX1 mRNA expression levels were positively correlated with SOX9 expression, and negatively correlated with SOX10 expression in melanoma tissues. Furthermore, transcription of the long noncoding RNA (lncRNA), survival-associated mitochondrial melanoma-specific oncogenic noncoding RNA (SAMMSON), was decreased in PITX1-overexpressing cells. Taken together, the findings in this study indicate that PITX1 may act as a negative regulatory factor in the development and progression of melanoma via direct targeting of the SOX signaling.
Collapse
|
4
|
Ballester V, Taylor WR, Slettedahl SW, Mahoney DW, Yab TC, Sinicrope FA, Boland CR, Lidgard GP, Cruz-Correa MR, Smyrk TC, Boardman LA, Ahlquist DA, Kisiel JB. Novel methylated DNA markers accurately discriminate Lynch syndrome associated colorectal neoplasia. Epigenomics 2020; 12:2173-2187. [PMID: 33350853 PMCID: PMC7923255 DOI: 10.2217/epi-2020-0132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Acquired molecular changes in Lynch syndrome (LS) colorectal tumors have been largely unstudied. We identified methylated DNA markers (MDMs) for discrimination of colorectal neoplasia in LS and determined if these MDMs were comparably discriminant in sporadic patients. Patients & methods: For LS discovery, we evaluated DNA from 53 colorectal case and control tissues using next generation sequencing. For validation, blinded methylation-specific PCR assays to the selected MDMs were performed on 197 cases and controls. Results: OPLAH was the most discriminant MDM with areas under the receiver operating characteristic curve ≥0.97 for colorectal neoplasia in LS and sporadic tissues. ALKBH5, was uniquely hypermethylated in LS neoplasms. Conclusion: Highly discriminant MDMs for colorectal neoplasia in LS were identified with potential use in screening and surveillance.
Collapse
Affiliation(s)
- Veroushka Ballester
- Division of Digestive & Liver Diseases, Columbia University, New York, NY 10032, USA
| | - William R Taylor
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Tracy C Yab
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Frank A Sinicrope
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Marcia R Cruz-Correa
- Comprehensive Cancer Center, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA
| | - Thomas C Smyrk
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Lisa A Boardman
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - David A Ahlquist
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - John B Kisiel
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Paired like homeodomain 1 and SAM and SH3 domain-containing 1 in the progression and prognosis of head and neck squamous cell carcinoma. Int J Biochem Cell Biol 2020; 127:105846. [PMID: 32905855 DOI: 10.1016/j.biocel.2020.105846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/14/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy with high morbidity and mortality rates. In spite of numerous advancements have been made in therapeutic methods, the prognosis of HNSCC patients remains poor. Therefore, investigation of crucial genes during HNSCC tumorigenesis which could be exploited as biomarkers and therapeutic targets is greatly needed. In this study, original data of four independent datasets was downloaded from the Gene Expression Omnibus database and analyzed through R language to screen out differentially expressed genes. Paired like homeodomain 1 and SAM and SH3 domain-containing 1 were selected to be further explored through multiple online databases. Quantitative real-time polymerase chain reaction analysis and immunohistochemistry assay were adopted to validate the downregulation of paired like homeodomain 1 and SAM and SH3 domain-containing 1 in HNSCC and statistical analysis indicated their close associations with patient prognosis. In vitro experiments demonstrated the inhibitory effect of paired like homeodomain 1 and SAM and SH3 domain-containing 1 on HNSCC progression. Overall, we identified the aberrant downregulation of paired like homeodomain 1 and SAM and SH3 domain-containing 1 in HNSCC and suggested the potential of utilizing them as therapeutic targets or efficient biomarkers for diagnosis and prognosis evaluation. Our findings may provide novel evidences for the development of new strategies for HNSCC treatment.
Collapse
|
6
|
Ashrafizadeh M, Rafiei H, Mohammadinejad R, Farkhondeh T, Samarghandian S. Wnt-regulating microRNAs role in gastric cancer malignancy. Life Sci 2020; 250:117547. [PMID: 32173311 DOI: 10.1016/j.lfs.2020.117547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is responsible for high morbidity and mortality worldwide. This cancer claims fifth place among other cancers. There are a number of factors associated with GC development such as alcohol consumption and tobacco smoking. It seems that genetic factors play significant role in GC malignancy and progression. MicroRNAs (miRs) are short non-coding RNA molecules with negative impact on the expression of target genes. A variety of studies have elucidated the potential role of miRs in GC growth. Investigation of molecular pathways has revealed that miRs function as upstream modulators of Wnt signaling pathway. This signaling pathway involves in important biological processes such as cell proliferation and differentiation, and its dysregulation is associated with GC invasion. At the present review, we demonstrate that how miRs regulate Wnt signaling pathway in GC malignancy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Hossein Rafiei
- Department of Biology, Faculty of Sciences, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
7
|
Liu L, Tian YC, Mao G, Zhang YG, Han L. MiR-675 is frequently overexpressed in gastric cancer and enhances cell proliferation and invasion via targeting a potent anti-tumor gene PITX1. Cell Signal 2019; 62:109352. [DOI: 10.1016/j.cellsig.2019.109352] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/21/2019] [Accepted: 06/27/2019] [Indexed: 12/29/2022]
|
8
|
Ohira T, Kojima H, Kuroda Y, Aoki S, Inaoka D, Osaki M, Wanibuchi H, Okada F, Oshimura M, Kugoh H. PITX1 protein interacts with ZCCHC10 to regulate hTERT mRNA transcription. PLoS One 2019; 14:e0217605. [PMID: 31404068 PMCID: PMC6690549 DOI: 10.1371/journal.pone.0217605] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/29/2019] [Indexed: 01/21/2023] Open
Abstract
Telomerase is a ribonucleoprotein ribonucleic enzyme that is essential for cellular immortalization via elongation of telomere repeat sequences at the end of chromosomes. Human telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase holoenzyme, is a key regulator of telomerase activity. Telomerase activity, which has been detected in the majority of cancer cells, is accompanied by hTERT expression, suggesting that this enzyme activity contributes to an unlimited replication potential of cancer cells via regulation of telomere length. Thus, hTERT is an attractive target for cancer-specific treatments. We previously reported that pared-like homeodomain 1 (PITX1) is a negative regulator of hTERT through direct binding to the hTERT promoter. However, the mechanism by which the function of PITX1 contributes to transcriptional silencing of the hTERT gene remains to be clarified. Here, we show that PITX1 and zinc finger CCHC-type containing 10 (ZCCHC10) proteins cooperate to facilitate the transcriptional regulation of the hTERT gene by functional studies via FLAG pull-down assay. Co-expression of PITX1 and ZCCHC10 resulted in inhibition of hTERT transcription, in melanoma cell lines, whereas mutate-deletion of homeodomain in PITX1 that interact with ZCCHC10 did not induce similar phenotypes. In addition, ZCCHC10 expression levels showed marked decrease in the majority of melanoma cell lines and tissues. Taken together, these results suggest that ZCCHC10-PITX1 complex is the functional unit that suppresses hTERT transcription, and may play a crucial role as a novel tumor suppressor complex.
Collapse
Affiliation(s)
- Takahito Ohira
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
| | - Hirotada Kojima
- Department of Immunology, Graduate School of Medicine, Osaka City University, Asahi-machi, Abeno-ku, Osaka, Japan
| | - Yuko Kuroda
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
| | - Sayaka Aoki
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
| | - Daigo Inaoka
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
| | - Mitsuhiko Osaki
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
- Division of Pathological Biochemistry, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Graduate School of Medicine, Osaka City University, Asahi-machi, Abeno-ku, Osaka, Japan
| | - Futoshi Okada
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
- Division of Pathological Biochemistry, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Mitsuo Oshimura
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
| | - Hiroyuki Kugoh
- Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, Yonago, Tottori, Japan
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori, Japan
- * E-mail:
| |
Collapse
|
9
|
Shen X, Gu Y, Yu S, Gong P, Mao Y, Li Y, Zheng Y, Qiao F, Zhao Z, Fan H. Silenced PITX1 promotes chemotherapeutic resistance to 5-fluorocytosine and cisplatin in gastric cancer cells. Exp Ther Med 2019; 17:4046-4054. [PMID: 31007741 PMCID: PMC6468935 DOI: 10.3892/etm.2019.7459] [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] [Received: 07/10/2018] [Accepted: 01/31/2019] [Indexed: 12/11/2022] Open
Abstract
Resistance to chemotherapeutic drugs leads to a poor prognosis in gastric cancer (GC). The present study aimed to assess the association between pituitary homeobox paired homeodomain transcription 1 (PITX1) expression and the sensitivity of GC cells to the chemotherapeutic drugs 5-fluorouracil (5-FU) and cisplatin (CDDP). In the present study, the gastric cancer cell lines GES-1, AGS, BGC-823, MCG-803 and SGC-7901 were used. The expression of PITX1 was determined via reverse transcription-quantitative polymerase chain reaction in GC cell lines. AGS and BGC-823 cells, which exhibit a decreased PITX1 expression, were transfected with a PITX1 cDNA construct and its control vector. MCG-803 and SGC-7901 cells, which exhibit an increased PITX1 expression, were transfected with siRNA against PITX1 and its control scramble sequence. A Cell Counting kit-8 assay was performed to determine the impact of PITX1 expression on the sensitivity of GC cells to 5-FU and CDDP. The Cancer Genome Atlas database was used to analyze the expression of PITX1 with GC prognosis in the Asian population and to assess the potential mechanism of PITX1 in 5-FU and CDDP resistance. The results revealed that the overexpression of PIXT1 increased the sensitivity of GC cells to 5-FU/CDDP. The combination of 5-FU/CDDP and PITX1 overexpression also reduced the proliferation of GC cells. Additionally, PIXT1 knockdown decreased the sensitivity of GC cells to 5-FU/CDDP. TCGA data revealed that a lower expression of PITX1 is exhibited in Asian GC patients than in normal individuals. GC patients with a lower expression of PITX1 had a poor prognosis. The expression of PITX1 affected the sensitivity of GC cells to 5-FU/CDDP, indicating that PITX1 may increase the efficacy of treatment in GC patients.
Collapse
Affiliation(s)
- Xiaohui Shen
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yuejun Gu
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Shengling Yu
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Pihai Gong
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yuhang Mao
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yiping Li
- Department of Pathology, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ying Zheng
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Fengchang Qiao
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhujiang Zhao
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Hong Fan
- Department of Medical Genetics and Developmental Biology, Medical School of Southeast University, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
10
|
Gunathilake MN, Lee J, Cho YA, Oh JH, Chang HJ, Sohn DK, Shin A, Kim J. Interaction between physical activity, PITX1 rs647161 genetic polymorphism and colorectal cancer risk in a Korean population: a case-control study. Oncotarget 2018; 9:7590-7603. [PMID: 29484135 PMCID: PMC5800927 DOI: 10.18632/oncotarget.24136] [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: 06/21/2017] [Accepted: 12/26/2017] [Indexed: 12/31/2022] Open
Abstract
This study assessed the interaction between physical activity and colorectal cancer (CRC) risk based on a polymorphism in the paired-like homeodomain 1 (PITX1) gene in Koreans. In total, 923 cases and 1,846 controls were enrolled at the National Cancer Center, Korea. Subjects who did regular exercise showed a significantly reduced risk of CRC than those did not exercise regularly (OR = 0.37, 95% CI = 0.30-0.45). Subjects in the highest tertile of metabolic equivalents of task (MET)-minutes per week showed a significantly lower risk of CRC (OR = 0.62, 95% CI = 0.48-0.79, p-trend < 0.001). In the dominant model, minor allele carriers showed a significantly higher risk of CRC than subjects homozygous for the major allele (OR = 1.46, 95% CI = 1.18-1.80). The PITX1 genetic variant showed significant interactions with regular exercise and CRC risk (p-interaction = 0.018) and colon cancer risk (p-interaction = 0.029) among all subjects. Subjects who carried at least one minor allele and did not regularly exercise showed a greater risk of CRC (OR = 1.81, 95% CI = 1.37-2.41). Subjects who were homozygous for the major allele with high physical activity showed a significantly reduced risk of CRC (OR = 0.56, 95% CI = 0.38-0.82). Thus, individuals with PITX1 genetic variants can have benefit from physical activity regarding prevention of CRC risk in a Korean population.
Collapse
Affiliation(s)
- Madhawa Neranjan Gunathilake
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Jeonghee Lee
- Department of Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Young Ae Cho
- Department of Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Jae Hwan Oh
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Hee Jin Chang
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Dae Kyung Sohn
- Center for Colorectal Cancer, National Cancer Center Hospital, National Cancer Center, Goyang-si, Gyeonggi-do 10408, South Korea
| | - Aesun Shin
- Department of Preventive Medicine, Seoul National University College of Medicine, Jongno-gu, Seoul 03080, South Korea
| | - Jeongseon Kim
- Department of Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, Gyeonggi-do 10408, South Korea
| |
Collapse
|
11
|
DNA hypermethyation and silencing of PITX1 correlated with advanced stage and poor postoperative prognosis of esophageal squamous cell carcinoma. Oncotarget 2017; 8:84434-84448. [PMID: 29137437 PMCID: PMC5663609 DOI: 10.18632/oncotarget.21375] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/03/2017] [Indexed: 11/25/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is associated with the accumulation of genetic and epigenetic changes in the background mucosa. Dysregulated DNA methylation is known to lead to the inactivation of tumor suppressor genes and the activation of oncogenes. To identify the genes whose expression is perturbed by abnormal DNA methylation in ESCC, integrative transcriptomics by serial analysis of gene expression (SAGE) and methylome sequencing by methyl-DNA immunoprecipitation (MeDIP) analysis were performed. We found 159 genes with significantly decreased expression in ESCC compared to that in noncancerous esophageal mucosa. MeDIP-seq analysis identified hypermethylation in the promoter region of 56 of these genes. Using surgically resected tissues of 40 cases, we confirmed that the paired-like homeodomain 1 (PITX1) gene was hypermethylated in ESCC compared to that in normal tissues (P < 0.0001) by pyrosequencing. PITX1 overexpression in ESCC cell lines inhibited cell growth and colony formation, whereas PITX1 knockdown accelerated cell growth. A PITX1-transfected ESCC cell line, KYSE30, formed smaller tumors in nude mice than in mock-transfected cells. Hypermethylation of PITX1 was associated with tumor depth (P = 0.0011) and advanced tumor stage (P = 0.0052) and predicted poor survival in ESCC (hazard ratio, 0.1538; 95% confidence interval, 0.03159–0.7488; P = 0.0169). In this study, we found a novel tumor suppressor gene of ESCC, PITX1, which is silenced by DNA hypermethylation. Downregulation of PITX1 contributes to the growth and progression of ESCC. Hypermethylation of the PITX1 in ESCC correlated with tumor progression and advanced stage cancer, and may predict a poor prognosis.
Collapse
|
12
|
Barut F, Udul P, Kokturk F, Kandemir NO, Keser SH, Ozdamar SO. Clinicopathological features and pituitary homeobox 1 gene expression in the progression and prognosis of cutaneous malignant melanoma. Kaohsiung J Med Sci 2016; 32:494-500. [PMID: 27742032 DOI: 10.1016/j.kjms.2016.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/14/2016] [Accepted: 06/27/2016] [Indexed: 11/27/2022] Open
Abstract
The evidence that PITX1 (pituitary homeobox 1) is a significant tumor suppressor in human cancer remains largely circumstantial, but it clearly warrants further study as little is known about the tumor-inhibitory roles of PITX1 in cutaneous malignant melanoma. The aims of this study were to investigate PITX1 gene expression in patients with cutaneous malignant melanoma and to evaluate its potential relevance to clinicopathological characteristics and tumor cell proliferation. Clinicopathological findings of patients with cutaneous malignant melanoma were analyzed retrospectively. PITX1 and Ki-67 expression were detected by immunohistochemistry in malignant melanoma and healthy tissue samples from each patient. Labeling indices were calculated based on PITX1 gene and Ki-67 expression. The correlation between PITX1and Ki-67 expressions was analyzed in cutaneous malignant melanoma cases. The relationship between PITX1 expression intensity and clinicopathological characteristics was also analyzed. PITX1 expression was observed in all (100%) normal healthy skin tissue samples. In addition, PITX1 expression was found in 56 (80%) and was absent in 14 (20%) of the 70 cutaneous malignant melanoma cases. Ki-67 positive expression was only detected in the 14 (20%) PITX1-negative cases. PITX1-positive tumor cells were observed on the surface, but Ki-67 positive tumor cells were observed in deeper zones of the tumor nests. PITX1 expression was downregulated in human cutaneous malignant melanoma lesions compared with healthy skin tissue, but Ki-67 expression was upregulated in concordance with the progression of cutaneous malignant melanoma. PITX1 expression may be involved in tumor progression and is a potential tumor suppressor gene and prognostic marker for cutaneous malignant melanoma.
Collapse
Affiliation(s)
- Figen Barut
- Department of Pathology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey.
| | - Perihan Udul
- Department of Pathology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Furuzan Kokturk
- Department of Biostatistics, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Nilufer Onak Kandemir
- Department of Pathology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| | - Sevinc Hallac Keser
- Department of Pathology, Dr. Lutfi Kırdar Training and Research Hospital, Istanbul, Turkey
| | - Sukru Oguz Ozdamar
- Department of Pathology, Faculty of Medicine, Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
13
|
Takenobu M, Osaki M, Fujiwara K, Fukuhara T, Kitano H, Kugoh H, Okada F. PITX1 is a novel predictor of the response to chemotherapy in head and neck squamous cell carcinoma. Mol Clin Oncol 2016; 5:89-94. [PMID: 27330773 DOI: 10.3892/mco.2016.880] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/19/2016] [Indexed: 12/24/2022] Open
Abstract
The pituitary homeobox 1 (PITX1) protein is essential for developmental processes in humans. Previously, PITX1 was identified as a possible tumor suppressor gene in various types of human carcinoma. However, the association between PITX1 and human head and neck squamous cell carcinoma (HNSCC) remains to be elucidated. Immunohistochemical analysis was performed to examine the expression levels of PITX1 in 47 cases of HNSCC, and in 4 control cases. The expression of p53 was also examined in these cases. The labeling indices (LIs) were calculated, and the correlations between clinical factors (chemosensitivity, prognosis and the degree of differentiation) and the LIs were assessed. The PITX1 LI in HNSCC was 27.4±14.5%, which was significantly lower compared with the LIs of the control samples: 76.9±6.97% (P<0.05). Additionally, the PITX1 LIs were 39.9±6.2, 26.9±16.9 and 24.2±11.8% in the complete response (CR), partial response (PR), stable disease or progressive disease (SD/PD) groups, respectively. The PITX1 LI in the CR group revealed the highest result between the all groups, and it was significantly greater compared with that in the SD/PD group (P<0.01). The p53 LIs were 24.5±19.9, 25.7±16.9 and 19.8±13.8 in the CR, PR and SD/PD groups, respectively (P>0.05). Neither the PITX1 nor the p53 LIs were a statistically significant indicator of the prognosis. PITX1 is a candidate tumor suppressor gene and a possible predictive biomarker of chemosensitivity of human HNSCC.
Collapse
Affiliation(s)
- Masao Takenobu
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan; Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Mitsuhiko Osaki
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan; Chromosome Engineering Research Center, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Kazunori Fujiwara
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Takahiro Fukuhara
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Hiroya Kitano
- Department of Otolaryngology, Head and Neck Surgery, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan
| | - Hiroyuki Kugoh
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori 683-8503, Japan; Division of Molecular Genetics and Biofunction, Graduate School of Medical Science; Tottori University, Yonago, Tottori 683-8503, Japan
| | - Futoshi Okada
- Division of Pathological Biochemistry, Department of Biomedical Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8503, Japan; Chromosome Engineering Research Center, Tottori University, Yonago, Tottori 683-8503, Japan
| |
Collapse
|
14
|
Studies of Tumor Suppressor Genes via Chromosome Engineering. Cancers (Basel) 2015; 8:cancers8010004. [PMID: 26729168 PMCID: PMC4728451 DOI: 10.3390/cancers8010004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/19/2015] [Accepted: 12/21/2015] [Indexed: 12/01/2022] Open
Abstract
The development and progression of malignant tumors likely result from consecutive accumulation of genetic alterations, including dysfunctional tumor suppressor genes. However, the signaling mechanisms that underlie the development of tumors have not yet been completely elucidated. Discovery of novel tumor-related genes plays a crucial role in our understanding of the development and progression of malignant tumors. Chromosome engineering technology based on microcell-mediated chromosome transfer (MMCT) is an effective approach for identification of tumor suppressor genes. The studies have revealed at least five tumor suppression effects. The discovery of novel tumor suppressor genes provide greater understanding of the complex signaling pathways that underlie the development and progression of malignant tumors. These advances are being exploited to develop targeted drugs and new biological therapies for cancer.
Collapse
|
15
|
Ke J, Lou J, Chen X, Li J, Liu C, Gong Y, Yang Y, Zhu Y, Zhang Y, Gong J. Identification of a Potential Regulatory Variant for Colorectal Cancer Risk Mapping to Chromosome 5q31.1: A Post-GWAS Study. PLoS One 2015; 10:e0138478. [PMID: 26381143 PMCID: PMC4575091 DOI: 10.1371/journal.pone.0138478] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/29/2015] [Indexed: 02/07/2023] Open
Abstract
Large-scale genome-wide association studies (GWAS) have established chromosome 5q31.1 as a susceptibility locus for colorectal cancer (CRC), which was still lack of causal genetic variants. We searched potentially regulatory single nucleotide polymorphisms (SNPs) in the overlap region between linkage disequilibrium (LD) block of 5q31.1 and regulatory elements predicted by histone modifications, then tested their association with CRC via a case-control study. Among three candidate common variants, we found rs17716310 conferred significantly (heterozygous model: OR = 1.273, 95% confidence interval (95%CI) = 1.016–1.595, P = 0.036) and marginally (dominant model: OR = 1.238, 95%CI = 1.000–1.532, P = 0.050) increase risk for CRC in a Chinese population including 695 cases and 709 controls. This variation was suggested to be regulatory altering the activity of enhancer that control PITX1 expression. Using epigenetic information such as chromatin immunoprecipitation-sequencing (ChIP-seq) data might help researchers to interpret the results of GWAS and locate causal variants for diseases in post-GWAS era.
Collapse
Affiliation(s)
- Juntao Ke
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Lou
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xueqin Chen
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaoyuan Li
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Liu
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajie Gong
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Yang
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Zhang
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Gong
- State Key Laboratory of Environment Health (Incubation), MOE (Ministry of Education) Key Laboratory of Environment & Health, Ministry of Environmental Protection Key Laboratory of Environment and Health (Wuhan), and Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| |
Collapse
|
16
|
Kong G, Liu Z, Wu K, Zhang Y, Deng Z, Feng W, Chen S, Wang H. Strong expression of paired-like homeodomain transcription factor 1 (PITX1) is associated with a favorable outcome in human osteosarcoma. Tumour Biol 2015; 36:7735-41. [PMID: 25936343 DOI: 10.1007/s13277-015-3512-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
Paired-like homeodomain transcription factor 1 (PITX1) has been implicated as a tumor suppressor in various cancers. However, the biological and clinical significance of PITX1 in osteosarcoma has not been fully elucidated. Here, we studied the expression and clinical significance of PITX1 in 6 normal lower limb bone tissue specimens and 35 osteosarcoma tissue samples by immunohistochemistry. PITX1 was expressed in all normal tissues (6/6, 100 %) and in 85.7 % (30/35) of tumor tissues (P > 0.05). In addition, all normal tissue specimens showed high PITX1 expression (6/6, 100 %) while only 23.3 % (7/30) osteosarcoma tissue specimens had high PITX1 expression (P < 0.05). Patients with median overall survival (OS) >12 months had significantly higher PITX1 levels compared with those whose median OS was less than or equal to 12 months (P < 0.05 or 0.001). Furthermore, patients with lung metastasis had significantly lower PITX1 levels than patients without lung metastasis. In conclusion, PITX1 expression is downregulated in osteosarcoma and correlates with patient survival and lung metastasis.
Collapse
Affiliation(s)
- Gengbin Kong
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Kezhou Wu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Ying Zhang
- Department of Pathology, Shantou University Medical College, No.22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhihua Deng
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Weili Feng
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Shubiao Chen
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Hu Wang
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
17
|
miR-19b regulates hTERT mRNA expression through targeting PITX1 mRNA in melanoma cells. Sci Rep 2015; 5:8201. [PMID: 25643913 PMCID: PMC4314654 DOI: 10.1038/srep08201] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) plays a crucial role in cancer development. We previously identified paired-like homeodomain1 (PITX1) as an hTERT suppressor gene. However, the underlying mechanisms that are involved in the regulation of PITX1 remain unknown. Here, we report that the microRNA-19b (miR-19b) regulates hTERT expression and cell proliferation through inhibition of PITX1. Compared with normal melanocyte cells, miR-19b expression was higher in most melanoma cells and was accompanied by downregulation of PITX1. Moreover, overexpression of miR-19b inhibited PITX1 mRNA translation through a miR-19b binding site within the 3'UTR of the PITX1 mRNA. Our combined findings indicate the participation of miR-19b as a novel upstream effector of hTERT transcription via direct targeting of PITX1.
Collapse
|
18
|
Yu Z, Chen D, Su Z, Li Y, Yu W, Zhang Q, Yang L, Li C, Yang S, Ni L, Gui Y, Mao Z, Lai Y. miR‑886‑3p upregulation in clear cell renal cell carcinoma regulates cell migration, proliferation and apoptosis by targeting PITX1. Int J Mol Med 2014; 34:1409-16. [PMID: 25190136 DOI: 10.3892/ijmm.2014.1923] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/28/2014] [Indexed: 11/06/2022] Open
Abstract
miR‑886‑3p has been discovered to be involved in the oncogenesis, progression and metastasis of several types of human cancer. The aim of the present study was to identify the biological function of miR‑886‑3p in clear cell renal cell carcinoma (ccRCC) and to determine its possible molecular mechanisms. miR‑886‑3p was found to be significantly upregulated in ccRCC tissues (P<0.05), in accordance with a previous sequencing result. Functional experiments revealed that forced downregulation of miR‑886‑3p significantly inhibited cellular migration, suppressed cell proliferation and induced cell apoptosis of renal cancer cells. Paired‑like homeodomain 1 (PITX1), which has been identified as a tumor suppressor, was found to be downregulated in ccRCC tissues and identified as a target gene of miR‑886‑3p. Further experiments demonstrated that the protein level, and not the mRNA level, of PITX1 was significantly decreased or increased when miR‑886‑3p was upregulated or downregulated, respectively, indicating that miR‑886‑3p acted as an oncogene by directly regulating the protein expression of PITX1 at a post‑transcriptional level. In conclusion, this study revealed that miR‑886‑3p was upregulated in ccRCC and was involved in cellular migration, proliferation and apoptosis of renal cancer cells by directly targeting the tumor suppressor gene, PITX1.
Collapse
Affiliation(s)
- Zuhu Yu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Duqun Chen
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Zhengming Su
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Yifan Li
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Wenshui Yu
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Qiang Zhang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Lihua Yang
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Cailing Li
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Shangqi Yang
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Liangchao Ni
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Yaoting Gui
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| | - Zebin Mao
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100083, P.R. China
| | - Yongqing Lai
- Department of Urology, Peking University Shenzhen Hospital, Institute of Urology, Shenzhen PKU‑HKUST Medical Center, Shenzhen 518036, P.R. China
| |
Collapse
|
19
|
Single nucleotide polymorphisms associated with colorectal cancer susceptibility and loss of heterozygosity in a Taiwanese population. PLoS One 2014; 9:e100060. [PMID: 24968322 PMCID: PMC4072675 DOI: 10.1371/journal.pone.0100060] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 05/22/2014] [Indexed: 01/01/2023] Open
Abstract
Given the significant racial and ethnic diversity in genetic variation, we are intrigued to find out whether the single nucleotide polymorphisms (SNPs) identified in genome-wide association studies of colorectal cancer (CRC) susceptibility in East Asian populations are also relevant to the population of Taiwan. Moreover, loss of heterozygosity (LOH) may provide insight into how variants alter CRC risk and how regulatory elements control gene expression. To investigate the racial and ethnic diversity of CRC-susceptibility genetic variants and their relevance to the Taiwanese population, we genotyped 705 CRC cases and 1,802 healthy controls (Taiwan Biobank) for fifteen previously reported East Asian CRC-susceptibility SNPs and four novel genetic variants identified by whole-exome sequencing. We found that rs10795668 in FLJ3802842 and rs4631962 in CCND2 were significantly associated with CRC risk in the Taiwanese population. The previously unreported rs1338565 was associated with a significant increased risk of CRC. In addition, we also genotyped tumor tissue and paired adjacent normal tissues of these 705 CRC cases to search for LOH, as well as risk-associated and protective alleles. LOH analysis revealed preferential retention of three SNPs, rs12657484, rs3802842, and rs4444235, in tumor tissues. rs4444235 has been recently reported to be a cis-acting regulator of BMP4 gene; in this study, the C allele was preferentially retained in tumor tissues (p = 0.0023). rs4631962 and rs10795668 contribute to CRC risk in the Taiwanese and East Asian populations, and the newly identified rs1338565 was specifically associated with CRC, supporting the ethnic diversity of CRC-susceptibility SNPs. LOH analysis suggested that the three CRC risk variants, rs12657484, rs3802842, and rs4444235, exhibited somatic allele-specific imbalance and might be critical during neoplastic progression.
Collapse
|
20
|
PITX1 is a reliable biomarker for predicting prognosis in patients with oral epithelial dysplasia. Oncol Lett 2013; 7:750-754. [PMID: 24527083 PMCID: PMC3919858 DOI: 10.3892/ol.2013.1775] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 12/11/2013] [Indexed: 01/08/2023] Open
Abstract
Paired-like homeodomain 1 (PITX1) genes are essential in human development. In the present study, PITX1 protein expression was evaluated in human normal oral mucosa, oral epithelial dysplasia and oral squamous cell carcinoma (OSCC), with the aim of examining the expression patterns of these critical genes during the multi-stage transformation of oral epithelial dysplasia to OSCC. PITX1 and Ki-67 expression were assessed by immunohistochemistry in 26 individuals with normal oral mucosa, 106 patients with oral epithelial dysplasia and 97 OSCC patients. The labeling indices (LIs) of PITX1 and Ki-67 were calculated and their correlation with the incidence of malignancy was evaluated. The PITX1 LI of the dysplasia specimens was significantly lower than that of the normal oral mucosa samples, but significantly higher than that of the OSCC samples. The oral epithelial dysplasia patients that exhibited low PITX1 expression showed a significantly higher incidence of malignant transformation than those exhibiting high PITX1 expression, regardless of the histological grades of their oral epithelial dysplasias. On the other hand, no correlation was observed between the Ki-67 LI and the incidence of malignancy. These results suggested that PITX1 suppression is associated with malignant transformation in the oral epithelium and that PITX1 expression may serve as a novel biomarker for predicting prognosis in oral epithelial dysplasia.
Collapse
|
21
|
Booy EP, Howard R, Marushchak O, Ariyo EO, Meier M, Novakowski SK, Deo SR, Dzananovic E, Stetefeld J, McKenna SA. The RNA helicase RHAU (DHX36) suppresses expression of the transcription factor PITX1. Nucleic Acids Res 2013; 42:3346-61. [PMID: 24369427 PMCID: PMC3950718 DOI: 10.1093/nar/gkt1340] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
RNA Helicase associated with AU-rich element (RHAU) (DHX36) is a DEAH (Aspartic acid, Glumatic Acid, Alanine, Histidine)-box RNA helicase that can bind and unwind G4-quadruplexes in DNA and RNA. To detect novel RNA targets of RHAU, we performed an RNA co-immunoprecipitation screen and identified the PITX1 messenger RNA (mRNA) as specifically and highly enriched. PITX1 is a homeobox transcription factor with roles in both development and cancer. Primary sequence analysis identified three probable quadruplexes within the 3′-untranslated region of the PITX1 mRNA. Each of these sequences, when isolated, forms stable quadruplex structures that interact with RHAU. We provide evidence that these quadruplexes exist in the endogenous mRNA; however, we discovered that RHAU is tethered to the mRNA via an alternative non–quadruplex-forming region. RHAU knockdown by small interfering RNA results in significant increases in PITX1 protein levels with only marginal changes in mRNA, suggesting a role for RHAU in translational regulation. Involvement of components of the microRNA machinery is supported by similar and non-additive increases in PITX1 protein expression on Dicer and combined RHAU/Dicer knockdown. We also demonstrate a requirement of argonaute-2, a key RNA-induced silencing complex component, to mediate RHAU-dependent changes in PITX1 protein levels. These results demonstrate a novel role for RHAU in microRNA-mediated translational regulation at a quadruplex-containing 3′-untranslated region.
Collapse
Affiliation(s)
- Evan P Booy
- Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada, University of Manitoba, Winnipeg, Manitoba, Canada, Department of Biochemistry and Molecular Biology, University of British Columbia, V6T 1Z4 Vancouver, British Columbia, Canada and Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada, R3T 2N2
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Jia WH, Zhang B, Matsuo K, Shin A, Xiang YB, Jee SH, Kim DH, Ren Z, Cai Q, Long J, Shi J, Wen W, Yang G, Delahanty RJ, Ji BT, Pan ZZ, Matsuda F, Gao YT, Oh JH, Ahn YO, Park EJ, Li HL, Park JW, Jo J, Jeong JY, Hosono S, Casey G, Peters U, Shu XO, Zeng YX, Zheng W. Genome-wide association analyses in East Asians identify new susceptibility loci for colorectal cancer. Nat Genet 2012; 45:191-6. [PMID: 23263487 PMCID: PMC3679924 DOI: 10.1038/ng.2505] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 11/29/2012] [Indexed: 12/12/2022]
Abstract
To identify novel genetic factors for colorectal cancer (CRC), we conducted a genome-wide association study in East Asians. By analyzing genome-wide data in 2,098 cases and 5,749 controls, we selected 64 promising SNPs for replication in an independent set of samples including up to 5,358 cases and 5,922 controls. We identified four SNPs with a P-value of 8.58 × 10−7 to 3.77 × 10−10 in the combined analysis of all East Asian samples. Three of the four SNPs were replicated in a study conducted among 26,060 European descendants with a combined P-value of 1.22 × 10−10 for rs647161 (5q31.1), 6.64 × 10−9 for rs2423279 (20p12.3), and 3.06 × 10−8 for rs10774214 (12p13.32 near the CCND2 gene), respectively, derived from the meta-analysis of data from both East Asian and European populations. This study identified three new CRC susceptibility loci and provides additional insight into the genetics and biology of CRC.
Collapse
Affiliation(s)
- Wei-Hua Jia
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Knösel T, Chen Y, Hotovy S, Settmacher U, Altendorf-Hofmann A, Petersen I. Loss of desmocollin 1-3 and homeobox genes PITX1 and CDX2 are associated with tumor progression and survival in colorectal carcinoma. Int J Colorectal Dis 2012; 27:1391-9. [PMID: 22438068 DOI: 10.1007/s00384-012-1460-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/09/2012] [Indexed: 02/04/2023]
Abstract
BACKGROUND Genomewide expression profiling has identified a number of genes differentially expressed in colorectal carcinomas (CRCs) compared to normal tissue. Some of these genes were linked to epithelial-mesenchymal transition. We tested whether genes including desmocollins and homeobox genes were distinct on the protein level and correlated the expression with clinicopathological data. METHODS Tissue microarrays of 402 R0-resected colorectal carcinomas of UICC stage II or III were constructed to evaluate ten biomarkers. Furthermore, mRNA expression of desmocollins was evaluated in eight colon cancer cell lines. Demethylation test was performed by treatment with 5-aza-2´-deoxycytide in five colon cancer cell lines. RESULTS On protein level, high expression of desmocollin 1 (DSC1) was observed in 41.6%, DSC2 in 58.0%, DSC3 in 61.4%, E-cadherin in 71.4%, CDX2 in 58.0%, PITX1 in 55.0%, CDK4 in 0.2%, TLE1 in 1.3%, Factor H in 42.5%, and MDM2 in 0.2%. Reduced expression of DSC1-3 was statistically linked to higher grading and DSC2, E-cadherin and CDX2 with shorter survival in high-grade carcinomas. Multivariate analysis showed that pathological stage and low PITX1 expression were statistically associated with shorter patients survival. On mRNA level, seven out of eight cell lines exhibited no expression of DSC1, and four out of seven restored DSC1 expression after demethylation test. CONCLUSIONS Reduced expression of PITX1 was independently correlated to shorter patients survival and could serve as a prognostic marker. Decreased expression of DSC1-3 is significantly correlated with higher tumor grading. Downregulation of DSC1 could be explained by DNA hypermethylation in colon cancer cells.
Collapse
Affiliation(s)
- Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Thalkirchnerstr. 36, 80337 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Regulation of the human catalytic subunit of telomerase (hTERT). Gene 2012; 498:135-46. [PMID: 22381618 DOI: 10.1016/j.gene.2012.01.095] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been much interest in the regulation of telomerase, the enzyme responsible for maintaining the integrity of chromosomal ends, and its crucial role in cellular immortalization, tumorigenesis, and the progression of cancer. Telomerase activity is characterized by the expression of the telomerase reverse transcriptase (TERT) gene, suggesting that TERT serves as the major limiting agent for telomerase activity. Recent discoveries have led to characterization of various interactants that aid in the regulation of human TERT (hTERT), including numerous transcription factors; further supporting the pivotal role that transcription plays in both the expression and repression of telomerase. Several studies have suggested that epigenetic modulation of the hTERT core promoter region may provide an additional level of regulation. Although these studies have provided essential information on the regulation of hTERT, there has been ambiguity of the role of methylation within the core promoter region and the subsequent binding of various activating and repressive agents. As a result, we found it necessary to consolidate and summarize these recent developments and elucidate these discrepancies. In this review, we focus on the co-regulation of hTERT via transcriptional regulation, the presence or absence of various activators and repressors, as well as the epigenetic pathways of DNA methylation and histone modifications.
Collapse
|
25
|
Chen H, Yang XW, Zhang H, Yang Q, Wang Z, Liu Y, Lu FL, Zhou BY, Qiu-Xi CH, Lu SL. In vivo and in vitro expression of the RASAL1 gene in human gastric adenocarcinoma and its clinicopathological significance. Oncol Lett 2011; 3:535-540. [PMID: 22740946 DOI: 10.3892/ol.2011.513] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 11/28/2011] [Indexed: 12/30/2022] Open
Abstract
Recent studies have suggested that the RAS protein activator like-1 (RASAL1) is a potential tumor suppressor, which is found to be reduced in certain human cancers. Its downregulation is involved in the progression of malignancies. However, whether or not RASAL1 plays a role in the development of gastric cancer remains to be determined. Our study aimed to clarify the role of RASAL1 in the progression of gastric adenocarcinoma. The expression of RASAL1 in primary gastric adenocarcinoma tissue specimens was determined by immunohistochemistry. The expression of RASAL1 mRNA and protein was detected by RT-PCR and western blotting in gastric adenocarcinoma cell lines with varying differentiation statuses, including well-differentiated MKN-28, moderately differentiated SGC-7901 and poorly differentiated BGC-823, respectively. A normal gastric epithelial cell line, GES-l, was used as the control line. The immunohistochemical results revealed that the expression of the RASAL1 protein was mainly observed in the cytoplasm. Among 50 cases of gastric adenocarcinoma tissues, 12 cases were identified as (-), 23 cases (+), 13 cases (++) and 2 cases (+++). Among 50 cases of normal gastric tissues, 16 cases were (++) and 34 cases (+++). The expression of the RASAL1 protein was found to be decreased in the gastric adenocarcinoma tissue compared with normal gastric tissue (p<0.01). Moreover, in the gastric carcinoma tissues, the expression of RASAL1 was correlated with carcinoma diameter, differentiation grades, invasive depth, lymph node metastasis and TNM. Additionally, the RASAL1 mRNA and proteins were decreased in the three gastric adenocarcinoma cell lines compared with the normal gastric epithelial cell line GES-l. In addition, the downregulation of RASAL1 correlated with the differentiation status of cancer cell lines. Based on the above investigation, we conclude that expression of the RASAL1 gene is decreased in gastric carcinoma tissues and cell lines. The results indicate that RASAL1 may be important in the tumorigenesis and development of gastric carcinoma.
Collapse
Affiliation(s)
- Hong Chen
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Libório TN, Acquafreda T, Matizonkas-Antonio LF, Silva-Valenzuela MG, Ferraz AR, Nunes FD. In situ hybridization detection of homeobox genes reveals distinct expression patterns in oral squamous cell carcinomas. Histopathology 2011; 58:225-33. [DOI: 10.1111/j.1365-2559.2011.03751.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
27
|
Identification of PITX1 as a TERT suppressor gene located on human chromosome 5. Mol Cell Biol 2011; 31:1624-36. [PMID: 21300782 DOI: 10.1128/mcb.00470-10] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Telomerase, a ribonucleoprotein enzyme that maintains telomere length, is crucial for cellular immortalization and cancer progression. Telomerase activity is attributed primarily to the expression of telomerase reverse transcriptase (TERT). Using microcell-mediated chromosome transfer (MMCT) into the mouse melanoma cell line B16F10, we previously found that human chromosome 5 carries a gene, or genes, that can negatively regulate TERT expression (H. Kugoh, K. Shigenami, K. Funaki, J. Barrett, and M. Oshimura, Genes Chromosome Cancer 36:37-47, 2003). To identify the gene responsible for the regulation of TERT transcription, we performed cDNA microarray analysis using parental B16F10 cells, telomerase-negative B16F10 microcell hybrids with a human chromosome 5 (B16F10MH5), and its revertant clones (MH5R) with reactivated telomerase. Here, we report the identification of PITX1, whose expression leads to the downregulation of mouse tert (mtert) transcription, as a TERT suppressor gene. Additionally, both human TERT (hTERT) and mouse TERT (mtert) promoter activity can be suppressed by PITX1. We show that three and one binding site within the hTERT and mtert promoters, respectively, that express a unique conserved region are responsible for the transcriptional activation of TERT. Furthermore, we showed that PITX1 binds to the TERT promoter both in vitro and in vivo. Thus, PITX1 suppresses TERT transcription through direct binding to the TERT promoter, which ultimately regulates telomerase activity.
Collapse
|
28
|
Bibliography. Current world literature. Diabetes and the endocrine pancreas II. Curr Opin Endocrinol Diabetes Obes 2008; 15:383-93. [PMID: 18594281 DOI: 10.1097/med.0b013e32830c6b8e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|