1
|
Ma J, Li C, Qian H, Zhang Y. MTA1: A Vital Modulator in Prostate Cancer. Curr Protein Pept Sci 2022; 23:456-464. [PMID: 35792131 DOI: 10.2174/1389203723666220705152713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) is the most frequent cancer of the male genitourinary system and the second most common cancer in men worldwide. PCa has become one of the leading diseases endangering men's health in Asia in recent years, with a large increase in morbidity and mortality. MTA1 (metastasis-associated antigen-1), a transcriptional coregulator involved in histone deacetylation and nucleosome remodeling, is a member of the MTA family. MTA1 is involved in cell signaling, chromosomal remodeling, and transcriptional activities, all of which are important for epithelial cell progression, invasion, and growth. MTA1 has been demonstrated to play a significant role in the formation, progression, and metastasis of PCa, and MTA1 expression is specifically linked to PCa bone metastases. Therefore, MTA1 may be a potential target for PCa prevention and treatment. Here, we reviewed the structure, function, and expression of MTA1 in PCa as well as drugs that target MTA1 to highlight a potential new treatment for PCa.
Collapse
Affiliation(s)
- Jialu Ma
- Graduate School of Hebei Medical University, Shijiazhuang, China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China
- Department of Urology Surgery, National Cancer Center/ National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Du L, Wang L, Gan J, Yao Z, Lin W, Li J, Guo Y, Chen Y, Zhou F, Jim Yeung SC, Coppes RP, Zhang D, Zhang H. MTA3 Represses Cancer Stemness by Targeting the SOX2OT/SOX2 Axis. iScience 2019; 22:353-368. [PMID: 31810000 PMCID: PMC6909183 DOI: 10.1016/j.isci.2019.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/05/2019] [Accepted: 11/05/2019] [Indexed: 02/05/2023] Open
Abstract
Cancer cell stemness (CCS) plays critical roles in both malignancy maintenance and metastasis, yet the underlying molecular mechanisms are far from complete. Although the importance of SOX2 in cancer development and CCS are well recognized, the role of MTA3 in these processes is unknown. In this study, we used esophageal squamous cell carcinoma (ESCC) as a model system to demonstrate that MTA3 can repress both CCS and metastasis in vitro and in vivo. Mechanistically, by forming a repressive complex with GATA3, MTA3 downregulates SOX2OT, subsequently suppresses the SOX2OT/SOX2 axis, and ultimately represses CCS and metastasis. More importantly, MTA3low/SOX2high is associated with poor prognosis and could serve as an independent prognostic factor. These findings altogether indicate that MTA3/SOX2OT/SOX2 axis plays an indispensable role in CCS. Therefore, this axis could be potentially used in cancer stratification and serves as a therapeutic target.
Collapse
Affiliation(s)
- Liang Du
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; Institute of Precision Cancer Medicine and Pathology, Department of Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China; Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen 9700 AD, the Netherlands
| | - Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; Institute of Precision Cancer Medicine and Pathology, Department of Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China
| | - Jinfeng Gan
- Institute of Precision Cancer Medicine and Pathology, Department of Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China; Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhimeng Yao
- Institute of Precision Cancer Medicine and Pathology, Department of Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China; Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wan Lin
- Cancer Research Center, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Junkuo Li
- The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang, Henan 455001, China; Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, Henan 455001, China
| | - Yi Guo
- Endoscopy Center, Affiliated Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yuping Chen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Fuyou Zhou
- The Fourth Affiliated Hospital of Henan University of Science and Technology, Anyang, Henan 455001, China; Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang, Henan 455001, China.
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert P Coppes
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen 9700 AD, the Netherlands
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, 4170 City Avenue, Philadelphia, PA 19131, USA; Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Hao Zhang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China; Institute of Precision Cancer Medicine and Pathology, Department of Pathology, Jinan University Medical College, Guangzhou, Guangdong 510632, China; Research Centre of Translational Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515063, China.
| |
Collapse
|
3
|
Bartlett AH, Liang JW, Sandoval-Sierra JV, Fowke JH, Simonsick EM, Johnson KC, Mozhui K. Longitudinal study of leukocyte DNA methylation and biomarkers for cancer risk in older adults. Biomark Res 2019; 7:10. [PMID: 31149338 PMCID: PMC6537435 DOI: 10.1186/s40364-019-0161-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022] Open
Abstract
Background Changes in DNA methylation over the course of life may provide an indicator of risk for cancer. We explored longitudinal changes in CpG methylation from blood leukocytes, and likelihood of future cancer diagnosis. Methods Peripheral blood samples were obtained at baseline and at follow-up visit from 20 participants in the Health, Aging and Body Composition prospective cohort study. Genome-wide CpG methylation was assayed using the Illumina Infinium Human MethylationEPIC (HM850K) microarray. Results Global patterns in DNA methylation from CpG-based analyses showed extensive changes in cell composition over time in participants who developed cancer. By visit year 6, the proportion of CD8+ T-cells decreased (p-value = 0.02), while granulocytes cell levels increased (p-value = 0.04) among participants diagnosed with cancer compared to those who remained cancer-free (cancer-free vs. cancer-present: 0.03 ± 0.02 vs. 0.003 ± 0.005 for CD8+ T-cells; 0.52 ± 0.14 vs. 0.66 ± 0.09 for granulocytes). Epigenome-wide analysis identified three CpGs with suggestive p-values ≤10− 5 for differential methylation between cancer-free and cancer-present groups, including a CpG located in MTA3, a gene linked with metastasis. At a lenient statistical threshold (p-value ≤3 × 10− 5), the top 10 cancer-associated CpGs included a site near RPTOR that is involved in the mTOR pathway, and the candidate tumor suppressor genes REC8, KCNQ1, and ZSWIM5. However, only the CpG in RPTOR (cg08129331) was replicated in an independent data set. Analysis of within-individual change from baseline to Year 6 found significant correlations between the rates of change in methylation in RPTOR, REC8 and ZSWIM5, and time to cancer diagnosis. Conclusion The results show that changes in cellular composition explains much of the cross-sectional and longitudinal variation in CpG methylation. Additionally, differential methylation and longitudinal dynamics at specific CpGs could provide powerful indicators of cancer development and/or progression. In particular, we highlight CpG methylation in the RPTOR gene as a potential biomarker of cancer that awaits further validation. Electronic supplementary material The online version of this article (10.1186/s40364-019-0161-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra H Bartlett
- 1Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN USA
| | - Jane W Liang
- 1Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN USA
| | | | - Jay H Fowke
- 1Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN USA
| | - Eleanor M Simonsick
- 2Intramural Research Program, National Institute on Aging, Baltimore, MD USA
| | - Karen C Johnson
- 1Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN USA
| | - Khyobeni Mozhui
- 1Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN USA
| |
Collapse
|
4
|
Zhang L, Cao Y, Kou X, Che L, Zhou X, Chen G, Zhao J. Long non-coding RNA HCG11 suppresses the growth of glioma by cooperating with the miR-4425/MTA3 axis. J Gene Med 2019; 21:e3074. [PMID: 30706982 DOI: 10.1002/jgm.3074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 01/18/2019] [Accepted: 01/25/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Glioma is a type of malignant tumor that occurs in the central nervous system of adults. Long non-coding RNAs (lncRNAs) that potentially participate in the initiation and progression of glioma have been widely reported. As a now-found lncRNA, HLA complex group 11 (HCG11) has not yet been studied in glioma. The present study aimed to determine the role of HCG11 in the tumorigenesis of glioma. METHODS A quantitative real-time polymerase chain reaction assay was performed to examine the expression pattern of HCG11 in 84 glioma tissues and cell lines. The overall survival rate of glioma patients with a high or low level of HCG11 or metastasis-associated 1 family member 3 (MTA3) was analyzed by Kaplan-Meier analysis. The effect of HCG11 on glioma cell growth was determined by in vitro and in vivo experiments. MicroRNAs (miRNAs) that potentially interact with HCG11 were searched and determined by bioinformatics analysis and a luciferase reporter assay. Similarly, the target of miRNA-4425 was identified. Finally, rescue assays were conducted to determine the bio-function of the competing endogenous RNA pathway. RESULTS HCG11 was downregulated in 84 pairs of glioma tissues and cell lines. Moreover, a low level of HCG11 indicted the lower overall survival rate of glioma patients. Regarding the mechanism, HCG11 was abundant in the cytoplasm of glioma cells and interacted with miR-4425 to release the expression of MTA3. miR-4425 and MTA3 participated in HCG11-mediated glioma growth. CONCLUSIONS LncRNA HCG11 suppresses the growth of glioma by cooperating with the miR-4425/MTA3 axis.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Clinical Laboratory, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Yanfei Cao
- Department of Clinical Laboratory, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Xiaonan Kou
- Department of Clinical Laboratory, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Lu Che
- Department of Clinical Laboratory, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Xiaona Zhou
- Department of Clinical Laboratory, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Gang Chen
- Department of Neurosurgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang, China
| | - Jiang Zhao
- Department of Neurosurgery, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Pfeifer A, Rusinek D, Żebracka-Gala J, Czarniecka A, Chmielik E, Zembala-Nożyńska E, Wojtaś B, Gielniewski B, Szpak-Ulczok S, Oczko-Wojciechowska M, Krajewska J, Polańska J, Jarząb B. Novel TG-FGFR1 and TRIM33-NTRK1 transcript fusions in papillary thyroid carcinoma. Genes Chromosomes Cancer 2019; 58:558-566. [PMID: 30664823 PMCID: PMC6594006 DOI: 10.1002/gcc.22737] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
Papillary thyroid carcinoma (PTC) is most common among all thyroid cancers. Multiple genomic alterations occur in PTC, and gene rearrangements are one of them. Here we screened 14 tumors for novel fusion transcripts by RNA‐Seq. Two samples harboring RET/PTC1 and RET/PTC3 rearrangements were positive controls whereas the remaining ones were negative regarding the common PTC alterations. We used Sanger sequencing to validate potential fusions. We detected 2 novel potentially oncogenic transcript fusions: TG‐FGFR1 and TRIM33‐NTRK1. We detected 4 novel fusion transcripts of unknown significance accompanying the TRIM33‐NTRK1 fusion: ZSWIM5‐TP53BP2, TAF4B‐WDR1, ABI2‐MTA3, and ARID1B‐PSMA1. Apart from confirming the presence of RET/PTC1 and RET/PTC3 in positive control samples, we also detected known oncogenic fusion transcripts in remaining samples: TFG‐NTRK1, ETV6‐NTRK3, MKRN1‐BRAF, EML4‐ALK, and novel isoform of CCDC6‐RET.
Collapse
Affiliation(s)
- Aleksandra Pfeifer
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Dagmara Rusinek
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Jadwiga Żebracka-Gala
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Agnieszka Czarniecka
- Department of Oncological and Reconstructive Surgery, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Ewa Chmielik
- Tumor Pathology Department, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Ewa Zembala-Nożyńska
- Tumor Pathology Department, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Bartosz Wojtaś
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Bartłomiej Gielniewski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Sylwia Szpak-Ulczok
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Małgorzata Oczko-Wojciechowska
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Jolanta Krajewska
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| | - Joanna Polańska
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Gliwice, Poland
| | - Barbara Jarząb
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Institute - Oncology Center Gliwice Branch, Gliwice, Poland
| |
Collapse
|
6
|
Yao Z, Du L, Xu M, Li K, Guo H, Ye G, Zhang D, Coppes RP, Zhang H. MTA3-SOX2 Module Regulates Cancer Stemness and Contributes to Clinical Outcomes of Tongue Carcinoma. Front Oncol 2019; 9:816. [PMID: 31552166 PMCID: PMC6736560 DOI: 10.3389/fonc.2019.00816] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/09/2019] [Indexed: 02/05/2023] Open
Abstract
Cancer cell plasticity plays critical roles in both tumorigenesis and tumor progression. Metastasis-associated protein 3 (MTA3), a component of the nucleosome remodeling and histone deacetylase (NuRD) complex and multi-effect coregulator, can serve as a tumor suppressor in many cancer types. However, the role of MTA3 in tongue squamous cell cancer (TSCC) remains unclear although it is the most prevalent head and neck cancer and often with poor prognosis. By analyzing both published datasets and clinical specimens, we found that the level of MTA3 was lower in TSCC compared to normal tongue tissues. Data from gene set enrichment analysis (GSEA) also indicated that MTA3 was inversely correlated with cancer stemness. In addition, the levels of MTA3 in both samples from TSCC patients and TSCC cell lines were negatively correlated with SOX2, a key regulator of the plasticity of cancer stem cells (CSCs). We also found that SOX2 played an indispensable role in MTA3-mediated CSC repression. Using the mouse model mimicking human TSCC we demonstrated that the levels of MTA3 and SOX2 decreased and increased, respectively, during the process of tumorigenesis and progression. Finally, we showed that the patients in the MTA3low/SOX2high group had the worst prognosis suggesting that MTA3low/SOX2high can serve as an independent prognostic factor for TSCC patients. Altogether, our data suggest that MTA3 is capable of repressing TSCC CSC properties and tumor growth through downregulating SOX2 and MTA3low/SOX2high might be a potential prognostic factor for TSCC patients.
Collapse
Affiliation(s)
- Zhimeng Yao
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Liang Du
- Cancer Research Center, Shantou University Medical College, Shantou, China
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Min Xu
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Kai Li
- Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Haipeng Guo
- Department of Head and Neck Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Guodong Ye
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Robert P. Coppes
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, China
- Research Centre of Translational Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Hao Zhang
| |
Collapse
|
7
|
Hu C, Zhou Y, Liu C, Kang Y. Risk assessment model constructed by differentially expressed lncRNAs for the prognosis of glioma. Oncol Rep 2018; 40:2467-2476. [PMID: 30106138 PMCID: PMC6151882 DOI: 10.3892/or.2018.6639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 08/01/2018] [Indexed: 02/05/2023] Open
Abstract
A risk assessment model was constructed using differentially expressed long non‑coding (lnc)RNAs for the prognosis of glioma. Transcriptome sequencing of the lncRNAs and mRNAs from glioma samples were obtained from the TCGA database. The samples were divided into bad and good prognosis groups based on survival time, then differently expressed lncRNAs between these two groups were screened using DEseq and edgeR packages. Multivariate Cox regression analysis was performed to establish a risk assessment system according to the weighted regression coefficient of lncRNA expression. Survival analysis and receiver operating characteristic curve were conducted for the risk assessment model. Furthermore, the co‑expression network of the screened lncRNAs was constructed, followed by the functional enrichment analysis for associated genes. A total of 117 lncRNAs were screened using edgeR and DEseq packages. Among all differently expressed lncRNAs, five lncRNAs (RP3‑503A6, LINC00940, RP11‑453M23, AC009411 and CDRT7) were identified to establish the risk assessment model. The risk assessment model demonstrated a good prognostic function with high area under the curve values in the training, validation and entire sets. The risk score was certified as an independent prognostic factor for gliomas. Multiple genes were screened to be co‑expressed with these five lncRNAs. Functional enrichment analysis demonstrated that they were involved in cytoskeleton, adhesion and Janus kinase/signal transducer and activator of transcription signaling pathway‑associated processes. The present study established a risk assessment model integrating five significantly different expressed lncRNAs, which may help to assess the prognosis of patients with glioma with increased accuracy.
Collapse
Affiliation(s)
- Chenggong Hu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yongfang Zhou
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Chang Liu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
8
|
Huang Y, Li Y, He F, Wang S, Li Y, Ji G, Liu X, Zhao Q, Li J. Metastasis-associated protein 3 in colorectal cancer determines tumor recurrence and prognosis. Oncotarget 2018; 8:37164-37171. [PMID: 28418887 PMCID: PMC5514899 DOI: 10.18632/oncotarget.16332] [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: 06/11/2016] [Accepted: 02/14/2017] [Indexed: 12/26/2022] Open
Abstract
Metastasis-associated protein family (MTA) promotes tumor cell invasion and metastasis of human malignancies. However, the novel component of MTA family, MTA3 was found to play conflicting roles in human malignancies. While the expression pattern and potential function of MTA3 in colorectal cancer has not been addressed yet. In the present study, we investigated the protein expression of MTA3 by immunohistochemistry assay, analyzed its association with tumor progression, recurrence and prognosis in239 cases of patients. Results showed that MTA3 expression in colorectal cancer was significantly decreased in colorectal cancer compared with normal specimens. Its expression was found to be correlated with tumor differentiation, metastases and TNM stage. Kaplan–Meier analysis proved that MTA3 was associated with both disease-free survival and overall survival of patients with colorectal cancer that patients with negative MTA3 expression tend to have unfavorable outcome. Moreover, cox's proportional hazards analysis showed that negative MTA3 expression was an independent prognostic marker of poor outcome. These results provided the first evidence that MTA3 level was decreased in colorectal cancer and significantly correlated with tumor cell invasion and metastasis. It also demonstrated that MTA3 might serve as a potential marker of tumor recurrence and prognosis of colorectal cancer.
Collapse
Affiliation(s)
- Yi Huang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University. Xi'an, China
| | - Yunlong Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Fenfei He
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Shiqi Wang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yaohui Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Gang Ji
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xiaonan Liu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qingchuan Zhao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jipeng Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Mu N, Gu J, Liu N, Xue X, Shu Z, Zhang K, Huang T, Chu C, Zhang W, Gong L, Zhao H, Jia B, Gao D, Shang L, Zhang W, Guo Q. PRL-3 is a potential glioblastoma prognostic marker and promotes glioblastoma progression by enhancing MMP7 through the ERK and JNK pathways. Am J Cancer Res 2018; 8:1527-1539. [PMID: 29556339 PMCID: PMC5858165 DOI: 10.7150/thno.22699] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/10/2017] [Indexed: 01/08/2023] Open
Abstract
Purpose: Glioblastoma is the most common and aggressive type of primary brain malignancy and is associated with a poor prognosis. Previously, we found that phosphatase of regenerating liver-3 (PRL-3) was significantly up-regulated in glioblastoma as determined by a microarray analysis. However, the function of PRL-3 in glioblastoma remains unknown. We aimed to investigate the clinical relationship between PRL-3 and glioblastoma, and uncover the mechanisms of PRL-3 in the process of glioblastoma. Methods: PRL-3 expression was evaluated in 61 glioblastoma samples and 4 cell lines by RT-qPCR and immunohistochemistry. Kaplan-Meier analysis was performed to evaluate the prognostic value of PRL-3 for overall survival (OS) and progression-free survival (PFS) for glioblastoma patients. Proliferation was evaluated by Cell Counting Kit-8 (CCK-8) assay and EdU proliferation assay, migration and invasion by wound-closure/Transwell assays, and qRT-PCR/immunoblotting/IHC were used for both in vivo and in vitro investigations. Result: A high PRL-3 expression level was closely correlated with unfavorable OS and PFS for glioblastoma patients, and was also significantly correlated with Ki-67 expression. Down-regulation of PRL-3 inhibited glioma cell proliferation, invasion and migration through ERK/JNK/matrix metalloproteinase 7 (MMP7) in vitro and in vivo. Conclusions: PRL-3 expression enhances the invasion and proliferation of glioma cells, highlighting this phosphatase as a novel prognostic candidate and an attractive target for future therapy in glioblastoma.
Collapse
|
10
|
Oroxyloside inhibits human glioma progression by suppressing proliferation, metastasis and inducing apoptosis related pathways. Biomed Pharmacother 2017; 97:1564-1574. [PMID: 29793319 DOI: 10.1016/j.biopha.2017.09.100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/02/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022] Open
Abstract
Malignant glioma are linked to a high mortality rate. Therefore, it is necessary to explore and develop effective therapeutic strategy. Oroxyloside is a metabolite of oroxylin A. However, its inhibitory effects on cancer are little to be known. In the present study, we investigated the effects of oroxyloside on cell proliferation, migration, and apoptosis in vitro and in vivo in human glioma. The results indicated that oroxyloside significantly suppressed the proliferation of human glioma cells through inducing cell cycle arrest at G0/G1 phase through reducing Cyclin D1 and cyclin-dependent kinase 2 (CDK2) while enhancing p53 and p21 expressions. In addition, the migration of glioma cells was dramatically inhibited by oroxyloside in a dose-dependent manner, which was related to its modulation on extracellular matrix (ECM), as evidenced by up-regulated E-cadherin, and metastasis-associated protein 3 (MTA3), whereas down-regulated N-cadherin, Vimentin, Twist, alpha-smooth muscle actin (α-SMA) and Syndecan-2. Furthermore, oroxyloside treatment markedly induced apoptosis in glioma cells through improving Caspase-9, Caspase-3 and PARP cleavage, accompanied with high release of cytochrome c (Cyto-c) into cytoplasm and subsequently increase of apoptotic protease-activating factor 1 (Apaf-1). In vivo, oroxyloside administration significantly inhibited the glioma cell xenograft tumorigenesis through various signaling pathways, including suppression of Cyclin D1/CDK2 and ECM pathways, as well as potentiation of p53/p21 and Caspases pathways. Together, the findings above illustrated that oroxyloside, for the first time, was used as a promising candidate against human glioma.
Collapse
|
11
|
Voigt A, Nowick K, Almaas E. A composite network of conserved and tissue specific gene interactions reveals possible genetic interactions in glioma. PLoS Comput Biol 2017; 13:e1005739. [PMID: 28957313 PMCID: PMC5634634 DOI: 10.1371/journal.pcbi.1005739] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 10/10/2017] [Accepted: 08/24/2017] [Indexed: 02/08/2023] Open
Abstract
Differential co-expression network analyses have recently become an important step in the investigation of cellular differentiation and dysfunctional gene-regulation in cell and tissue disease-states. The resulting networks have been analyzed to identify and understand pathways associated with disorders, or to infer molecular interactions. However, existing methods for differential co-expression network analysis are unable to distinguish between various forms of differential co-expression. To close this gap, here we define the three different kinds (conserved, specific, and differentiated) of differential co-expression and present a systematic framework, CSD, for differential co-expression network analysis that incorporates these interactions on an equal footing. In addition, our method includes a subsampling strategy to estimate the variance of co-expressions. Our framework is applicable to a wide variety of cases, such as the study of differential co-expression networks between healthy and disease states, before and after treatments, or between species. Applying the CSD approach to a published gene-expression data set of cerebral cortex and basal ganglia samples from healthy individuals, we find that the resulting CSD network is enriched in genes associated with cognitive function, signaling pathways involving compounds with well-known roles in the central nervous system, as well as certain neurological diseases. From the CSD analysis, we identify a set of prominent hubs of differential co-expression, whose neighborhood contains a substantial number of genes associated with glioblastoma. The resulting gene-sets identified by our CSD analysis also contain many genes that so far have not been recognized as having a role in glioblastoma, but are good candidates for further studies. CSD may thus aid in hypothesis-generation for functional disease-associations.
Collapse
Affiliation(s)
- André Voigt
- Network Systems Biology Group, Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Katja Nowick
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
- Bioinformatics, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
- Human Biology, Institute for Biology, Free University Berlin, Berlin, Germany
| | - Eivind Almaas
- Network Systems Biology Group, Department of Biotechnology, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and General Practice, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
12
|
Wang C, Li G, Li J, Li J, Li T, Yu J, Qin C. Overexpression of the metastasis-associated gene MTA3 correlates with tumor progression and poor prognosis in hepatocellular carcinoma. J Gastroenterol Hepatol 2017; 32:1525-1529. [PMID: 27992674 DOI: 10.1111/jgh.13680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/09/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) is one of the most common and aggressive cancers in the world. However, there remains a lack of effective diagnostic and treatment markers. We aimed to explore metastasis-associated protein 3 (MTA3) expression and function in HCC and its relationship with clinicopathological factors. METHODS We investigated the expression pattern and clinicopathological significance of MTA3 in 90 patients with HCC via immunohistochemistry and explored MTA3 function via gene knockdown of MTA3. RESULTS MTA3 was overexpressed in HCC cell nuclei and downregulated in HCC cell cytoplasm. The former finding correlated with metastasis (P = 0.010) and poor prognosis (P = 0.018). In addition, deleting MTA3 inhibited HCC cell growth, invasion, and metastasis in vitro, as shown in the colony formation, migration, and wound-healing assays. CONCLUSIONS These results indicate that MTA3 is an oncogene of HCC, predicts poor prognosis of HCC, and may be a future marker of HCC treatment.
Collapse
Affiliation(s)
- Chuanxi Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Guanzhen Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jiamei Li
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jie Li
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Tao Li
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Jinyu Yu
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Chengyong Qin
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| |
Collapse
|
13
|
Du L, Ning Z, Zhang H, Liu F. Corepressor metastasis-associated protein 3 modulates epithelial-to-mesenchymal transition and metastasis. CHINESE JOURNAL OF CANCER 2017; 36:28. [PMID: 28279208 PMCID: PMC5345190 DOI: 10.1186/s40880-017-0193-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/22/2017] [Indexed: 02/05/2023]
Abstract
Worldwide, metastasis is the leading cause of more than 90% of cancer-related deaths. Currently, no specific therapies effectively impede metastasis. Metastatic processes are controlled by complex regulatory networks and transcriptional hierarchy. Corepressor metastasis-associated protein 3 (MTA3) has been confirmed as a novel component of nucleosome remodeling and histone deacetylation (NuRD). Increasing evidence supports the theory that, in the recruitment of transcription factors, coregulators function as master regulators rather than passive passengers. As a master regulator, MTA3 governs the target selection for NuRD and functions as a transcriptional repressor. MTA3 dysregulation is associated with tumor progression, invasion, and metastasis in various cancers. MTA3 is also a key regulator of E-cadherin expression and epithelial-to-mesenchymal transition. Elucidating the functions of MTA3 might help to find additional therapeutic approaches for targeting components of NuRD.
Collapse
Affiliation(s)
- Liang Du
- Cancer Research Center, Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
| | - Zhifeng Ning
- Basic Medicine College, Hubei University of Science and Technology, Xianning, 437100 Hubei P. R. China
| | - Hao Zhang
- Cancer Research Center, Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
- Department of Biotherapy, Affiliated Cancer Hospital of Shantou University Medical College, Shantou, 515031 Guangdong P. R. China
| | - Fuxing Liu
- Basic Medicine College, Hubei University of Science and Technology, Xianning, 437100 Hubei P. R. China
| |
Collapse
|
14
|
Advances in clinical neurology through the journal "Neurological Sciences" (2015-2016). Neurol Sci 2017; 38:9-18. [PMID: 28093657 DOI: 10.1007/s10072-017-2815-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Soga T, Lim WL, Khoo ASB, Parhar IS. Kisspeptin Activates Ankrd 26 Gene Expression in Migrating Embryonic GnRH Neurons. Front Endocrinol (Lausanne) 2016; 7:15. [PMID: 26973595 PMCID: PMC4771921 DOI: 10.3389/fendo.2016.00015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/28/2016] [Indexed: 12/27/2022] Open
Abstract
Kisspeptin, a newly discovered neuropeptide, regulates gonadotropin-releasing hormone (GnRH). Kisspeptins are a large RF-amide family of peptides. The kisspeptin coded by KiSS-1 gene is a 145-amino acid protein that is cleaved to C-terminal peptide kisspeptin-10. G-protein-coupled receptor 54 (GPR54) has been identified as a kisspeptin receptor, and it is expressed in GnRH neurons and in a variety of cancer cells. In this study, enhanced green fluorescent protein (EGFP) labeled GnRH cells with migratory properties, which express GPR54, served as a model to study the effects of kisspeptin on cell migration. We monitored EGFP-GnRH neuronal migration in brain slide culture of embryonic day 14 transgenic rat by live cell imaging system and studied the effects of kisspeptin-10 (1 nM) treatment for 36 h on GnRH migration. Furthermore, to determine kisspeptin-induced molecular pathways related with apoptosis and cytoskeletal changes during neuronal migration, we studied the expression levels of candidate genes in laser-captured EGFP-GnRH neurons by real-time PCR. We found that there was no change in the expression level of genes related to cell proliferation and apoptosis. The expression of ankyrin repeat domain-containing protein (ankrd) 26 in EGFP-GnRH neurons was upregulated by the exposure to kisspeptin. These studies suggest that ankrd 26 gene plays an unidentified role in regulating neuronal movement mediated by kisspeptin-GPR54 signaling, which could be a potential pathway to suppress cell migration.
Collapse
Affiliation(s)
- Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Wei Ling Lim
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Alan Soo-Beng Khoo
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur, Malaysia
| | - Ishwar S. Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
- *Correspondence: Ishwar S. Parhar,
| |
Collapse
|