1
|
Zeng Q, Liu T, Qin L, Wang C, Peng G, Liu Z, Tao J. Screening of potential hub genes involved in Kidney Wilms tumor via bioinformatics analysis and experimental validation. BMC Cancer 2024; 24:771. [PMID: 38937666 PMCID: PMC11209955 DOI: 10.1186/s12885-024-12541-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Wilms tumor (WT) is the most common pediatric embryonal tumor. Improving patient outcomes requires advances in understanding and targeting the multiple genes and cellular control pathways, but its pathogenesis is currently not well-researched. We aimed to identify the potential molecular biological mechanism of WT and develop new prognostic markers and molecular targets by comparing gene expression profiles of Wilms tumors and fetal normal kidneys. METHODS Differential gene expression analysis was performed on Wilms tumor transcriptomic data from the GEO and TARGET databases. For biological functional analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were utilized. Out of 24 hub genes identified, nine were found to be prognostic-related through univariate Cox regression analysis. These nine genes underwent LASSO regression analysis to enhance the predictive capability of the model. The key hub genes were validated in the GSE73209 datasets, and cell function experiments were conducted to identify the genes' functions in WiT-49 cells. RESULTS The enrichment analysis revealed that DEGs were significantly involved in the regulation of angiogenesis and regulation of cell differentiation. 24 DEGs were identified through PPI networks and the MCODE algorithm, and 9 of 24 genes were related to WT patients' prognosis. EMCN and CCNA1 were identified as key hub genes, and related to the progression of WT. Functionally, over-expression of EMCN and CCNA1 knockdown inhibited cell viability, proliferation, migration, and invasion of Wilms tumor cells. CONCLUSIONS EMCN and CCNA1 were identified as key prognostic markers in Wilms tumor, suggesting their potential as therapeutic targets. Differential gene expression and enrichment analyses indicate significant roles in angiogenesis and cell differentiation.
Collapse
Affiliation(s)
- Qiang Zeng
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
- Department of Pediatric Surgery, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330100, Jiangxi, China
| | - Tingting Liu
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
- Department of Pediatric Surgery, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330100, Jiangxi, China
| | - Lilu Qin
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
| | - Chen Wang
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
| | - Guangbei Peng
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
| | - Zhong Liu
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China
| | - Junfeng Tao
- Department of Pediatric Surgery, Jiangxi Maternal and Child Health Hospital, Nanchang, 330100, Jiangxi, China.
- Department of Pediatric Surgery, Jiangxi Hospital Affiliated to Children's Hospital of Chongqing Medical University, Nanchang, 330100, Jiangxi, China.
| |
Collapse
|
2
|
Hsieh CC, Yang CY, Peng B, Ho SL, Tsao CH, Lin CK, Lin CS, Lin GJ, Lin HY, Huang HC, Chang SC, Sytwu HK, Chia WT, Chen YW. Allyl Isothiocyanate Suppresses the Proliferation in Oral Squamous Cell Carcinoma via Mediating the KDM8/CCNA1 Axis. Biomedicines 2023; 11:2669. [PMID: 37893043 PMCID: PMC10604360 DOI: 10.3390/biomedicines11102669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
The dysregulated expression of cyclin genes can lead to the uncontrolled proliferation of cancer cells. Histone demethylase Jumonji-C domain-containing protein 5 (KDM8, JMJD5) and cyclin A1 (CCNA1) are pivotal in cell cycle progression. A promising candidate for augmenting cancer treatment is Allyl isothiocyanate (AITC), a natural dietary chemotherapeutic and epigenetic modulator. This study aimed to investigate AITC's impact on the KDM8/CCNA1 axis to elucidate its role in oral squamous cell carcinoma (OSCC) tumorigenesis. The expression of KDM8 and CCNA1 was assessed using a tissue microarray (TMA) immunohistochemistry (IHC) assay. In vitro experiments with OSCC cell lines and in vivo experiments with patient-derived tumor xenograft (PDTX) and SAS subcutaneous xenograft tumor models were conducted to explore AITC's effects on their expression and cell proliferation. The results showed elevated KDM8 and CCNA1 levels in the OSCC patient samples. AITC exhibited inhibitory effects on OSCC tumor growth in vitro and in vivo. Additionally, AITC downregulated KDM8 and CCNA1 expression while inducing histone H3K36me2 expression in oral cancer cells. These findings underscore AITC's remarkable anticancer properties against oral cancer, highlighting its potential as a therapeutic option for oral cancer treatment by disrupting the cell cycle by targeting the KDM8/CCNA1 axis.
Collapse
Grants
- TSGH-C01-109017, TSGH-C05-110035, TSGH-C04-111037, TSGH-D-110148, TSGH-D-110149, TSGH-D-110151, TSGH-D-110152, TSGH-D-110154, TSGH-C02-112032 Tri-Service General Hospital, Taiwan, Republic of China
- MAB-E-109003, MAB-D-110003, MND-MAB-110-043, MND-MAB-110-076, MND-MAB-C-111036, MAB-E-111002, MND-MAB-D-111149, MND-MAB-D-112176, MND-MAB-C08-112033 Ministry of National Defense, Taiwan, Republic of China
- MOST 108-2314-B-016-005 Ministry of Science and Technology, Taiwan, Republic of China
- KAFGH-E-111047, KAFGH_E_112061 Kaohsiung Armed Forces General Hospital, Taiwan, Republic of China
- KSVGH112-135 Kaohsiung Veterans General Hospital, Taiwan, Republic of China
- HAFGH_E_112018 Hualien Armed Forces General Hospital, Taiwan, Republic of China
- CTH107A-2C01 Cardinal Tien Hospital, Taipei, Taiwan, Republic of China
Collapse
Affiliation(s)
- Cheng-Chih Hsieh
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- School of Pharmacy and Institute of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Cheng-Yu Yang
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Bo Peng
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Sien-Lin Ho
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
| | - Chang-Huei Tsao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
- Department of Medical Research, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Chih-Kung Lin
- Division of Anatomic Pathology, Taipei Tzu Chi Hospital, New Taipei City 231, Taiwan
| | - Chun-Shu Lin
- Department of Radiation Oncology, Tri-Service General Hospital, National Defense Medical Centre, Taipei 114, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 114, Taiwan
| | - Heng-Yi Lin
- Department of Dentistry, Cardinal Tien Hospital, New Taipei City 231, Taiwan
| | - Hung-Chi Huang
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Dentistry, Hualien Armed Forces General Hospital, Hualien 971, Taiwan
| | - Szu-Chien Chang
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Dentistry, Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Huey-Kang Sytwu
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli 350, Taiwan
| | - Wei-Tso Chia
- Department of Orthopedics, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan
- Department of Nursing, Yuan Pie University of Medical Technology, Hsinchu 302, Taiwan
- Tri-Service General Hospital, Taipei 114, Taiwan
| | - Yuan-Wu Chen
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| |
Collapse
|
3
|
Musat MG, Kwon CS, Masters E, Sikirica S, Pijush DB, Forsythe A. Treatment Outcomes of High-Risk Non-Muscle Invasive Bladder Cancer (HR-NMIBC) in Real-World Evidence (RWE) Studies: Systematic Literature Review (SLR). Clinicoecon Outcomes Res 2022; 14:35-48. [PMID: 35046678 PMCID: PMC8759992 DOI: 10.2147/ceor.s341896] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/18/2021] [Indexed: 12/30/2022]
Abstract
Background To date, there has been limited synthesis of RWE studies in high-risk non-muscle invasive bladder cancer (HR-NMIBC). The objective of this research was to conduct a systematic review of published real-world evidence to better understand the real-world burden and treatment patterns in HR-NMIBC. Methods An SLR was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines with the scope defined by the Population, Intervention Comparators, Outcomes, and Study design (PICOS) criteria. EMBASE, MEDLINE, and Cochrane databases (Jan 2015–Jul 2020) were searched, and relevant congress abstracts (Jan 2018–Jul 2020) identified. The final analysis only included studies that enrolled ≥100 patients with HR-NMIBC from the US, Europe, Canada, and Australia. Results The SLR identified 634 RWE publications in NMIBC, of which 160 studies reported data in HR-NMIBC. The average age of patients in the studies was 71 years, and 79% were males. The rates of BCG intravesical instillations ranged from 3% to 86% (29–95% for induction and 8–83% for maintenance treatment). Five-year outcomes were 17–89% recurrence-free survival (longest survival in patients completing BCG maintenance), 58–89% progression-free survival, 71–96% cancer-specific survival (lowest survival in BCG-unresponsive patients), and 28–90% overall survival (lowest survival in patients who did not receive BCG or instillation therapy). Conclusion BCG treatment rates and survival outcomes in patients with HR-NMIBC vary in the real world, with better survival seen in patients completing maintenance BCG, responding to treatment, and not progressing to muscle-invasive disease. There is a need to better understand the factors associated with BCG use and discontinuation and for an effective treatment that improves outcomes in HR-NMIBC. Generalization of these results is limited by variations in data collection, reporting, and methodologies used across RWE studies.
Collapse
Affiliation(s)
- Mihaela Georgiana Musat
- Evidence Generation, Purple Squirrel Economics, a Wholly Owned Subsidiary of Cytel, Inc., Waltham, MA, USA
| | - Christina Soeun Kwon
- Evidence Generation, Purple Squirrel Economics, a Wholly Owned Subsidiary of Cytel, Inc., Waltham, MA, USA
| | | | - Slaven Sikirica
- Global Health Economics and Outcomes Research, Pfizer, New York, NY, USA
| | - Debduth B Pijush
- Global Health Economics and Outcomes Research, Pfizer, New York, NY, USA
| | - Anna Forsythe
- Value and Access, Purple Squirrel Economics, a Wholly Owned Subsidiary of Cytel, Inc., Waltham, MA, USA
| |
Collapse
|
4
|
Bourova-Flin E, Derakhshan S, Goudarzi A, Wang T, Vitte AL, Chuffart F, Khochbin S, Rousseaux S, Aminishakib P. The combined detection of Amphiregulin, Cyclin A1 and DDX20/Gemin3 expression predicts aggressive forms of oral squamous cell carcinoma. Br J Cancer 2021; 125:1122-1134. [PMID: 34290392 PMCID: PMC8505643 DOI: 10.1038/s41416-021-01491-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/02/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
Background Large-scale genetic and epigenetic deregulations enable cancer cells to ectopically activate tissue-specific expression programmes. A specifically designed strategy was applied to oral squamous cell carcinomas (OSCC) in order to detect ectopic gene activations and develop a prognostic stratification test. Methods A dedicated original prognosis biomarker discovery approach was implemented using genome-wide transcriptomic data of OSCC, including training and validation cohorts. Abnormal expressions of silent genes were systematically detected, correlated with survival probabilities and evaluated as predictive biomarkers. The resulting stratification test was confirmed in an independent cohort using immunohistochemistry. Results A specific gene expression signature, including a combination of three genes, AREG, CCNA1 and DDX20, was found associated with high-risk OSCC in univariate and multivariate analyses. It was translated into an immunohistochemistry-based test, which successfully stratified patients of our own independent cohort. Discussion The exploration of the whole gene expression profile characterising aggressive OSCC tumours highlights their enhanced proliferative and poorly differentiated intrinsic nature. Experimental targeting of CCNA1 in OSCC cells is associated with a shift of transcriptomic signature towards the less aggressive form of OSCC, suggesting that CCNA1 could be a good target for therapeutic approaches.
Collapse
Affiliation(s)
- Ekaterina Bourova-Flin
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France
| | - Samira Derakhshan
- Oral and Maxillofacial Pathology Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Goudarzi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tao Wang
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France
| | - Anne-Laure Vitte
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France
| | - Florent Chuffart
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France
| | - Saadi Khochbin
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France
| | - Sophie Rousseaux
- CNRS UMR 5309/INSERM U1209/University Grenoble-Alpes/Institute for Advanced Biosciences, La Tronche, France.
| | - Pouyan Aminishakib
- Oral and Maxillofacial Pathology Department, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Institute Hospital, IKHC, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
CCNA1 gene as a potential diagnostic marker in papillary thyroid cancer. Acta Histochem 2020; 122:151635. [PMID: 33007517 DOI: 10.1016/j.acthis.2020.151635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 09/03/2020] [Accepted: 09/23/2020] [Indexed: 11/24/2022]
Abstract
The malignancy that most affects the endocrine system is thyroid neoplasm, with an increasing incidence over the years. The most prevalent histological type of the carcinomas that affect the thyroid gland is papillary carcinoma with a prevalence of 80 % worldwide. The current diagnostic methodology may present inconclusive results, emphasizing the need for new effective and sensitive techniques to aid the diagnosis. For this, it is necessary to understand molecular and protein mechanisms in the identification of diagnostic and predictive markers in the lesions. The Cyclin A1 protein, encoded by the CCNA1 gene, is an important cell cycle regulator, belonging to the MAPK/ERK signaling pathway directly involved with thyroid cancer. The aim of this study was to evaluate the CCNA1 gene and Cyclin A1 protein expression in papillary thyroid carcinoma, follicular thyroid carcinoma, and benign thyroid lesions, by real time quantitative PCR and immunohistochemistry analysis, respectively, to verify their roles as potential diagnostic and predictive markers to future applications in the clinical routine. Overexpression of CCNA1 gene was observed in the papillary carcinoma group compared to the normal group (P = 0.0023), benign lesions (P = 0.0011), colloid goiter (P = 0.0124), and follicular carcinoma (P = 0.0063). No differential expression was observed in the papillary primary tumor group from negative lymph nodes compared with the one from positive lymph nodes (P = 0.3818). Although an increased expression of Cyclin A1 was observed in the PTC group compared to the other one in the IHC analysis, no significant difference was observed (Fisher's exact Test). A Cyclin A1 overexpression was detected with weak to mid-moderate immunoreactivity in the benign group (k = 0.56), (score 1.5); mid-moderate to moderate in the goiter group (k = 0.58); weak in the FTC group (k = 0.33); and mid-moderate to moderate in the PTC group (k = 0.48). Due to the small sample size in the IHC analysis and to the fact that not all RNA is translated into protein, the diagnostic potential of Cyclin A1 could not be assessed. However, these findings highlight the potential of the CCNA1 gene as a diagnostic marker for papillary thyroid carcinoma.
Collapse
|
6
|
He X, Li S, Shi W, Lin Q, Ma J, Liu Y, Feng T, Cao X. Cyclin A1 is associated with poor prognosis in oesophageal squamous cell carcinoma. Oncol Lett 2019; 18:706-712. [PMID: 31289545 PMCID: PMC6546994 DOI: 10.3892/ol.2019.10377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of cyclin A1 (CCNA1) is implicated in the carcinogenesis, progression and metastasis of many types of solid tumours. In the present study, an mRNA single-channel expression profile chip experiment revealed that the CCNA1 mRNA levels in oesophageal squamous cell carcinoma (ESCC) were increased >10-fold compared with those in the adjacent non-cancer tissues. Reverse transcription-quantitative polymerase chain reaction and immunohistochemistry analyses were performed to additionally investigate the role of CCNA1 in the development and progression of ESCC in patients treated by radical resection of the oesophagus. The association between CCNA1 mRNA expression and the clinicopathological parameters of patients with ESCC was statistically analysed. The results indicated that upregulation of CCNA1 occurred in ~70% of patients with ESCC, and increased CCNA1 mRNA expression was significantly associated with advanced clinical stage, lymph node metastasis, invasiveness and poor clinical outcome, including disease-free survival and overall survival rates. Taken together, the data suggested that CCNA1 had an important function in ESCC development and progression, and may serve as a prognostic biomarker and therapeutic target in ESCC.
Collapse
Affiliation(s)
- Xiaoting He
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Medical Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214043, P.R. China
| | - Suqing Li
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Weihong Shi
- Department of Clinical Medicine, Yancheng Vocational Institute of Health Sciences, Yancheng, Jiangsu 224005, P.R. China
| | - Qingfeng Lin
- Department of Medical Oncology, Jiangyin People's Hospital, Jiangyin, Jiangsu 214400, P.R. China
| | - Jian Ma
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yang Liu
- Department of Pharmacy, No. 401 Hospital of Chinese People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Tingting Feng
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xiufeng Cao
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Surgery, Nanjing Medical University, Taikang Xianlin Drum Tower Hospital, Jiangsu 210000, P.R. China
| |
Collapse
|
7
|
Li XS, Shen FZ, Huang LY, Hui L, Liu RH, Ma YJ, Jin BZ. lncRNA small nucleolar RNA host gene 20 predicts poor prognosis in glioma and promotes cell proliferation by silencing P21. Onco Targets Ther 2019; 12:805-814. [PMID: 30774368 PMCID: PMC6352861 DOI: 10.2147/ott.s192641] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background In multiple cancers, long non-coding RNA small nucleolar RNA host gene 20 (lncRNA SNHG20) is generally dysregulated. In the present study, both the biological role and clinicopathological value of lncRNA SNHG20 in glioma are explored. Methods Real-time PCR was employed to determine lncRNA SNHG20 expression in glioma patients. The prognostic role of expression of lncRNA SNHG20 was evaluated in a retrospective cohort study. In addition, the association between lncRNA SNHG20 expression and the clinicopathological features of glioma patients, such as tumor recurrence, survival status, follow-up time, WHO grade, resection extent, tumor location, Karnofsky performance scale score, cystic change, tumor size, gender and age, was discussed. By constructing and transfecting siRNAs that targeted lncRNA SNHG20 into the glioma U87 cells, the effects of lncRNA SNHG20 on the proliferation and cell cycle of U87 cells were assessed through cell counting kit-8, colony formation and cell cycle assays, respectively. In addition, Western blot and real-time PCR measured the expression levels of P21 and CCNA1 in U87 cells after being transfected with SNHG20 siRNA. Results Our results suggested the high expression of lncRNA SNHG20 in human glioma tissues compared with normal brain tissues, which was related to recurrence-free survival and poor overall survival in glioma patients. According to the existing retrospective cohort study, high lncRNA SNHG20 expression was associated with tumor size, extent of resection, WHO grade, follow-up time, survival status and recurrence. Besides, knocking down the expression of lncRNA SNHG20 could inhibit the proliferation and colony formation abilities of glioma U87 cells through cell cycle arrest. Consequently, the expression of CCNA1 was inhibited, and the expression of P21 was up-regulated in U87 cells. Conclusion A high lncRNA SNHG20 expression level predicts the poor prognosis for glioma patients. Moreover, lncRNA SNHG20 can promote glioma proliferation through silencing P21 and thus lncRNA SNHG20 is an independent potential prognostic biomarker for glioma patients.
Collapse
Affiliation(s)
- Xiang-Sheng Li
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| | - Fa-Zheng Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| | - Li-Yong Huang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| | - Lei Hui
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| | - Rui-Hua Liu
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| | - Yan-Juan Ma
- Department of Emergency, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China
| | - Bao-Zhe Jin
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453000, Henan Province, People's Republic of China,
| |
Collapse
|
8
|
Munari E, Caliò A, Brunelli M, Martignoni G. Predicting progression in T1 non-muscle-invasive bladder cancer: back to histology. BJU Int 2018; 122:914-915. [PMID: 30460787 DOI: 10.1111/bju.14465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria Hospital, Negrar, VR, Italy
| | - Anna Caliò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,Department of Pathology, Pederzoli Hospital, Peschiera del Garda, VR, Italy
| |
Collapse
|
9
|
|
10
|
HDAC inhibition as a treatment concept to combat temsirolimus-resistant bladder cancer cells. Oncotarget 2017; 8:110016-110028. [PMID: 29299126 PMCID: PMC5746361 DOI: 10.18632/oncotarget.22454] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/12/2017] [Indexed: 12/16/2022] Open
Abstract
Introduction Although the mechanistic target of rapamycin (mTOR) might be a promising molecular target to treat advanced bladder cancer, resistance develops under chronic exposure to an mTOR inhibitor (everolimus, temsirolimus). Based on earlier studies, we proposed that histone deacetylase (HDAC) blockade might circumvent resistance and investigated whether HDAC inhibition has an impact on growth of bladder cancer cells with acquired resistance towards temsirolimus. Results The HDAC inhibitor valproic acid (VPA) significantly inhibited growth, proliferation and caused G0/G1 phase arrest in RT112res and UMUC-3res. cdk1, cyclin B, cdk2, cyclin A and Skp1 p19 were down-regulated, p27 was elevated. Akt-mTOR signaling was deactivated, whereas acetylation of histone H3 and H4 in RT112res and UMUC-3res increased in the presence of VPA. Knocking down cdk2 or cyclin A resulted in a significant growth blockade of RT112res and UMUC-3res. Materials And Methods Parental (par) and resistant (res) RT112 and UMUC-3 cells were exposed to the HDAC inhibitor VPA. Tumor cell growth, proliferation, cell cycling and expression of cell cycle regulating proteins were then evaluated. siRNA blockade was used to investigate the functional impact of the proteins. Conclusions HDAC inhibition induced a strong response of temsirolimus-resistant bladder cancer cells. Therefore, the temsirolimus-VPA-combination might be an innovative strategy for bladder cancer treatment.
Collapse
|
11
|
Hoque MO. Differences in the Molecular Characteristics of Bladder Cancer between Smokers and Nonsmokers. Eur Urol Focus 2017; 4:98-99. [PMID: 28753779 DOI: 10.1016/j.euf.2016.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/09/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Mohammad Obaidul Hoque
- Department of Otolaryngology and Head and Neck Surgery, Urology, and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Eskaros AR, Egloff SAA, Boyd KL, Richardson JE, Hyndman ME, Zijlstra A. Larger core size has superior technical and analytical accuracy in bladder tissue microarray. J Transl Med 2017; 97:335-342. [PMID: 28112755 DOI: 10.1038/labinvest.2016.151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 11/16/2016] [Accepted: 12/05/2016] [Indexed: 12/17/2022] Open
Abstract
The construction of tissue microarrays (TMAs) with cores from a large number of paraffin-embedded tissues (donors) into a single paraffin block (recipient) is an effective method of analyzing samples from many patient specimens simultaneously. For the TMA to be successful, the cores within it must capture the correct histologic areas from the donor blocks (technical accuracy) and maintain concordance with the tissue of origin (analytical accuracy). This can be particularly challenging for tissues with small histological features such as small islands of carcinoma in situ (CIS), thin layers of normal urothelial lining of the bladder, or cancers that exhibit intratumor heterogeneity. In an effort to create a comprehensive TMA of a bladder cancer patient cohort that accurately represents the tumor heterogeneity and captures the small features of normal and CIS, we determined how core size (0.6 vs 1.0 mm) impacted the technical and analytical accuracy of the TMA. The larger 1.0 mm core exhibited better technical accuracy for all tissue types at 80.9% (normal), 94.2% (tumor), and 71.4% (CIS) compared with 58.6%, 85.9%, and 63.8% for 0.6 mm cores. Although the 1.0 mm core provided better tissue capture, increasing the number of replicates from two to three allowed with the 0.6 mm core compensated for this reduced technical accuracy. However, quantitative image analysis of proliferation using both Ki67+ immunofluorescence counts and manual mitotic counts demonstrated that the 1.0 mm core size also exhibited significantly greater analytical accuracy (P=0.004 and 0.035, respectively, r2=0.979 and 0.669, respectively). Ultimately, our findings demonstrate that capturing two or more 1.0 mm cores for TMA construction provides superior technical and analytical accuracy over the smaller 0.6 mm cores, especially for tissues harboring small histological features or substantial heterogeneity.
Collapse
Affiliation(s)
- Adel Rh Eskaros
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shanna A Arnold Egloff
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce E Richardson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Eric Hyndman
- Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andries Zijlstra
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
13
|
Li C, Zhou L, He J, Fang XQ, Zhu SW, Xiong MM. Increased long noncoding RNA SNHG20 predicts poor prognosis in colorectal cancer. BMC Cancer 2016; 16:655. [PMID: 27543107 PMCID: PMC4992210 DOI: 10.1186/s12885-016-2719-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
Background Long noncoding RNAs (lncRNAs) have been suggested to be involved in the development and progression of malignancies. However, the investigation of small nucleolar RNA host gene 20 (SNHG20) on cancer progression remains unknown. The present study aims to explore the clinical significance of SNHG20 and its potential molecular mechanism in colorectal cancer (CRC). Methods Quantitative real-time PCR (qRT-PCR) was used to measure the SNHG20 expression in a total of 107 CRC tissues and CRC cell lines. Loss of function approach was employed to explore the biological roles of SNHG20 in vitro. Its potential molecular mechanism was further verified by western blotting and qRT-PCR. Results The results suggested that SNHG20 expression was significantly upregulated in CRC tissues compared to corresponding normal tissues from 107 CRC patients. High expression of SNHG20 was remarkably associated with advanced TNM stage in patients with CRC. Multivariate analyses unraveled that SNHG20 expression was an independent prognostic factor for overall survival in CRC patients. Further functional assays revealed that knockdown of SNHG20 suppressed cell proliferation, invasion and migration, and cell cycle progression in CRC cells. Moreover, SNHG20 regulated cell growth through modulation of a series of cell cycle-associated genes. Conclusions Our findings suggest that dysregulation of SNHG20 participates in CRC progression and may serve as a potential therapeutic target in CRC patients.
Collapse
Affiliation(s)
- Cong Li
- Department of Minimally Invasive Surgery, The People's Hospital of Chizhou, Chizhou, 247000, China
| | - Li Zhou
- Department of Minimally Invasive Surgery, The People's Hospital of Chizhou, Chizhou, 247000, China
| | - Jun He
- Department of Minimally Invasive Surgery, The People's Hospital of Chizhou, Chizhou, 247000, China
| | - Xue-Qing Fang
- Department of Minimally Invasive Surgery, The People's Hospital of Chizhou, Chizhou, 247000, China
| | - Shao-Wen Zhu
- Department of Minimally Invasive Surgery, The People's Hospital of Chizhou, Chizhou, 247000, China
| | - Mao-Ming Xiong
- Department of General Surgery, First Hospital Affiliated to Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
14
|
Bäumer N, Bäumer S, Haak M, Koschmieder S, Schönig K, Berdel WE, Müller-Tidow C. A Limited Role for the Cell Cycle Regulator Cyclin A1 in Murine Leukemogenesis. PLoS One 2015; 10:e0129147. [PMID: 26080083 PMCID: PMC4469679 DOI: 10.1371/journal.pone.0129147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/05/2015] [Indexed: 12/04/2022] Open
Abstract
The quest for novel therapeutic targets in acute myeloid leukemia (AML) is still ongoing. One of such targets, cyclin A1, was shown to be overexpressed in AML including AML stem cells. However, the function of cyclin A1 in AML is largely unknown, and the data on its impact on patients´ survival remain controversial. Therefore, we developed a transgenic mouse model of stem cell-directed inducible cyclin A1 overexpression and crossed these mice with PML-RARα-knockin mice, which develop an AML M3-like phenotype. To observe the effects of cyclin A1 loss-of-function, we also crossed PML-RARα-knockin mice to cyclin A1-knockout mice. Neither overexpression nor loss of cyclin A1 significantly altered leukemogenesis in PML-RARα-knockin mice. These findings imply that upregulation of cyclin A1 is not essential for leukemogenesis. Our data suggest that cyclin A1 does not represent a suitable target for AML therapy.
Collapse
Affiliation(s)
- Nicole Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- * E-mail: (NB); (CMT)
| | - Sebastian Bäumer
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Miriam Haak
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Steffen Koschmieder
- Department of Medicine (Hematology, Oncology, Hemostaseology, and SCT), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Kai Schönig
- Central Institute of Mental Health, Department of Molecular Biology, Heidelberg University, Mannheim, Germany
| | - Wolfgang E. Berdel
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
| | - Carsten Müller-Tidow
- Department of Medicine A, Hematology/Oncology, University of Muenster, Muenster, Germany
- Dept. of Medicine IV, Hematology and Oncology, University of Halle, Halle, Germany
- * E-mail: (NB); (CMT)
| |
Collapse
|