1
|
Zhu J, Zhang L. Construction of DNA methylation-based nomogram for predicting biochemical-recurrence-free survival in prostate cancer. Medicine (Baltimore) 2022; 101:e32205. [PMID: 36626527 PMCID: PMC9750565 DOI: 10.1097/md.0000000000032205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
This study aimed to develop a DNA methylation-based nomogram for predicting biochemical recurrence in patients with prostate cancer. A DNA methylation signature was obtained via univariate, lasso, and stepwise multivariate Cox regression models. A 11-DNA methylation signature yielded a high evaluative performance for biochemical-recurrence-free survival. Cox regression analysis indicated that 11-DNA methylation signature and Gleason score served as independent risk factors. A nomogram was constructed based on the 11-DNA methylation signature and Gleason score, and C-index as well as the calibration plots demonstrated good performance and clinical application of the nomogram. A DNA methylation-associated nomogram serve as a prognosis stratification tool to predict the biochemical recurrence of prostate cancer patients after radical prostatectomy.
Collapse
Affiliation(s)
- Jiayu Zhu
- Department of Oncology, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine (Xiaoshan Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Le Zhang
- Department of Oncology, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine (Xiaoshan Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- * Correspondence: Le Zhang, Department of Oncology, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine (Xiaoshan Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang 310016, China (e-mail: )
| |
Collapse
|
2
|
Xie Q, Liu R, Zou Z, Feng Y, Huang Y, Xu G, Sun W, Liang Y, Zhong W. MYPT1 inhibits the metastasis of renal clear cell carcinoma via the MAPK8/N-cadherin pathway. FEBS Open Bio 2022; 12:2083-2095. [PMID: 36106411 PMCID: PMC9623519 DOI: 10.1002/2211-5463.13487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/23/2022] [Accepted: 09/14/2022] [Indexed: 01/25/2023] Open
Abstract
Myosin phosphatase target subunit 1 (MYPT1) is a subunit of myosin phosphatase that is capable of regulating smooth muscle contraction. MYPT1 has been reported to be involved in a wide variety of tumours, but its expression and biological functions in renal clear cell carcinoma (ccRCC) remain obscure. Herein, we analysed the relationship between patient clinicopathological characteristics and MYPT1 expression levels in ccRCC patients using a tissue microarray (TMA) and data retrieved from the TCGA-KIRC dataset. MYPT1 was overexpressed or depleted using siRNA in ccRCC cells to assess the effects on migration and invasion in vitro and in vivo. Additionally, RNA-sequencing and bioinformatics analysis were performed to investigate the precise mechanism. MYPT1 expression in ccRCC tissues was observed to be lower than that in nonmalignant tissues (P < 0.05). In addition, MYPT1 downregulation was closely linked to advanced pathological stage (P < 0.05), and poor OS (overall survival; P < 0.05). Functionally, increased expression of MYPT1 suppressed ccRCC migration and invasion in vitro, and inhibited tumour metastasis in vivo. In addition, MYPT1 overexpression exerted its suppressive effects via the MAPK8/N-cadherin pathway in ccRCC.
Collapse
Affiliation(s)
- Qingling Xie
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ren Liu
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Zhihao Zou
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yiqiao Huang
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Guibin Xu
- Department of UrologyThe Fifth Affiliated Hospital of Guangzhou Medical UniversityChina
| | - Wei Sun
- Department of Urology, Huadu District People's HospitalSouthern Medical UniversityGuangzhouChina
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Weide Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
3
|
Global Reprogramming of Apoptosis-Related Genes during Brain Development. Cells 2021; 10:cells10112901. [PMID: 34831124 PMCID: PMC8616463 DOI: 10.3390/cells10112901] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/20/2022] Open
Abstract
To enable long-term survival, mammalian adult neurons exhibit unique apoptosis competence. Questions remain as to whether and how neurons globally reprogram the expression of apoptotic genes during development. We systematically examined the in vivo expression of 1923 apoptosis-related genes and associated histone modifications at eight developmental ages of mouse brains. Most apoptotic genes displayed consistent temporal patterns across the forebrain, midbrain, and hindbrain, suggesting ubiquitous robust developmental reprogramming. Although both anti- and pro-apoptotic genes can be up- or downregulated, half the regulatory events in the classical apoptosis pathway are downregulation of pro-apoptotic genes. Reduced expression in initiator caspases, apoptosome, and pro-apoptotic Bcl-2 family members restrains effector caspase activation and attenuates neuronal apoptosis. The developmental downregulation of apoptotic genes is attributed to decreasing histone-3-lysine-4-trimethylation (H3K4me3) signals at promoters, where histone-3-lysine-27-trimethylation (H3K27me3) rarely changes. By contrast, repressive H3K27me3 marks are lost in the upregulated gene groups, for which developmental H3K4me3 changes are not predictive. Hence, developing brains remove epigenetic H3K4me3 and H3K27me3 marks on different apoptotic gene groups, contributing to their downregulation and upregulation, respectively. As such, neurons drastically alter global apoptotic gene expression during development to transform apoptosis controls. Research into neuronal cell death should consider maturation stages as a biological variable.
Collapse
|
4
|
Wang M, Wu Q, Fang M, Huang W, Zhu H. miR-152-3p Sensitizes Glioblastoma Cells Towards Cisplatin Via Regulation Of SOS1. Onco Targets Ther 2019; 12:9513-9525. [PMID: 31807027 PMCID: PMC6857816 DOI: 10.2147/ott.s210732] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022] Open
Abstract
Background Accumulating evidences suggest that microRNAs (miRNAs) play key roles in mediating glioblastoma progression. Decreased expression of miR-152-3p was reported in several cancer types including glioblastoma. Methods The sensitivity of glioblastoma cells to cisplatin was assessed by the cell counting kit-8 assay and flow cytometry analysis. The expression of miR-152-3p was determined by RT-qPCR method. Bioinformatic analysis, dual luciferase reporter assay and Western blot were used to explore the target gene of miR-152-3p. The association between miR-152-3p and SOS1 was confirmed in glioblastoma tissues by Pearson correlation analysis. Results In the current study, we discovered that overexpression of miR-152-3p increased cisplatin sensitivity while inhibition of miR-152-3p decreased cisplatin sensitivity in glioblastoma cells (T98G and U87). In addition, miR-152-3p augmented cell apoptosis induced by cisplatin treatment. It was further predicted and validated that SOS1, a protein involved in regulating chemotherapy sensitivity, was a direct target gene of miR-152-3p. SOS1 was proven to suppress the cytotoxic effect of cisplatin in glioblastoma. Transfection of recombinant SOS1 could effectively reverse the increased cisplatin sensitivity induced by miR-152-3p overexpression in T98G. Furthermore, overexpression of SOS1 reduced the percentage of apoptotic cells increased by miR-152-3p mimic in the presence of cisplatin in T98G. More importantly, a significant negative correlation between miR-152-3p levels and SOS1 levels was observed in glioblastoma tissues collected from 40 patients. Conclusion Our study identified miR-152-3p as a chemotherapy sensitizer in glioblastoma.
Collapse
Affiliation(s)
- Meihua Wang
- Department of Pathology, Changzhou Tumor Hospital, Affiliated to Soochow University, Changzhou, People's Republic of China
| | - Qi Wu
- Department of Histology and Embryology, Heze Medical College, Heze, People's Republic of China
| | - Mingming Fang
- Department of Radiotherapy, Changzhou Tumor Hospital, Affiliated to Soochow University, Changzhou, People's Republic of China
| | - Wu Huang
- Department of Neurosurgery, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, People's Republic of China
| | - Hong Zhu
- Department of Radiation Oncology, Minhang Branch of Cancer Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
5
|
Xu P, Zhang G, Hou S, Sha LG. MAPK8 mediates resistance to temozolomide and apoptosis of glioblastoma cells through MAPK signaling pathway. Biomed Pharmacother 2018; 106:1419-1427. [PMID: 30119215 DOI: 10.1016/j.biopha.2018.06.084] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE In this study, we aimed to evaluate the expression and functions of MAPK8 in temozolomide (TMZ) -resistant glioblastoma cells as well as to explore the mechanism of TMZ resistance in glioblastoma cells. METHODS Gene Expression Omnibus (GEO) database was used for identifying the differentially expressed genes (DEGs) in TMZ resistant samples. The functional partner genes of TMZ were screened out by Gene-drug interaction network (STITCH) and the glioblastoma-related genes were selected by gene search engine with evidence sentences (Digsee). The interactions among identified DEGs and glioblastoma-related genes were detected by Search Tool for the Retrieval of Interacting Genes (STRING). The dysregulated pathways were identified by Gene set enrichment analysis (GSEA). qRT-PCR was performed to detect the expression level of MAPK8 in glioblastoma cells. Western blot was used to detect the expressions of MAPK8 and MAPK signaling pathway-related proteins. MTT assay was utilized to measure the cell viability of TMZ sensitive and resistant cells. Colony formation assay was performed to detect the clone ability and flow cytometry (FCM) assay was applied to identify the apoptosis rate of TMZ resistant glioblastoma cells. RESULTS MAPK8 was one of the DEGs and was up-regulated in TMZ resistant glioblastoma cells. The MAPK signaling pathway was activated in TMZ resistant glioblastoma cells under the condition of over-expression of MAPK8. The inhibition of MAPK8 restrained the colony formation, inducing apoptosis of TMZ resistant glioblastoma cells and suppressed the MAPK signaling pathway. CONCLUSION MAPK8 promoted the resistance to TMZ, accelerated cell proliferation and inhibited the apoptosis of glioblastoma cells via activating MAPK signaling pathway.
Collapse
Affiliation(s)
- Peng Xu
- The Fourth Department of Geronotology, Jinan Military General Hospital, Jinan, 250031, Shandong, China
| | - Guofeng Zhang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Shaanxi, 710032, Xi'an, China
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Pudong, 201399, Shanghai, China.
| | - Long-Gui Sha
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Pudong, 201399, Shanghai, China.
| |
Collapse
|
6
|
Singla S, Chen J, Sethuraman S, Sysol JR, Gampa A, Zhao S, Machado RF. Loss of lung WWOX expression causes neutrophilic inflammation. Am J Physiol Lung Cell Mol Physiol 2017; 312:L903-L911. [PMID: 28283473 DOI: 10.1152/ajplung.00034.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor WW domain-containing oxidoreductase (WWOX) exhibits regulatory interactions with an array of transcription factors and signaling molecules that are positioned at the well-known crossroads between inflammation and cancer. WWOX is also subject to downregulation by genotoxic environmental exposures, making it of potential interest to the study of lung pathobiology. Knockdown of lung WWOX expression in mice was observed to cause neutrophil influx and was accompanied by a corresponding vascular leak and inflammatory cytokine production. In cultured human alveolar epithelial cells, loss of WWOX expression resulted in increased c-Jun- and IL-8-dependent neutrophil chemotaxis toward cell monolayers. WWOX was observed to directly interact with c-Jun in these cells, and its absence resulted in increased nuclear translocation of c-Jun. Finally, inhibition of the c-Jun-activating kinase JNK abrogated the lung neutrophil influx observed during WWOX knockdown in mice. Altogether, these observations represent a novel mechanism of pulmonary neutrophil influx that is highly relevant to the pathobiology and potential treatment of a number of different lung inflammatory conditions.
Collapse
Affiliation(s)
- Sunit Singla
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Jiwang Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Shruthi Sethuraman
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Justin R Sysol
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Amulya Gampa
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Shuangping Zhao
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
7
|
Pandey V, Bhaskara VK, Babu PP. Implications of mitogen-activated protein kinase signaling in glioma. J Neurosci Res 2015; 94:114-27. [PMID: 26509338 DOI: 10.1002/jnr.23687] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/22/2015] [Accepted: 10/12/2015] [Indexed: 12/22/2022]
Abstract
Gliomas are the most common primary central nervous system tumors. Gliomas originate from astrocytes, oligodendrocytes, and neural stem cells or their precursors. According to WHO classification, gliomas are classified into four different malignant grades ranging from grade I to grade IV based on histopathological features and related molecular aberrations. The induction and maintenance of these tumors can be attributed largely to aberrant signaling networks. In this regard, the mitogen-activated protein kinase (MAPK) network has been widely studied and is reported to be severely altered in glial tumors. Mutations in MAPK pathways most frequently affect RAS and B-RAF in the ERK, c-Jun N-terminal kinase (JNK), and p38 pathways leading to malignant transformation. Also, it is linked to both inherited and sequential accumulations of mutations that control receptor tyrosine kinase (RTK)-activated signal transduction pathways, cell cycle growth arrest pathways, and nonresponsive cell death pathways. Genetic alterations that modulate RTK signaling can also alter several downstream pathways, including RAS-mediated MAP kinases along with JNK pathways, which ultimately regulate cell proliferation and cell death. The present review focuses on recent literature regarding important deregulations in the RTK-activated MAPK pathway during gliomagenesis and progression.
Collapse
Affiliation(s)
- Vimal Pandey
- Laboratory of Neuroscience, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, AP, India
| | - Vasantha Kumar Bhaskara
- Laboratory of Neuroscience, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, AP, India
| | - Phanithi Prakash Babu
- Laboratory of Neuroscience, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, AP, India
| |
Collapse
|