1
|
Li L, Cui J, Li X, Zhu Y, Wu H, Zhou L. Prmt1-mediated histone H4R3me2a methylation regulates the proliferation, migration and invasion of laryngeal cancer cells by affecting the expression level of NCOA5. Front Oncol 2024; 14:1489164. [PMID: 39741976 PMCID: PMC11685131 DOI: 10.3389/fonc.2024.1489164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Background Laryngeal cancer is a common head and neck cancer, and its occurrence and development are closely related to a variety of epigenetic modifications. protein arginine methyltransferase 1 (PRMT1) is an important type I protein arginine methyltransferase, which catalyzes the monomethylation and asymmetric dimethylation of arginine and participates in the occurrence and development of a variety of cancers. Current research has found that the expression of PRMT1 is increased in laryngeal carcinoma tissues. Histone modifications play a key role in regulating gene expression and maintaining cellular function. In particular, histone H4 arginine 3 dimethylation (H4R3me2a) has been shown to be associated with the development of a variety of cancers. Nuclear Receptor Coactivator 5 (NCOA5) is an important nuclear receptor coactivator, which regulates gene expression through the interaction between various nuclear receptors and other transcription factors. The present study aimed to investigate how PRMT1-mediated H4R3me2a methylation affects the proliferation, migration and invasion of laryngeal cancer cells and to verify the role of NCOA5 in this process. Methods The expression of PRMT1 and NCOA5 was inhibited by siRNA mediated gene knockdown in laryngeal cancer cells. The changes of H4R3me2a protein levels were detected by Western Blotting, and cell proliferation, migration and invasion abilities were evaluated by CCK-8, cell scratch assay, Transwell migration and invasion assay. RT-qPCR was used to detect the mRNA expression levels of related genes. The overexpression experiment of NCOA5 was carried out by constructing overexpression vector to verify its effect. Results After PRMT1 knockdown, the expression of H4R3me2a in laryngeal cancer cells was significantly decreased, and the cell proliferation, migration and invasion abilities were weakened. Similarly, knockdown of NCOA5 expression also resulted in decreased H4R3me2a levels and attenuated cell proliferation, migration and invasion. Overexpression of NCOA5 partially restored H4R3me2a levels and cell proliferation, migration and invasion abilities.
Collapse
Affiliation(s)
| | | | | | | | | | - Lanzhu Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
2
|
Li Z, Yuan X, Wang Y, Sun Z, Ao J. DNAJA1 positively regulates amino acid-stimulated milk protein and fat synthesis in bovine mammary epithelial cells. Cell Biochem Funct 2024; 42:e3918. [PMID: 38269516 DOI: 10.1002/cbf.3918] [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: 10/25/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Several cellular processes, including the recovery of misfolded proteins, the folding of polypeptide chains, transit of polypeptides across the membrane, construction and disassembly of protein complexes, and modulation of protein control, are carried out by DnaJ homolog subfamily A member 1 (DNAJA1), which belongs to the DnaJ heat-shock protein family. It is unknown if DNAJA1 regulates the production of milk in bovine mammary epithelium cells (BMECs). Methionine and leucine increased DNAJA1 expression and nuclear location, as seen by us. In contrast to DNAJA1 knockdown, overexpression of DNAJA1 boosted the production of milk proteins and fats as well as mammalian target of rapamycin (mTOR) and sterol regulatory element binding protein-1c (SREBP-1c). As a result of amino acids, mTOR and SREBP-1c gene expression are stimulated, and DNAJA1 is a positive regulator of BMECs' amino acid-induced controlled milk protein and fat production.
Collapse
Affiliation(s)
- Zhuolin Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Xiaohan Yuan
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Yuanhao Wang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| | - Zheya Sun
- College of Food Science, Northeast Agricultural University, Harbin, China
| | - Jinxia Ao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, Northeast Agricultural University, Harbin, China
| |
Collapse
|
3
|
Wang T, Zhang F, Zhang P. Role of the TPX2/NCOA5 axis in regulating proliferation, migration, invasion and angiogenesis of breast cancer cells. Exp Ther Med 2023; 25:304. [PMID: 37229326 PMCID: PMC10203914 DOI: 10.3892/etm.2023.12003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/17/2023] [Indexed: 05/27/2023] Open
Abstract
Breast cancer is a common malignant tumor in women. Increasing evidence has demonstrated that nuclear receptor coactivator 5 (NCOA5) and targeting protein for xenopus kinesin-like protein 2 (TPX2) serve vital roles in the progression of breast cancer. However, to the best of our knowledge, the molecular mechanisms underlying the involvement of TPX2/NCOA5 in the development of breast cancer are not fully understood at present. In the present study, the expression levels of NCOA5 and TPX2 were compared between paired non-tumor and tumor tissues of patients with breast cancer using the TNMplot tool. Expression differences of NCOA5 and TPX2 in human breast epithelial cell lines (MCF10A and MCF12A) and human breast cancer cell lines (MCF7 and T47D) were assessed via reverse transcription-quantitative PCR and western blotting. Additionally, proliferation, migration and invasion of breast cancer cells were determined via Cell Counting Kit-8, would healing and transwell assays. In vitro angiogenesis was determined using a tube formation assay. Furthermore, TPX2 was identified as a high-confidence NCOA5 interactor based on BioPlex network data sets. A co-immunoprecipitation assay was adopted to confirm the interaction between TPX2 and NCOA5. The present study revealed that TPX2 and NCOA5 were highly expressed in breast cancer cells. TPX2 interacted with NCOA5 and there was a positive association between TPX2 and NCOA5 expression. NOCA5 knockdown repressed the proliferation, migration, invasion and in vitro angiogenesis of breast cancer cells. In addition, TPX2 knockdown suppressed the proliferation, migration and invasion of breast cancer cells, and inhibited in vitro angiogenesis, and all of these effects were reversed following NCOA5 overexpression. In conclusion, NCOA5 was a downstream target of TPX2 in enhancing proliferation, migration, invasion and angiogenesis of breast cancer cells.
Collapse
Affiliation(s)
- Tian Wang
- Department of Oncology Hematology, Yan'an People's Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Fulin Zhang
- Department of Oncology Hematology, Yan'an People's Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Peirong Zhang
- Department of Pathology, Yantian District People's Hospital, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
4
|
Song X, Qian D, Dai P, Li Q, Xi Q, Sun K. Expression and clinical significance of NCOA5 in epithelial ovarian cancer. Front Oncol 2023; 13:1117033. [PMID: 37197435 PMCID: PMC10183562 DOI: 10.3389/fonc.2023.1117033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
Background Nuclear receptor coactivator 5 (NCOA5) plays a significant role in the progression of human cancer. However, its expression in epithelial ovarian cancer (EOC) is unknown. The current study was designed to explore to investigate the clinical significance of NCOA5 and its correlation with the prognosis of EOC. Methods Immunohistochemistry was used to detect the expression of NCOA5 in 60 patients with EOC in this retrospective study and statistical analysis was performed to assess its relevance to clinicopathologic features and survival. Results NCOA5 expression was significantly higher in EOC than in normal ovarian tissues (P < 0.001). Its expression level was significantly correlated with FIGO stage (P <0. 05) and subtypes of ovarian cancer (P < 0.001), while not correlation with age , differentiation and lymph node metastasis (P>0.05). Correlation analysis showed that NCOA5 was significantly correlated with CA125 (P < 0.001) and HE4 (P < 0.01). In a Kaplan-Meier analysis of overall survival rates, the patients with low expression of NCOA5 had significantly longer survival than high expression of NCOA5 (p=0.038). Conclusion NCOA5 high expression is associated with EOC progression and can be an independent factor affecting the prognosis of EOC patients.
Collapse
Affiliation(s)
- Xiaoping Song
- Department of Gynaecology, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
- *Correspondence: Xiaoping Song,
| | - Da Qian
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| | - Ping Dai
- Department of Burn and Plastic Surgery-Hand Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| | - Qian Li
- Department of Gynaecology, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| | - Qiuping Xi
- Department of Gynaecology, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| | - Kailv Sun
- Department of Thyroid and Breast Surgery, Changshu Hospital Affiliated to Soochow University, Changshu No.1 People’s Hospital, Changshu, Jiangsu, China
| |
Collapse
|
5
|
Tan Y, Liu F, Xu P. Knockdown of NCOA5 suppresses viability, migration and epithelial-mesenchymal transition, and induces adhesion of breast cancer cells. Oncol Lett 2021; 22:694. [PMID: 34457049 DOI: 10.3892/ol.2021.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/27/2021] [Indexed: 11/06/2022] Open
Abstract
Nuclear receptor coactivator 5 (NCOA5) has been reported to be involved in the regulation of several malignancies; however, to the best of our knowledge, its role in breast cancer is still unknown. The present study aimed to reveal the biological function of NCOA5 in breast cancer cells. NCOA5 expression in breast cancer tissues and cell lines was examined using reverse transcription-quantitative PCR and western blotting. Small interfering RNA (siRNA) against NCOA5 (siNCOA5) was transfected into MDA-MB-453 and MCF-7 cells to knock down NCOA5. MTT, transwell migration and cell adhesion assays were performed to determine cell viability, migration and adhesion abilities of breast cancer cells, respectively. In addition, the expression levels of N-cadherin, Vimentin and E-cadherin were examined by western blotting. It was revealed that NCOA5 expression was significantly increased in breast cancer tissues and cell lines. Knockdown of NCOA5 suppressed breast cancer cell viability and migration, and induced cell adhesion. Compared with those in cells transfected with non-targeting negative control siRNA, the protein expression levels of N-cadherin and Vimentin were significantly decreased, whereas the protein expression levels of E-cadherin were significantly increased in cells transfected with siNCOA5. The present study demonstrated that knockdown of NCOA5 suppressed cell viability and migration, induced cell adhesion, and inhibited epithelial-mesenchymal transition of breast cancer cells, indicating that NCOA5 may serve a tumor-promoting role in breast cancer.
Collapse
Affiliation(s)
- Yanfang Tan
- Department of Clinical Laboratory, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Fuhui Liu
- Department of Blood Transfusion, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563000, P.R. China
| | - Pei Xu
- Department of Pathology, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| |
Collapse
|
6
|
NCOA5 is a master regulator of amino acid-induced mTOR activation and β-casein synthesis in bovine mammary epithelial cells. Biochem Biophys Res Commun 2020; 529:569-574. [DOI: 10.1016/j.bbrc.2020.05.193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022]
|
7
|
Deng H, Zhou T, Mo X, Liu C, Yin Y. Low-density lipoprotein promotes lymphatic metastasis of esophageal squamous cell carcinoma and is an adverse prognostic factor. Oncol Lett 2018; 17:1053-1061. [PMID: 30655865 PMCID: PMC6313071 DOI: 10.3892/ol.2018.9683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/19/2018] [Indexed: 01/14/2023] Open
Abstract
The purpose of the current study was to investigate the prognostic role of preoperative serum lipid levels in patients with esophageal squamous cell carcinoma (ESCC) and to preliminarily explore the mechanism of serum lipids in this disease. Preoperative lipids, including total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol and triglyceride levels, were assessed in 242 patients with ESCC. To eliminate the influence of nutritional status, all patients had previously undergone esophagectomy. Univariate and multivariate Cox regression analyses were performed to identify predictors of overall survival (OS). Associations between significant lipid targets and clinical features were then analyzed and the results were validated using TE-1 and ECa109 esophageal cancer cell lines. The cell proliferation was evaluated with a Cell Counting Kit-8 (CCK8) assay and the cell cycle was assessed with propidium iodide staining and flow cytometry. Univariate analysis revealed that HDL (P=0.048), LDL (P=0.020), Pathological T-staging status (pT status) (P=0.001), Pathological N-staging status (pN status) (P=0.001) and histological differentiation (P=0.002) were significantly associated with OS. Based on multivariate analysis, LDL [hazard ratio (HR)=2.164, P=0.005], pT status (HR=1.714, P=0.001), pN status (HR=1.966, P=0.001) and histological differentiation (HR=4.083, P=0.002) were risk factors in patients with ESCC. A high LDL level (>3.12 mmol/l) was associated with sex (P=0.001), tumor location (P=0.004) and a higher susceptibility to lymphatic metastasis (P=0.007). A CCK8 assay demonstrated that LDL promoted TE-1 and ECa109 cell proliferation, and flow cytometry analysis revealed that treatment with LDL at an appropriate concentration resulted in an accumulation of cells in G2 phase and decreased the number of cells in G1 phase. In summary, the current study identified that preoperative LDL serum level serves an important role in predicting ESCC outcome as LDL promotes lymphatic metastasis. Furthermore, a preliminary mechanism for this association has been validated in vitro.
Collapse
Affiliation(s)
- Hongbin Deng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China.,School of Medicine and Life Science, University of Jinan-Shandong Academy of Medical Science, Jinan, Shandong 25022, P.R. China
| | - Tao Zhou
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Xinkai Mo
- Department of Laboratory, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Chengxin Liu
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
8
|
Liang Y, Zhang T, Shi M, Zhang S, Guo Y, Gao J, Yang X. Low expression of NCOA5 predicts poor prognosis in human cervical cancer and promotes proliferation, migration, and invasion of cervical cancer cell lines by regulating notch3 signaling pathway. J Cell Biochem 2018; 120:6237-6249. [PMID: 30335900 DOI: 10.1002/jcb.27911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/25/2018] [Indexed: 12/15/2022]
Abstract
Nuclear receptor coactivator 5 (NCOA5) specifically enhances estrogen receptor α-modulated transcriptional activity. As a novel tumor suppressor, depletion of NCOA5 is associated with the development of a variety of tumors, but its function in cervical cancer is currently unclear. In this study, we addressed how expression of NCOA5 changed in the development of human cervical cancer and its association with clinicopathological features, prognosis, and biology characteristics of cervical cancer. Analysis of the microarrays in the Oncomine database indicated that NCOA5 expression was lower in human cervical squamous cell carcinoma tissues than that in normal cervical tissues. That was corroborated by our experiments using fresh tissues: the expression levels of NCOA5 messenger RNA and protein were both significantly decreased in cervical cancer tissues compared with paired adjacent nontumor tissues (P < 0.01). Low expression of NCOA5 is associated with the International Federation of Gynecology and Obstetrics stage ( P = 0.043) and histological grade ( P = 0.018) of human cervical cancer. In addition, patients possessing low NCOA5 expression had poorer prognosis. Univariate and multivariate Cox regression analyses indicated that low NCOA5 expression may be an independent prognostic factor for poorer overall survival in cervical cancer. Further, downregulation of NCOA5 expression results in a significant increase in proliferation, migration, and invasion of HeLa cells. Data of xenograft tumor on BALB/c nude mice manifested that HeLa cells with low NCOA5 expression tend to form larger tumors than negative control ones. In contrast, overexpression of NCOA5 expression leads to the opposite results. Finally, we found that NCOA5 might affect the biological function of human cervical cancer cells by mediating the notch3 signaling pathway. These findings suggest that NCOA5 acts as a tumor suppressor to inhibit tumorigenicity, migration, and invasion, and thus represents a potential novel prognostic marker for overall survival in cervical cancer.
Collapse
Affiliation(s)
- Ying Liang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tianli Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Mengdie Shi
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital, Suzhou, China
| | - Shuo Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaxing Guo
- Department of Obstetrics and Gynecology, Shandong Obstetrics and Gynecology Hospital, Jinan, China
| | - Jiwei Gao
- Department of Oncology and Pathology, Karolinska Institute, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Xingsheng Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
9
|
Zheng ZC, Wang QX, Zhang W, Zhang XH, Huang DP. A novel tumor suppressor gene NCOA5 is correlated with progression in papillary thyroid carcinoma. Onco Targets Ther 2018; 11:307-311. [PMID: 29391807 PMCID: PMC5769572 DOI: 10.2147/ott.s154158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background In contrast to the excellent prognosis for papillary thyroid carcinoma (PTC), the high incidence of lymph node metastasis (LNM) markedly increases the risk of recurrence and secondary surgery. Thus, novel biomarkers must be urgently identified to assess LNM for patients with PTC. NCOA5 is deeply involved in the progression of human cancer; however, its role in thyroid cancer remains unknown. Patients and methods Quantitative real-time polymerase chain reaction was conducted to investigate the expression of NCOA5 in PTC. RNA-seq data from The Cancer Genome Atlas (TCGA) database were downloaded to further understand the role of NCOA5 in PTC and its relationship with LNM. Results NCOA5 was significantly downregulated in PTC tissues when compared with that in adjacent noncancerous thyroid tissues both in our local cohort and TCGA database. Reduced expression of NCOA5 was significantly associated with aggressive clinicopathological features, including histological type, tumor stage, BRAF-V600E mutation, LNM, extrathyroid extension, and clinical stage. Moreover, logistic analysis indicated that reduced expression of NCOA5, age, histological type, and clinical stage are independent high-risk factors for LNM in PTC. Conclusion Our study provides new insights and evidence that NOCA5 was significantly correlated with the progression of PTC and was particularly involved in LNM.
Collapse
Affiliation(s)
- Zhou-Ci Zheng
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Qing-Xuan Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Wei Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiao-Hua Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Du-Ping Huang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
10
|
Xing S, Zheng X, Zeng T, Zeng MS, Zhong Q, Cao YS, Pan KL, Wei C, Hou F, Liu WL. Chitinase 3-like 1 secreted by peritumoral macrophages in esophageal squamous cell carcinoma is a favorable prognostic factor for survival. World J Gastroenterol 2017; 23:7693-7704. [PMID: 29209110 PMCID: PMC5703929 DOI: 10.3748/wjg.v23.i43.7693] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/13/2017] [Accepted: 09/26/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To identify whether chitinase 3-like 1 (CHI3L1) serves as a suitable biomarker for the prognosis of esophageal squamous cell carcinoma (ESCC) and to analyze this protein’s cellular source.
METHODS An ELISA was conducted to detect the concentration of CHI3L1 in the serum of 150 ESCC patients diagnosed between January 2001 and February 2005. The prognostic relevance of CHI3L1 was evaluated by a Kaplan-Meier and Cox regression analysis. The immunohistochemistry was reanalyzed, and fluorescent staining was utilized to explore the cellular origins of CHI3L1. We stimulated monocyte-derived macrophages (MDMs) with either IL-6 or the supernatant of the ESCC cell line Eca-109 and later investigated the level of CHI3L1 by qPCR and ELISA.
RESULTS The level of serum CHI3L1 was higher in older patients (≥ 60) than in patients under the age of 60 (P = 0.001). The patients with higher levels of CHI3L1 had a significantly shorter overall survival, whereas the traditional markers, carcinoembryonic antigen and squamous cell carcinoma antigen, were less effective (P > 0.05). A multivariate Cox analysis (P = 0.001) indicated that CHI3L1 was an independent prognostic factor for ESCC patients. Peritumoral macrophages in ESCC exhibited high levels of CHI3L1. Interleukin-6 (IL-6) and the supernatant of Eca-109 containing IL-6 stimulated MDMs to secrete CHI3L1. The serum concentration of CHI3L1 in the ESCC patients showed a weak correlation with the laboratory inflammatory parameters neutrophil (NEU, P = 0.045), neutrophil/lymphocyte rate (NLR, P = 0.016), and C-reactive protein (CRP, P < 0.001).
CONCLUSION Our study first established a connection between the pretreated CHI3L1 and patients with ESCC, and the serum CHI3L1 was primarily secreted by ESCC-surrounded macrophages.
Collapse
Affiliation(s)
- Shan Xing
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Xin Zheng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Tao Zeng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Qian Zhong
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
| | - Yue-Song Cao
- Department of Biotechnology, Gannan Medical University, Ganzhou 341000, Jiangxi Province, China
| | - Kai-Lu Pan
- Department of Clinical Laboratory, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Chu Wei
- Department of Clinical Laboratory, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Fan Hou
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| | - Wan-Li Liu
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, Guangdong Province, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
11
|
Sun K, Wang S, He J, Xie Y, He Y, Wang Z, Qin L. NCOA5 promotes proliferation, migration and invasion of colorectal cancer cells via activation of PI3K/AKT pathway. Oncotarget 2017; 8:107932-107946. [PMID: 29296214 PMCID: PMC5746116 DOI: 10.18632/oncotarget.22429] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 07/25/2017] [Indexed: 02/06/2023] Open
Abstract
The nuclear receptor coactivator 5 (NCOA5) displays both coactivator and corepressor functions. Previous studies showed that alteration of NCOA5 participates in carcinogenesis and progression. However, its roles in colorectal cancer (CRC) remain unknown. Herein, we demonstrated that expression of NCOA5 in human CRC tissues was notably higher than that in adjacent tissues, which significantly correlated with clinicopathological features such as length of tumor, regional lymph node staging and cancer staging. Knockdown of NCOA5 markedly suppressed proliferation, migration and invasion of SW620 high malignant CRC cells. Silencing of NCOA5 also inhibited in vivo growth of SW620 CRC subcutaneously xenografted tumors in athymic BALB/c nude mice. Meanwhile, Overexpression of NCOA5 facilitated these processes in SW480 low malignant CRC cells. Furthermore, knockdown of NCOA5 induced cell cycle G1 phase arrest in SW620 cells, whereas overexpression of NCOA5 promoted G1 to S phase transition in SW480 cells. Mechanistic studies revealed that NCOA5 upregulated phospho-protein kinase B (p-PKB/AKT), Cyclin D1 and matrix metalloproteinase 9 (MMP9) as well as downregulated P27 in CRC cells. Notably, PI3K inhibitor LY294002 obviously attenuated the effects of NCOA5 on p-AKT, Cyclin D1, P27 and MMP9. Moreover, LY294002 and knockdown of Cyclin D1 or MMP9 remarkably blocked the tumor-promoting activity of NCOA5. Collectively, NCOA5 promoted CRC cell proliferation, migration and invasion by upregulating Cyclin D1 and MMP9 while downregulating P27 to a great extent via activating PI3K/AKT signaling pathway. These findings suggested that NCOA5 exhibits an oncogenic effect in human CRC and represents a novel therapeutic target for CRC.
Collapse
Affiliation(s)
- Kailv Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yufeng Xie
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang He
- Ministry of Health Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhenxin Wang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
12
|
Kollipara L, Buchkremer S, Coraspe JAG, Hathazi D, Senderek J, Weis J, Zahedi RP, Roos A. In-depth phenotyping of lymphoblastoid cells suggests selective cellular vulnerability in Marinesco-Sjögren syndrome. Oncotarget 2017; 8:68493-68516. [PMID: 28978133 PMCID: PMC5620273 DOI: 10.18632/oncotarget.19663] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/28/2017] [Indexed: 12/18/2022] Open
Abstract
SIL1 is a ubiquitous protein of the Endoplasmic Reticulum (ER) acting as a co-chaperone for the ER-resident chaperone, BiP. Recessive mutations of the corresponding gene lead to vulnerability of skeletal muscle and central nervous system in man (Marinesco-Sjögren syndrome; MSS) and mouse. However, it is still unclear how loss of ubiquitous SIL1 leads to selective vulnerability of the nervous system and skeletal muscle whereas other cells and organs are protected from clinical manifestations. In this study we aimed to disentangle proteins participating in selective vulnerability of SIL1-deficient cells and tissues: morphological examination of MSS patient-derived lymphoblastoid cells revealed altered organelle structures (ER, nucleus and mitochondria) thus showing subclinical vulnerability. To correlate structural perturbations with biochemical changes and to identify proteins potentially preventing phenotypical manifestation, proteomic studies have been carried out. Results of proteomic profiling are in line with the morphological findings and show affection of nuclear, mitochondrial and cytoskeletal proteins as well as of such responsible for cellular viability. Moreover, expression patterns of proteins known to be involved in neuromuscular disorders or in development and function of the nervous system were altered. Paradigmatic findings were confirmed by immunohistochemistry of splenic lymphocytes and the cerebellum of SIL1-deficient mice. Ataxin-10, identified with increased abundance in our proteome profile, is necessary for the neuronal survival but also controls muscle fiber apoptosis, thus declaring this protein as a plausible candidate for selective tissue vulnerability. Our combined results provide first insights into the molecular causes of selective cell and tissue vulnerability defining the MSS phenotype.
Collapse
Affiliation(s)
- Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Stephan Buchkremer
- Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany
| | | | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Jan Senderek
- Friedrich-Baur-Institute, Medical Faculty, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Joachim Weis
- Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany
| | - René P Zahedi
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften-ISAS -e.V., 44227 Dortmund, Germany.,Institute of Neuropathology, University Hospital Aachen, RWTH Aachen, 5274 Aachen, Germany.,The John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
13
|
Ye XH, Huang DP, Luo RC. NCOA5 is correlated with progression and prognosis in luminal breast cancer. Biochem Biophys Res Commun 2017; 482:253-256. [DOI: 10.1016/j.bbrc.2016.11.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/10/2016] [Indexed: 11/26/2022]
|
14
|
Wang XP, Li XH, Zhang L, Lin JH, Huang H, Kang T, Mao MJ, Chen H, Zheng X. High level of serum apolipoprotein A-I is a favorable prognostic factor for overall survival in esophageal squamous cell carcinoma. BMC Cancer 2016; 16:516. [PMID: 27444612 PMCID: PMC4957343 DOI: 10.1186/s12885-016-2502-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/04/2016] [Indexed: 12/28/2022] Open
Abstract
Background Noninvasive prognostic tools for esophageal squamous cell carcinoma (ESCC) are urgently needed. Serum lipids and lipoproteins are used for the prognosis of certain diseases; however, the prognostic value of serum apolipoprotein A-I (ApoA-I) in ESCC has not been described. Methods Pre-treatment serum lipids and lipoprotein concentrations (including ApoA-I, Apo-B, HDL-C, LDL-C, TC and TG) were analyzed retrospectively and compared between 210 patients with ESCC and 219 healthy controls. The prognostic significance of serum lipids and lipoproteins was determined by univariate and multivariate Cox hazard models in ESCC. Results Clinical characteristics (age, sex, pT status, pN status, pM status, pTNM status, histological differentiation or alcohol index) had no influence on baseline ApoA-I level. Serum ApoA-I, HDL-C, LDL-C, and TC levels were significantly lower and Apo-B was significantly higher in ESCC patients than in normal controls. On univariate analysis, ApoA-I, alcohol index, pT status, pN status and pTNM status were associated with significantly poor survival, and ApoA-I (p = 0.039), alcohol index (p = 0.037) and pTNM status (p = 0.000) were identified as prognostic factors associated with shorter survival in the multivariate analysis. Conclusions Overall survival was shorter in ESCC patients with decreased pre-treatment ApoA-I levels. Our findings suggest that serum ApoA-I level should be evaluated as a predictor of survival in patients with ESCC.
Collapse
Affiliation(s)
- Xue-Ping Wang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Xiao-Hui Li
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Lin Zhang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China.,Guangdong Esophageal Cancer Institute, Guangzhou, Guangdong, China
| | - Jian-Hua Lin
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Hao Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ting Kang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Min-Jie Mao
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China
| | - Hao Chen
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China.
| | - Xin Zheng
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, People's Republic of China.
| |
Collapse
|