1
|
Gong L, Huang J, Bai X, Song L, Hang J, Guo J. Expression of HECTD2 predicts peritoneal metastasis of gastric cancer and reconstructs immune microenvironment. Cancer Cell Int 2024; 24:380. [PMID: 39548546 DOI: 10.1186/s12935-024-03553-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
Peritoneal metastasis (PM) is a common metastasis site and death cause of gastric cancer, which is a complex biological process, but there is currently a lack of effective prediction and treatment targets. In this study, we first analyzed the differential gene expression of gastric cancer patients with or without peritoneal metastasis, and identified the HECT domain E3 ubiquitin protein ligase 2 (HECTD2) as the core gene of PM in gastric cancer. The current study shows that the role of HECTD2 in tumor is contradictory. In this study, our results show that the low expression of HECTD2 indicates that the survival rate of overall survival (OS), progression-free survival (PFS), disease-specific survival (DSS), and disease-free survival (DFS) are better, and can be used as an important component of prognostic indicators. In addition, through pathway enrichment analysis, we found that HECTD2 was mainly involved in metastasis related pathways such as extracellular matrix remodeling and cell adhesion in gastric cancer, and high expression of HECTD2 could activate epithelial-mesenchymal transition (EMT) metastasis related pathways in gastric cancer. In regulating the metastasis of gastric cancer cells, HECTD2 can also change the surrounding microenvironment, induce the enrichment of interstitial components and build an immune microenvironment conducive to tumor progression, while patients with low expression of HECTD2 may be more likely to benefit from immunotherapy. In conclusion, HECTD2 may be a novel biomarker for the diagnosis and prognosis of peritoneal metastasis of gastric cancer, providing basis for the mechanism of peritoneal metastasis of cancer and clinical medication.
Collapse
Affiliation(s)
- Libao Gong
- The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, Guangdong Province, China
| | - Jiayi Huang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xue Bai
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Lin Song
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Junjie Hang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
- Changzhou No.2 People's Hospital, Changzhou, China.
| | - Jinfeng Guo
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
| |
Collapse
|
2
|
Su J, Xia H, He H, Tang H, Zhou J, Xun Y, Liu F, Su B, Su Q. Diallyl disulfide antagonizes DJ-1 mediated proliferation, epithelial-mesenchymal transition, and chemoresistance in gastric cancer cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:4105-4119. [PMID: 38642008 DOI: 10.1002/tox.24300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/31/2024] [Indexed: 04/22/2024]
Abstract
Diallyl disulfide (DADS), an organic component of allicin abstracted from garlic, possesses multi-target antitumor activity. DJ-1 performs a vital function in promoting AKT aberrant activation via down-regulating phosphatase and tensin homologue (PTEN) in tumors. It is unknown the involvement of DJ-1 in epithelial-mesenchymal transition (EMT) of gastric cancer (GC) cells. The purpose of this study is to investigate whether diallyl disulfide (DADS) intervenes in the role of DJ-1 in GC. Based on the identification that the correlation between high DJ-1 and low PTEN expression in GC was implicated in clinical progression, we illuminated that down-regulation of DJ-1 by DADS aided in an increase in PTEN expression and a decrease in phosphorylated AKT levels, which was in line with the results manifested in the DJ-1 knockdown and overexpressed cells, concurrently inhibiting proliferation, EMT, migration, and invasion. Furthermore, the antagonistic effects of DADS on DJ-1 were observed in in vivo experiments. Additionally, DADS mitigated the DJ-1-associated drug resistance. The current study revealed that DJ-1 is one of potential targets for DADS, which hopefully provides a promising strategy for prevention and adjuvant chemotherapy of GC.
Collapse
Affiliation(s)
- Jian Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Hong Xia
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Hui He
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Huan Tang
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Department of Oncology, Yongzhou Central Hospital, Yongzhou, China
| | - Juan Zhou
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Yi Xun
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Fang Liu
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Bo Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Institute of Pharmacy and Pharmacology, School of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Qi Su
- Hunan Clinical Research Center for Gastric Cancer Prevention and Treatment, Second Affiliated hospital, University of South China, Hengyang, China
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
3
|
The PI3K/AKT signaling pathway in cancer: Molecular mechanisms and possible therapeutic interventions. Exp Mol Pathol 2022; 127:104787. [DOI: 10.1016/j.yexmp.2022.104787] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 04/15/2022] [Accepted: 05/21/2022] [Indexed: 01/02/2023]
|
4
|
Roles of Nrf2 in Gastric Cancer: Targeting for Therapeutic Strategies. Molecules 2021; 26:molecules26113157. [PMID: 34070502 PMCID: PMC8198360 DOI: 10.3390/molecules26113157] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/15/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Nuclear Factor Erythroid 2-Related Factor 2 (Nrf2) is a specific transcription factor with potent effects on the regulation of antioxidant gene expression that modulates cell hemostasis under various conditions in tissues. However, the effects of Nrf2 on gastric cancer (GC) are not fully elucidated and understood. Evidence suggests that uncontrolled Nrf2 expression and activation has been observed more frequently in malignant tumors, including GC cells, which is then associated with increased antioxidant capacity, chemoresistance, and poor clinical prognosis. Moreover, Nrf2 inhibitors and the associated modulation of tumor cell redox balance have shown that Nrf2 also has beneficial effects on the therapy of various cancers, including GC. Based on previous findings on the important role of Nrf2 in GC therapy, it is of great interest to scientists in basic and clinical tumor research that Nrf2 can be active as both an oncogene and a tumor suppressor depending on different background situations.
Collapse
|
5
|
Qiu L, Ma Z, Li X, Deng Y, Duan G, Zhao LE, Xu X, Xiao L, Liu H, Zhu Z, Chen H. DJ-1 is involved in the multidrug resistance of SGC7901 gastric cancer cells through PTEN/PI3K/Akt/Nrf2 pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1202-1214. [PMID: 33079995 DOI: 10.1093/abbs/gmaa110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/18/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
Gastric cancer is a common malignancy worldwide. The occurrence of multidrug resistance (MDR) is the major obstacle for effective gastric cancer chemotherapy. In this study, the in-depth molecular mechanism of the DJ-1-induced MDR in SGC7901 gastric cancer cells was investigated. The results showed that DJ-1 expression level was higher in MDR variant SGC7901/VCR cells than that in its parental SGC7901 cells. Moreover, DJ-1 overexpression conferred the MDR phenotype to SGC7901 cells, while DJ-1 knockdown in SGC7901/VCR cells induced re-sensitization to adriamycin, vincristine, cisplatin, and 5-fluorouracil. These results suggested that DJ-1 mediated the development of MDR in SGC7901 gastric cancer cells. Importantly, further data revealed that the activation of PI3k/Akt and Nrf2 signaling pathway were required for the DJ-1-induced MDR phenotype in SGC7901 gastric cancer cells. Meanwhile, we found that PI3k/Akt pathway was activated probably through DJ-1 directly binding to and negatively regulating PTEN, consequently resulting in Nrf2 phosphorylation and activation, and thereby inducing Nrf2-dependent P-glycoprotein (P-gp) and Bcl-2 expressions in the DJ-1-mediated MDR of SGC7901 gastric cancer cells. Overall, these results revealed that activating PTEN/PI3K/Akt/Nrf2 pathway and subsequently upregulating P-gp and Bcl-2 expression could be a critical mechanism by which DJ-1 mediates the development of MDR in SGC7901 gastric cancer cells. The new findings may be helpful for understanding the mechanisms of MDR in gastric cancer cells, prompting its further investigation as a molecular target to overcome MDR.
Collapse
Affiliation(s)
- Lejia Qiu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhaoxia Ma
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Xiaoran Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Yizhang Deng
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Guangling Duan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - L e Zhao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Xingwang Xu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Lin Xiao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Haoyue Liu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Heping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| |
Collapse
|
6
|
Liu S, Gong Y, Xu XD, Shen H, Gao S, Bao HD, Guo SB, Yu XF, Gong J. MicroRNA-936/ERBB4/Akt axis exhibits anticancer properties of gastric cancer through inhibition of cell proliferation, migration, and invasion. Kaohsiung J Med Sci 2020; 37:111-120. [PMID: 33021020 DOI: 10.1002/kjm2.12304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/01/2020] [Accepted: 09/06/2020] [Indexed: 12/28/2022] Open
Abstract
Gastric cancer is one of the most common cancers globally and has a poor prognosis. MiR-936 has been reported to regulate cell activity and tumor progression in non-small cell lung cancer, glioma, and epithelial ovarian cancer. However, the specific role and mechanism of miR-936 in gastric cancer have not been explored. In present study, gastric cancer cells were transfected with miR-936 mimic, and cell proliferation, cell cycle distribution, cell apoptosis, migration and invasion were assessed via cell-counting kit-8, flow cytometry, wound healing, and transwell assay, respectively. Dual luciferase reporter assay was used to check miR-936 binding to its downstream target. It was shown that miR-936 was downregulated in gastric cancer tissues and cells. Erb-B2 Receptor Tyrosine Kinase 4 (ERBB4) was confirmed as a direct target of miR-936 and negatively regulated its expression by miR-936. Overexpression of miR-936 suppressed cell proliferation, cell cycle progression, cell migration and invasion, and enhanced cell apoptosis in gastric cancer cells, which could be reversed by further ERBB4 overexpression. Western blot results showed that miR-936/ERBB4 axis regulated Akt-related pathways to control gastric cancer cell activities. Therefore, our data suggest that miR-936 overexpression inhibits cell proliferation and invasion and promotes cell apoptosis through Akt-related pathways by targeting ERBB4, which provides novel insight to target miR-936 or miR-936/ERBB4 axis for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Shi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Gong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xue-Dong Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui Shen
- Department of Chinese Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuai Gao
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hai-Dong Bao
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shi-Bin Guo
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xue-Feng Yu
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jian Gong
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Jin W. Novel Insights into PARK7 (DJ-1), a Potential Anti-Cancer Therapeutic Target, and Implications for Cancer Progression. J Clin Med 2020; 9:jcm9051256. [PMID: 32357493 PMCID: PMC7288009 DOI: 10.3390/jcm9051256] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022] Open
Abstract
The expression of PARK7 is upregulated in various types of cancer, suggesting its potential role as a critical regulator of the pathogenesis of cancer and in the treatment of cancer and neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, and Huntington disease. PARK7 activates various intracellular signaling pathways that have been implicated in the induction of tumor progression, which subsequently enhances tumor initiation, continued proliferation, metastasis, recurrence, and resistance to chemotherapy. Additionally, secreted PARK7 has been identified as a high-risk factor for the pathogenesis and survival of various cancers. This review summarizes the current understanding of the correlation between the expression of PARK7 and tumor progression.
Collapse
Affiliation(s)
- Wook Jin
- Laboratory of Molecular Disease and Cell Regulation, Department of Biochemistry, School of Medicine, Gachon University, Incheon 406-840, Korea
| |
Collapse
|
8
|
Zhu X, Luo C, Lin K, Bu F, Ye F, Huang C, Luo H, Huang J, Zhu Z. Overexpression of DJ-1 enhances colorectal cancer cell proliferation through the cyclin-D1/MDM2-p53 signaling pathway. Biosci Trends 2020; 14:83-95. [PMID: 32132307 DOI: 10.5582/bst.2019.01272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Emerging evidence indicates that DJ-1 is highly expressed in different cancers. It modulates cancer progression, including cell proliferation, cell apoptosis, invasion, and metastasis. However, its role in colorectal cancer (CRC) remains poorly defined. The current study noted increased DJ-1 expression in CRC tumor tissue and found that its expression was closely related to clinical-pathological features. Similarly, DJ-1 increased in CRC cells (SW480, HT-29, Caco-2, LoVo, HCT116, and SW620), and especially in SW480 and HCT116 cells. Functional analyses indicated that overexpression of DJ-1 promoted CRC cell invasion, migration, and proliferation in vitro and in vivo. Mechanistic studies indicated that DJ-1 increased in CRC cell lines, activated specific protein cyclin-D1, and modulated the MDM2/p53 signaling pathway by regulating the levels of the downstream factors Bax, Caspase-3, and Bcl-2, which are related to the cell cycle and apoptosis. Conversely, knockdown of DJ-1 upregulated p53 expression by disrupting the interaction between p53 and MDM2 and inhibiting CRC cell proliferation, revealing the pro-oncogenic mechanism of DJ-1 in CRC. In conclusion, the current findings provide compelling evidence that DJ-1 might be a promoter of CRC cell invasion, proliferation, and migration via the cyclin-D1/MDM2-p53 signaling pathway. Findings also suggest its potential role as a postoperative adjuvant therapy for patients with CRC.
Collapse
Affiliation(s)
- Xiaojian Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chen Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Kang Lin
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fanqin Bu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Fan Ye
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Huang
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Zhengming Zhu
- The Second Affiliated Hospital Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
DJ-1 overexpression confers the multidrug resistance phenotype to SGC7901 cells by upregulating P-gp and Bcl-2. Biochem Biophys Res Commun 2019; 519:73-80. [PMID: 31477270 DOI: 10.1016/j.bbrc.2019.08.131] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 01/01/2023]
Abstract
Gastric cancer (GC) is one of the most malignant tumors with high incidence and mortality worldwide, and the multidrug resistance (MDR) often results in chemotherapy failure in GC. DJ-1 has been well indicated to be associated with drug resistance in multiple cancers. However, the role of DJ-1 in the MDR of gastric cancer cells and its possible mechanism remain to be elucidated. Therefore, the current study was investigated whether DJ-1 expression is differential in parental gastric cancer cell SGC7901 and vincristine (VCR)-induced gastric cancer MDR cell SGC7901/VCR, and whether DJ-1 plays a significant role in development of MDR in gastric cancer. The results showed that DJ-1 expression in SGC7901/VCR cells was significantly higher than its sensitive parental SGC7901 cells. Furthermore, DJ-1 overexpressed gastric cancer cell line SGC7901/LV-DJ-1 led to the increase of cell survival rate, the IC50 of chemotherapeutic drugs and number of cell clones as well as decrease of cell cycle G0/G1 phase ratio compared with its parental cells under the treatment of VCR, adriamycin (ADR), 5-Fluorouracil (5-FU) and cisplatin (DDP). However, the DJ-1 knockdown stable cell line SGC7901/VCR/shDJ-1 reversed the above mentioned series of MDR. Moreover, it was found that upregulation of DJ-1 protein expression promoted the pumping rate of GC cells to ADR and reduced the apoptotic index of GC cells treated with chemotherapeutic drugs by upregulating P-gp and Bcl-2. Similarly, knocking down DJ-1, P-gp or Bcl-2 displayed a converse effect. In conclusion, the current study demonstrated that DJ-1 overexpression confers the MDR phenotype to SGC7901 cells and this process is related to DJ-1 promoting active efflux of drugs and enhancing the anti-apoptotic ability of MDR GC cells by upregulating P-gp and Bcl-2.
Collapse
|
10
|
Liu R, Yang YN, Yi L, Qing J, Li QY, Wang WS, Wang J, Tang YX, Tan H. Diallyl disulfide effect on the invasion and migration ability of HL-60 cells with a high expression of DJ-1 in the nucleus through the suppression of the Src signaling pathway. Oncol Lett 2018; 15:6377-6385. [PMID: 29725397 PMCID: PMC5920463 DOI: 10.3892/ol.2018.8139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
The present study examined the effect of diallyl disulfide (DADS) on the invasion and migration ability of HL-60 cells with a high expression of parkinsonism associated deglycase (DJ-1) in the nucleus (HHDN), and its molecular mechanism. A western blot assay was used to measure the effects of DADS and an Src inhibitor on the expression of DJ-1 and the Src signal pathway in HHDN. The effects of DADS and Src inhibitors on the invasion and migration ability of HHDN was detected using Transwell migration and invasion chamber experiments. The experiments were divided into three groups: A control group (HL-60 cells), an empty vector group and a high expression group (HHDN cells). Western blot assays revealed that the expression of DJ-1 in HHDN was inhibited in a time-dependent manner following treatment with DADS for 24, 48 and 72 h. Following DADS treatment, the expression of phosphorylated Src (p-Src) and phosphorylated Fak (p-Fak) were significantly decreased in all groups compared with the untreated groups, however the expression level of Src, Fak and integrin did not change significantly. Western blot analysis results revealed that following treatment with DADS and Src inhibitor, the expression levels of p-Src and p-Fak significantly decreased in all three groups compared with untreated groups, whereas the expression levels of Src, Fak and integrin did not change significantly. The expression of DJ-1 in HHND was inhibited in time-dependent manner following treatment with DADS and Src inhibitor for 24, 48 and 72 h. Transwell migration and invasion assay results revealed that DADS and Src inhibitors may suppress migration and invasion in leukemic cells, and a combination of the two treatments may result in more efficient suppression. DADS may downregulate DJ-1-mediated invasion and migration in leukemic cells through suppressing the Src-Fak-Integrin signaling pathway, and the Src inhibitor may enhance the antitumor effect of DADS.
Collapse
Affiliation(s)
- Ran Liu
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China.,Department of Pathology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ye-Ning Yang
- Department of Pathology, The First People's Hospital of Youxian, Youxian, Hunan 412300, P.R. China
| | - Lan Yi
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Jing Qing
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Qing-Ye Li
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Wen-Song Wang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Juan Wang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Yu-Xian Tang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Hui Tan
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| |
Collapse
|
11
|
Vavougios G, Zarogiannis SG, Doskas T. The putative interplay between DJ-1/NRF2 and Dimethyl Fumarate: A potentially important pharmacological target. Mult Scler Relat Disord 2018. [PMID: 29529529 DOI: 10.1016/j.msard.2018.02.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent research has outlined that Dimethyl Fumarate (DMF) functions as a gene regulator via multiple pathways, critical among which is the NRF2 cytoprotective cascade. PARK7/DJ-1 is a multifunctional protein that acts as a redox sensor and effector of multiple cytoprotective pathways, including NRF2. Specifically, it prevents the association of NRF2 with its inhibitor KEAP1, allowing NRF2 to enter the nucleus and mediate cytoprotective and antioxidant cascades. It is our hypothesis that while the NRF2-KEAP1 inhibitory complex is reported the main pharmacological target for DMF's NRF dependent functions, no study to date has explored the effects of DMF on DJ-1's expression, and vice-versa, the possibility of a regulatory inadequacy in the upstream, oxidant-responsive DJ-1 activator of the NRF2 cascade.
Collapse
Affiliation(s)
- George Vavougios
- Department of Neurology, Athens Naval Hospital, Deinokratous 70, Athens, Greece.
| | - Sotirios G Zarogiannis
- Department of Physiology, Faculty of Medicine, University of Thessaly, BIOPOLIS, Larissa 41500, Greece
| | - Triantafylos Doskas
- Department of Neurology, Athens Naval Hospital, Deinokratous 70, Athens, Greece
| |
Collapse
|
12
|
Kwon HS, Park JH, Hwang HS, Sohn IS, Kim YH, Cho S. Effect of DJ-1 Downregulation on the Functions of the First Trimester Extravillous Trophoblasts. Reprod Sci 2017; 25:1436-1445. [PMID: 29258409 DOI: 10.1177/1933719117746760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
DJ-1 ( PARK7) has been reported to be causative gene of Parkinson disease and also an oncogene. A loss in DJ-1 function can lead to cell death in neurodegenerative disease, or a gain of it can cause unregulated cell survival in cancer, respectively. DJ-1 protein is known to be expressed mainly in trophoblastic cells in the placenta with increased expression in the first trimester compared to later in term. However, its role in trophoblast regulation remains unknown. This study aimed to investigate the effect of DJ-1 regulation on a first trimester extravillous trophoblast cell line, HTR-8/SVneo. The effect of DJ-1 downregulation induced by small-interfering RNA on cell apoptosis, migration, and the pathway to regulate the cell function was assessed. Data of this study showed that DJ-1 downregulation increased apoptosis and reduced migration by regulating matrix metalloproteinase 2 and matrix metalloproteinase 9 in HTR-8/SVneo cells under both ambient and oxidative stress. Changes in cell function were demonstrated to be at least partly dependent on the AKT/S6 kinase beta-1 (S6K1) pathway. In summary, DJ-1 might play a protective role in maintaining trophoblastic cell functions through the AKT/S6K1-based pathway.
Collapse
Affiliation(s)
- Han-Sung Kwon
- 1 Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Korea.,2 Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Park
- 3 Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.,4 Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Han-Sung Hwang
- 1 Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Korea
| | - In-Sook Sohn
- 1 Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Korea
| | - Young-Han Kim
- 5 Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Korea
| | - SiHyun Cho
- 3 Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.,4 Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Zeng R, Li B, Huang J, Zhong M, Li L, Duan C, Zeng S, Huang J, Liu W, Lu J, Tang Y, Zhou L, Liu Y, Li J, He Z, Wang Q, Dai Y. Lysophosphatidic Acid is a Biomarker for Peritoneal Carcinomatosis of Gastric Cancer and Correlates with Poor Prognosis. Genet Test Mol Biomarkers 2017; 21:641-648. [PMID: 28910191 DOI: 10.1089/gtmb.2017.0060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Ruolan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Junhui Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Meizuo Zhong
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Li Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Chaojun Duan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingchen Lu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youhong Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Lingming Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yiping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhuang Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengxi He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youyi Dai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Oh SE, Mouradian MM. Regulation of Signal Transduction by DJ-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1037:97-131. [PMID: 29147906 DOI: 10.1007/978-981-10-6583-5_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The ability of DJ-1 to modulate signal transduction has significant effects on how the cell regulates normal processes such as growth, senescence, apoptosis, and autophagy to adapt to changing environmental stimuli and stresses. Perturbations of DJ-1 levels or function can disrupt the equilibrium of homeostatic signaling networks and set off cascades that play a role in the pathogenesis of conditions such as cancer and Parkinson's disease.DJ-1 plays a major role in various pathways. It mediates cell survival and proliferation by activating the extracellular signal-regulated kinase (ERK1/2) pathway and the phosphatidylinositol-3-kinase (PI3K)/Akt pathway. It attenuates cell death signaling by inhibiting apoptosis signal-regulating kinase 1 (ASK1) activation as well as by inhibiting mitogen-activated protein kinase kinase kinase 1 (MEKK1/MAP3K1) activation of downstream apoptotic cascades. It also modulates autophagy through the ERK, Akt, or the JNK/Beclin1 pathways. In addition, DJ-1 regulates the transcription of genes essential for male reproductive function, such as spermatogenesis, by relaying nuclear receptor androgen receptor (AR) signaling. In this chapter, we summarize the ways that DJ-1 regulates these pathways, focusing on how its role in signal transduction contributes to cellular homeostasis and the pathologic states that result from dysregulation.
Collapse
Affiliation(s)
- Stephanie E Oh
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - M Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
15
|
Unique proteome signature of post-chemotherapy ovarian cancer ascites-derived tumor cells. Sci Rep 2016; 6:30061. [PMID: 27470985 PMCID: PMC4965858 DOI: 10.1038/srep30061] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022] Open
Abstract
Eighty % of ovarian cancer patients diagnosed at an advanced-stage have complete remission after initial surgery and chemotherapy. However, most patients die within <5 years due to episodes of recurrences resulting from the growth of residual chemoresistant cells. In an effort to identify mechanisms associated with chemoresistance and recurrence, we compared the expression of proteins in ascites-derived tumor cells isolated from advanced-stage ovarian cancer patients obtained at diagnosis (chemonaive, CN) and after chemotherapy treatments (chemoresistant/at recurrence, CR) by using in-depth, high-resolution label-free quantitative proteomic profiling. A total of 2,999 proteins were identified. Using a stringent selection criterion to define only significantly differentially expressed proteins, we report identification of 353 proteins. There were significant differences in proteins encoding for immune surveillance, DNA repair mechanisms, cytoskeleton rearrangement, cell-cell adhesion, cell cycle pathways, cellular transport, and proteins involved with glycine/proline/arginine synthesis in tumor cells isolated from CR relative to CN patients. Pathway analyses revealed enrichment of metabolic pathways, DNA repair mechanisms and energy metabolism pathways in CR tumor cells. In conclusion, this is the first proteomics study to comprehensively analyze ascites-derived tumor cells from CN and CR ovarian cancer patients.
Collapse
|
16
|
DJ-1 Mediates the Delayed Cardioprotection of Hypoxic Preconditioning Through Activation of Nrf2 and Subsequent Upregulation of Antioxidative Enzymes. J Cardiovasc Pharmacol 2016; 66:148-58. [PMID: 25915512 DOI: 10.1097/fjc.0000000000000257] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
We have recently shown that DJ-1 is implicated in the delayed cardioprotective effect of hypoxic preconditioning (HPC) against hypoxia/reoxygenation (H/R) injury as an endogenous protective protein. This study aims to further investigate the underlying mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress. Using a well-characterized cellular model of HPC from rat heart-derived H9c2 cells, we found that HPC promoted nuclear factor erythroid 2-related factor 2 (Nrf2) and its cytoplasmic inhibitor Kelch-like ECH-associated protein-1 (Keap1) dissociation and resulted in increased nuclear translocation, antioxidant response element-binding, and transcriptional activity of Nrf2 24 hours after HPC, with subsequent upregulation of manganese superoxide dismutase (MnSOD) and heme oxygenase-1 (HO-1), which provided delayed protection against H/R-induced oxidative stress in normal H9c2 cells. However, the aforementioned effects of HPC were abolished in DJ-1-knockdown H9c2 cells, which were restored by restoration of DJ-1 expression. Importantly, we showed that inhibition of the Nrf2 pathway in H9c2 cells mimicked the effects of DJ-1 knockdown and abolished HPC-derived induction of antioxidative enzymes (MnSOD and HO-1) and the delayed cardioprotection. In addition, inhibition of Nrf2 also reversed the effects of restored DJ-1 expression on induction of antioxidative enzymes and delayed cardioprotection by HPC in DJ-1-knockdown H9c2 cells. Taken together, this work revealed that activation of Nrf2 pathway and subsequent upregulation of antioxidative enzymes could be a critical mechanism by which DJ-1 mediates the delayed cardioprotection of HPC against H/R-induced oxidative stress in H9c2 cells.
Collapse
|
17
|
Fernandez-Caggiano M, Schröder E, Cho HJ, Burgoyne J, Barallobre-Barreiro J, Mayr M, Eaton P. Oxidant-induced Interprotein Disulfide Formation in Cardiac Protein DJ-1 Occurs via an Interaction with Peroxiredoxin 2. J Biol Chem 2016; 291:10399-410. [PMID: 26945066 DOI: 10.1074/jbc.m115.699850] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 12/24/2022] Open
Abstract
The role and responses of the dimeric DJ-1 protein to cardiac oxidative stress is incompletely understood. H2O2 induces a 50-kDa DJ-1 interprotein homodimer disulfide, known to form between Cys-53 on each subunit. A trimeric 75-kDa DJ-1 complex that mass spectrometry shows contained 2-Cys peroxiredoxin also formed and precedes the appearance of the disulfide dimer. These observations may represent peroxiredoxin sensing and transducing the oxidant signal to DJ-1. The dimeric disulfide DJ-1 complex was stabilized by auranofin, suggesting that thioredoxin recycles it in cells. Higher concentrations of H2O2 concomitantly induce DJ-1 Cys-106 hyperoxidation (sulfination or sulfonation) in myocytes, perfused heart, or HEK cells. An oxidation-resistant C53A DJ-1 shows potentiated H2O2-induced Cys-106 hyperoxidation. DJ-1 also forms multiple disulfides with unknown target proteins during H2O2 treatment, the formation of which is also potentiated in cells expressing the C53A mutant. This suggests that the intersubunit disulfide induces a conformational change that limits Cys-106 forming heterodisulfide protein complexes or from hyperoxidizing. High concentrations of H2O2 also induce cell death, with DJ-1 Cys-106 sulfonation appearing causal in these events, as expressionof C53A DJ-1 enhanced both Cys-106 sulfonation and cell death. Nonetheless, expression of the DJ-1 C106A mutant, which fully prevents hyperoxidation, also showed exacerbated cell death responses to H2O2 A rational explanation for these findings is that DJ-1 Cys-106 forms disulfides with target proteins to limit oxidant-induced cell death. However, when Cys-106 is hyperoxidized, formation of these potentially protective heterodimeric disulfide complexes is limited, and so cell death is exacerbated.
Collapse
Affiliation(s)
- Mariana Fernandez-Caggiano
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Ewald Schröder
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Hyun-Ju Cho
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Joseph Burgoyne
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Javier Barallobre-Barreiro
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, King's College Hospital, London SE5 9NU, United Kingdom
| | - Manuel Mayr
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, King's College Hospital, London SE5 9NU, United Kingdom
| | - Philip Eaton
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| |
Collapse
|
18
|
Tanti GK, Pandey S, Goswami SK. SG2NA enhances cancer cell survival by stabilizing DJ-1 and thus activating Akt. Biochem Biophys Res Commun 2015; 463:524-31. [DOI: 10.1016/j.bbrc.2015.05.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/18/2015] [Indexed: 01/20/2023]
|
19
|
Pylväs-Eerola M, Karihtala P, Puistola U. Preoperative serum 8-hydroxydeoxyguanosine is associated with chemoresistance and is a powerful prognostic factor in endometrioid-type epithelial ovarian cancer. BMC Cancer 2015; 15:493. [PMID: 26134400 PMCID: PMC4489129 DOI: 10.1186/s12885-015-1504-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/19/2015] [Indexed: 01/30/2023] Open
Abstract
Background Oxidative stress is a widely seen phenomenon in several carcinomas. Increasing evidence also suggests that it has a significant role in the development of epithelial ovarian carcinoma (EOC). 8-Hydroxydeoxyguanosine (8-OHdG) is one of the main indicators of oxidative stress and increased expression of 8-OHdG has previously been seen in EOC. DJ-1 is an oncoprotein connected to oxidative stress regulation, but its role in ovarian cancer is not well known. We investigated redox status in different histotypes of EOC by measuring serum 8-OHdG and DJ-1 concentrations and their associations with known prognostic factors. Methods Serum samples from newly diagnosed EOC patients were collected in 1996–2009 and stored at the Department of Obstetrics and Gynecology, Oulu University Hospital. Serum 8-OHdG and DJ-1 levels were measured by using commercially available ELISA kits. Clinical data was gathered retrospectively from the patients` files. Results were analyzed by using SPSS software. Results In total, 112 patient samples were analyzed (38 serous, 20 mucinous, 34 endometrioid and 20 clear-cell). High serum 8-OHdG levels were associated with poor overall survival (OS) (p = 0.019), poor disease-free survival (DFS) (p = 0.020), platinum resistance (p = 0.002), serous histology versus other (p = 0.033), stage III–IV versus I–II (p = 0.009) and suboptimal surgical outcome (p = 0.012). Regarding histotypes, in the endometrioid EOC group in particular, serum 8-OHdG levels were significantly associated with poor DFS (p = 0.005), suboptimal surgical outcome (p = 0.025), and platinum resistance (p = 0.007). The prognostic significance of 8-OHdG in patients with endometrioid cancer in terms of DFS was confirmed in Cox regression analysis. High DJ-1 levels were associated with high histological grade (p = 0.029) and nonsignificantly associated with serous histology vs. other histology (p = 0.089). Conclusions An elevated serum 8-OHdG level is a significant predictor of poor prognosis, especially in cases of the endometrioid subtype of ovarian carcinoma. High 8-OHdG levels are associated with all traditional factors of poor prognosis in ovarian cancer and they also predict earlier development of platinum resistance. These results could be valuable when deciding the primary treatment mode for EOC patients.
Collapse
Affiliation(s)
- Marjo Pylväs-Eerola
- Department of Obstetrics and Gynecology, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 22, FIN-90029, Oulu, Finland.
| | - Ulla Puistola
- Department of Obstetrics and Gynecology, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.
| |
Collapse
|
20
|
Vavougios G, Kerenidi T, Tsilioni I, Zarogiannis SG, Gourgoulianis KI. Pleural effusion levels of DJ-1 are increased in elderly lung cancer patients with malignant pleural effusions. Redox Rep 2015; 20:254-8. [PMID: 26125099 DOI: 10.1179/1351000215y.0000000023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVES DJ-1 is a multifunctional protein implicated in redox dependent cell fate decisions. The aim of our study was to determine the pleural fluid (PF) levels of DJ-1 in malignant pleural effusions (MPEs) secondary to lung cancer. Additionally, we opted to assess potential correlations of DJ-1 PF levels with the PF levels of superoxide dismutase-1 (SOD1) and 8-isoprostane that are known antioxidant enzymes and have been previously reported in MPEs. METHODS Forty lung cancer patients with cytological proof of MPE were enrolled in this study. The PF levels of DJ-1, SOD1, and 8-isoprostane were measured by means of enzyme-linked immunosorbent assay. RESULTS The median PF levels of DJ-1 were 826 ng/mL (interquartile range, IQR: 482-1010 ng/mL). DJ-1 PF levels significantly correlated with PF Cu/Zn-SOD1 and PF 8-isoprostane levels (Spearman's rho, r; r = -0.476, P = 0.002 and r = -0.264, P = 0.033, respectively), PF lactate dehydrogenase (r = -0.497, P = 0.001) and total PF cell counts (r = -0.325, P = 0.041). Finally, in patients aged over 65 the PF DJ-1 levels were significantly higher than patients aged less than 65 (875 ng/mL vs. 607 ng/mL, respectively, P = 0.037). DISCUSSION To our knowledge, this is the first report to determine DJ-1's levels in MPEs due to lung cancer. The negative correlations between DJ-1, SOD1, and 8-isorpostane warrant further investigation regarding the altered redox regulation associated with MPEs.
Collapse
|
21
|
DJ-1: a promising marker in metastatic uveal melanoma. J Cancer Res Clin Oncol 2014; 141:315-21. [PMID: 25129821 DOI: 10.1007/s00432-014-1804-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 08/08/2014] [Indexed: 01/30/2023]
Abstract
PURPOSE Overexpression of DJ-1 was associated with metastatic uveal melanoma (UM). The purpose of this study was to evaluate the potential of serum DJ-1 as a biomarker for metastasis of uveal melanoma. METHODS Serum DJ-1 levels were determined by ELISA assays in 27 patients with metastatic UM metastatic uveal melanoma and in 76 patients who were disease free for at least 10 years and 30 age- and sex-matched controls. Receiver operating characteristic (ROC) curve was used to evaluate the feasibility of DJ-1 in detection of metastatic uveal melanoma. RESULTS Serum DJ-1 levels were significantly higher in patients with metastatic UM compared with patients who were disease free for at least 10 years (P < 0.001) or with controls (P < 0.001). ROC curve for DJ-1 revealed an area under the curve of 86.3%, and when 3.350 ng/mL was used as the cutoff value, a sensitivity of 74.1% and a specificity of 94.3% were achieved. Comparison of DJ-1 and liver function tests (LFTs) ROC curves indicated that DJ-1 was superior to LFTs in detection of metastatic UM. CONCLUSIONS Our data suggest that DJ-1 might be a promising serum marker for monitoring metastatic uveal melanoma.
Collapse
|