1
|
Lou R, Shao H, Wu P. Expression of RSK4 protein in non-small cell lung cancer tissues, adjacent tissues and its correlation with clinicopathological features. Am J Transl Res 2024; 16:3273-3279. [PMID: 39114716 PMCID: PMC11301506 DOI: 10.62347/zleq9498] [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: 03/29/2024] [Accepted: 06/16/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE To evaluate the expression of Ribosomal S6 kinase 4 (RSK4) protein in non-small cell lung cancer (NSCLC) tissues and adjacent non-tumor tissues, and to elucidate its correlation with clinicopathological features of NSCLC. METHODS We analyzed 100 NSCLC patients treated at the Second Affiliated Hospital of Zhejiang University School of Medicine from June 2020 to June 2022. Patient demographics and clinical data, including gender, age, history of diabetes, tumor location, degree of tumor differentiation, lymph node metastasis, and clinical stage, were collected. RSK4 protein expression was assessed in tissue samples via immunohistochemical staining. RESULTS RSK4 protein was positively expressed in 35.00% of cancerous tissues, significantly lower than the 69.00% observed in adjacent non-tumor tissues (P < 0.05). Patients with lower tumor differentiation, advanced Tumor Node Metastasis (TNM) stages, and lymph node metastases showed significantly reduced RSK4 expression compared to those with higher differentiation, earlier TNM stages, and no lymph node metastases (all P < 0.05). Cox regression analysis indicated that TNM stage, low differentiation, and lymph node metastases significantly influenced RSK4 expression (all P < 0.05). Survival analysis revealed a higher positive prognosis survival rate of 74.29% (26/35) among patients with positive RSK4 expression, versus 53.85% (35/65) in those with negative expression (P < 0.05). Spearman correlation analysis demonstrated a significant positive correlation of RSK4 expression with TNM stage, lymph node metastasis, and patient prognosis (all P < 0.05). CONCLUSION Positive RSK4 expression in NSCLC tissues is significantly correlated with advanced cancer stage, poor differentiation, and presence of lymph node metastasis, suggesting a potential tumor suppressor role for RSK4 in NSCLC. This association underscores its prognostic relevance in NSCLC patients.
Collapse
Affiliation(s)
- Ru Lou
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou 310000, Zhejiang, China
| | - Huawei Shao
- Department of Burns and Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou 310000, Zhejiang, China
| | - Pengcheng Wu
- Department of Burns and Wound Care Centre, The Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou 310000, Zhejiang, China
| |
Collapse
|
2
|
Wang H, Shan X, Peng Y, Zhou W. Circular RNAs in the chemoresistance of triple-negative breast cancer: A systematic review. Drug Dev Res 2023; 84:805-814. [PMID: 37114737 DOI: 10.1002/ddr.22069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023]
Abstract
This study aims to assess studies on circular RNAs (circRNAs) in the chemoresistance of triple-negative breast cancer (TNBC) and provide relevant references for the development of new TNBC chemotherapy sensitivity biomarkers and therapeutic targets. The PubMed, Embase, Web of Knowledge, Cochrane Library, and four Chinese databases were searched up to January 27, 2023, and studies related to TNBC chemoresistance were included. The basic characteristics of the studies and the mechanisms of circRNAs in regulating TNBC chemoresistance were analyzed. A total of 28 studies published between 2018 and 2023 were included, and the chemotherapeutics included adriamycin, paclitaxel, docetaxel, 5-fluorouracil, lapatinib, and so forth. A total of 30 circRNAs were identified, 86.67% (n = 26) of these circRNAs were reported to act as microRNA (miRNA) sponges to regulate chemotherapy sensitivity, while only two circRNAs (circRNA-MTO1 and circRNA-CREIT) interacted with proteins. A total of 14, 12, and 2 circRNAs were reported to be associated with chemoresistance to adriamycin, taxanes, and 5-fluorouracil, respectively. Six circRNAs were found to act as miRNA sponges that promote chemotherapy resistance by regulating the PI3K/Akt signalling pathway. CircRNAs participate in the regulation of TNBC chemoresistance and can be used as biomarkers and therapeutic targets for improving chemotherapy sensitivity. However, further studies are needed to confirm the role of circRNAs in TNBC chemoresistance.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuefeng Shan
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Arechavaleta-Velasco F, Dominguez-Lopez P, Acosta-Jimenez E, Diaz-Cueto L. Ribosomal S6 kinase 4 (RSK4) tumor suppressor gene promoter methylation status in ovarian cancer. Mol Biol Rep 2023; 50:6863-6870. [PMID: 37402066 DOI: 10.1007/s11033-023-08609-9] [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: 05/05/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND Previously, we reported lower RSK4 mRNA and protein levels in malignant ovarian tumors compared to normal and benign ovarian tissues. Also, we observed a significant inverse correlation between the advanced ovarian cancer stages and RSK4 mRNA levels. We did not investigate the mechanisms involved in RSK4-reduced expression in ovarian cancer. Thus, this study investigates whether RSK4 promoter methylation in ovarian cancer tissues is responsible for its low expression. Additionally, the reactivation of RSK4 expression and its effect was studied in ovarian cancer cell lines. METHODS AND RESULTS RSK4 promoter methylation percentage in malignant and benign ovarian tumors and normal ovary tissues was determined by combined bisulfite restriction analysis. The reactivation of RSK4 expression by decitabine treatment was studied in OVCAR3, SKOV3, TOV-112D, and TOV-21G cells by Western blotting. Cell proliferation was determined by XTT. A significantly high methylation percentage of the RSK4 promoter was observed among malignant and benign ovarian tumors but not in normal ovarian tissue. RSK4 promoter methylation was not associated with age, histological subtype, or stages of ovarian cancer. RSK4 promoter methylation correlates weakly but not significantly with RSK4 protein expression. No correlation was shown between RSK4 methylation and RSK4 mRNA expression. Decitabine induces RSK4 reactivation in all cell lines. However, cell proliferation was reduced only in TOV-112D cells. CONCLUSION These data indicate that although RSK4 promoter methylation is increased in malignant ovarian tumors, this mechanism is unlikely to regulate its expression in ovarian cancer. RSK4 reactivation reduced cell proliferation only in the endometroid histological subtype.
Collapse
Affiliation(s)
- Fabian Arechavaleta-Velasco
- Unidad de Investigación Médica en Medicina Reproductiva, UMAE Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Av. Rio Magdalena No. 289, Sexto piso. Tizapán, San Angel, CP 01090, Mexico City, Mexico
| | - Pablo Dominguez-Lopez
- Unidad de Investigación Médica en Medicina Reproductiva, UMAE Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Av. Rio Magdalena No. 289, Sexto piso. Tizapán, San Angel, CP 01090, Mexico City, Mexico
| | - Ernesto Acosta-Jimenez
- Unidad de Investigación Médica en Medicina Reproductiva, UMAE Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Av. Rio Magdalena No. 289, Sexto piso. Tizapán, San Angel, CP 01090, Mexico City, Mexico
- Hospital Angeles del Pedregal, Mexico City, Mexico
| | - Laura Diaz-Cueto
- Unidad de Investigación Médica en Medicina Reproductiva, UMAE Hospital de Gineco-Obstetricia No. 4 "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Av. Rio Magdalena No. 289, Sexto piso. Tizapán, San Angel, CP 01090, Mexico City, Mexico.
| |
Collapse
|
4
|
Zeng F, Carrasco G, Li B, Sophocleous A, Idris AI. TRAF6 as a potential target in advanced breast cancer: a systematic review, meta-analysis, and bioinformatics validation. Sci Rep 2023; 13:4646. [PMID: 36944688 PMCID: PMC10029787 DOI: 10.1038/s41598-023-31557-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
TRAF6 has emerged as a key regulator of breast cancer (BCa). However, the TRAF family constitutes of seven members that exhibit distinct and overlapping functions. To explore which TRAF represents a potential druggable target for BCa treatment, we searched Medline, Web of Science and Scopus for relevant studies from inception to June 27, 2021. We identified 14 in vitro, 11 in vivo and 4 human articles. A meta-analysis of pharmacological studies showed that in vitro inhibition of TRAF2/4 (mean difference (MD): - 57.49, 95% CI: - 66.95, - 48.02, P < 0.00001) or TRAF6 (standard(Std.)MD: - 4.01, 95% CI: - 5.75, - 2.27, P < 0.00001) is associated with reduction in BCa cell migration. Consistently, inhibition of TRAF2/4 (MD: - 51.08, 95% CI: - 64.23, - 37.94, P < 0.00001) and TRAF6 (Std.MD: - 2.80, 95% CI: - 4.26, - 1.34, P = 0.0002) is associated with reduced BCa cell invasion, whereas TRAF2/4 inhibition (MD: - 40.54, 95% CI: - 52.83, - 28.26, P < 0.00001) is associated with reduced BCa cell adhesion. Interestingly, only inhibition of TRAF6 (MD: - 21.46, 95% CI: - 30.40, - 12.51, P < 0.00001) is associated with reduced cell growth. In animal models of BCa, administration of pharmacological inhibitors of TRAF2/4 (Std.MD: - 3.36, 95% CI: - 4.53, - 2.18, P < 0.00001) or TRAF6 (Std.MD: - 4.15, 95% CI: - 6.06, - 2.24, P < 0.0001) in mice is associated with reduction in tumour burden. In contrast, TRAF6 inhibitors (MD: - 2.42, 95% CI: - 3.70, - 1.14, P = 0.0002) reduced BCa metastasis. In BCa patients, high expression of TRAF6 (Hazard Ratio: 1.01, CI: 1.01, 1.01, P < 0.00001) is associated with poor survival rate. Bioinformatics validation of clinical and pathway and process enrichment analysis in BCa patients confirmed that gain/amplification of TRAF6 is associated with secondary BCa in bone (P = 0.0079), and poor survival rate (P < 0.05). Overall, TRAF6 inhibitors show promise in the treatment of metastatic BCa. However, low study number and scarcity of evidence from animal and human studies may limit the translation of present findings into clinical practice.
Collapse
Affiliation(s)
- Feier Zeng
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Giovana Carrasco
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Boya Li
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenes Street, 1516, Nicosia, Cyprus
| | - Aymen I Idris
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
5
|
The role of long non-coding RNA HCG18 in cancer. Clin Transl Oncol 2023; 25:611-619. [PMID: 36346572 DOI: 10.1007/s12094-022-02992-8] [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: 07/23/2022] [Accepted: 10/23/2022] [Indexed: 11/10/2022]
Abstract
The incidence of cancer is increasing worldwide and is becoming the most common cause of death. Identifying new biomarkers for cancer diagnosis and prognosis is important for developing cancer treatment strategies and reducing mortality. Long non-coding RNAs (lncRNAs) are non-coding, single-stranded RNAs that play an important role as oncogenes or tumor suppressors in the occurrence and development of human tumors. Abnormal expression of human leukocyte antigen complex group 18 (HCG18) is observed in many types of cancer, and its imbalance is closely related to cancer progression. HCG18 regulates cell proliferation, invasion, metastasis, and anti-apoptosis through a variety of mechanisms. Therefore, HCG18 is a potential tumor biomarker and therapeutic target. However, the therapeutic significance of HCG18 has not been well studied, and future research may develop new intervention strategies to combat cancer. In this study, we reviewed the biological function, mechanism, and potential clinical significance of HCG18 in various cancers to provide a reference for future research.
Collapse
|
6
|
Dong X, Li X, Gan Y, Ding J, Wei B, Zhou L, Cui W, Li W. TRAF4-mediated ubiquitination-dependent activation of JNK/Bcl-xL drives radioresistance. Cell Death Dis 2023; 14:102. [PMID: 36765039 PMCID: PMC9918491 DOI: 10.1038/s41419-023-05637-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023]
Abstract
The E3 ligase TNF receptor-associated factor 4 (TRAF4) is upregulated and closely associated with tumorigenesis and the progression of multiple human malignancies. However, its effect on radiosensitivity in colorectal cancer (CRC) has not been elucidated. The present study found that TRAF4 was significantly increased in CRC clinical tumor samples. Depletion of TRAF4 impaired the malignant phenotype of CRC cells and sensitized irradiation-induced cell death. Irradiation activated the c-Jun N-terminal kinases (JNKs)/c-Jun signaling via increasing JNKs K63-linked ubiquitination and phosphorylation. Furthermore, c-Jun activation triggered the transcription of the antiapoptotic protein Bcl-xL, thus contributing to the radioresistance of CRC cells. TRAF4 was positively correlated with c-Jun and Bcl-xL, and blocking TRAF4 or inhibiting Bcl-xL with inhibitor markedly promoted ionizing radiation (IR)-induced intrinsic apoptosis and sensitized CRC cells to radiotherapy in vitro and in vivo. Our findings illustrate a potential mechanism of radioresistance, emphasizing the clinical value of targeting the TRAF4/Bcl-xL axis in CRC therapy.
Collapse
Affiliation(s)
- Xin Dong
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jie Ding
- Department of Anesthesia, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Baojun Wei
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Wei Cui
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
7
|
Wang Y, Luo X, Wu N, Liao Q, Wang J. SRC-3/TRAF4 facilitates ovarian cancer development by activating the PI3K/AKT signaling pathway. Med Oncol 2023; 40:76. [PMID: 36625999 PMCID: PMC9831961 DOI: 10.1007/s12032-022-01944-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Ovarian cancer is the seventh most common cancer in women, and it causes many deaths in women worldwide. Patients with ovarian cancer have a poor prognosis and low survival rate. This study aimed to explore the role of the SRC-3/TRAF4/PI3K/AKT pathway in ovarian cancer development. METHODS SRC-3 and TRAF4 expression in ovarian cancer cell lines were assessed using qRT-PCR and western-blotting. The expression of SRC-3 and TRAF4 in ovarian cancer cells was downregulated by transient transfection with sh-RNAs. An MTT assay was performed to evaluate cell proliferation. Cell migration and invasion were measured using a Transwell assay. Cell stemness was detected using a cell spheroidization assay and western blotting. The expression levels of stem cell factors and PI3K/AKT pathway proteins were determined by qRT-PCR and western blot analysis. RESULTS SRC-3 and TRAF4 were upregulated in ovarian cancer cell lines. TRAF4 is a downstream factor of SRC-3, and the protein level of TRAF4 was regulated by SRC-3. SRC-3 knockdown reduced TRAF4 expression. Silencing SRC-3 or TRAF4 inhibited cell proliferation, migration, and invasion, as well as the expression of stem cell factors. Furthermore, sh-TRAF4 as well as treatment with LY294002, the PI3K/Akt inhibitor, inhibited the phosphorylation of Akt and PI3K, thus repressing the activation of PI3K/AKT signaling pathway in ovarian cancer cell lines. However, TRAF4 overexpression reversed the effect of SRC-3 silencing on cell proliferation, migration, invasion, and stemness. CONCLUSION Our study demonstrated that SRC-3/TRAF4 promotes ovarian cancer cell growth, migration, invasion, and stemness by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Ying Wang
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xia Luo
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Nayiyuan Wu
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jing Wang
- grid.216417.70000 0001 0379 7164Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
8
|
Hao M, Zhang J, Sun M, Diao K, Wang J, Li S, Cao Q, Dai S, Mi X. TRAF4 Inhibits the Apoptosis and Promotes the Proliferation of Breast Cancer Cells by Inhibiting the Ubiquitination of Spindle Assembly-Associated Protein Eg5. Front Oncol 2022; 12:855139. [PMID: 35692762 PMCID: PMC9174544 DOI: 10.3389/fonc.2022.855139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor receptor associated factor 4 (TRAF4) is a RING domain E3 ubiquitin ligase that mediates the ubiquitination of various proteins and plays an important role in driving tumor progression. By studying the relationship between TRAF4 and Eg5, a member of the kinesin family that plays a critical role in spindle assembly, we demonstrated that TRAF4 regulated Eg5 ubiquitination and contributed to Eg5-mediated breast cancer proliferation and inhibited breast cancer apoptosis. TRAF4 and Eg5 were both highly expressed in breast cancer and their protein level was positively correlated. Relying on its Zinc fingers domain, TRAF4 interacted with Eg5 in the cytoplasm of breast cancer cells. TRAF4 was a mitosis-related protein, and by up-regulating the protein level of Eg5 TRAF4 participated in spindle assembly. Loss of TRAF4 resulted in monopolar spindles formation, but loss of function could be rescued by Eg5. Relying on its RING domain, TRAF4 up-regulated Eg5 protein levels by inhibition of Eg5 ubiquitination, thus stabilizing Eg5 protein level during mitosis. Furthermore, we found that Smurf2, a TRAF4-targeted ubiquitination substrate, mediated the regulation of Eg5 ubiquitination by TRAF4. TRAF4 inhibited the interaction between Smurf2 and Eg5, and down-regulated the protein level of Smurf2 by promoting its ubiquitination, thereby inhibited the Smurf2-catalyzed ubiquitination of Eg5 and up-regulated Eg5 protein levels. We also demonstrate that TRAF4 plays an important role in promoting cell proliferation and in inhibiting cell apoptosis induced by Eg5. In summary, our study suggests a new direction for investigating the role of TRAF4 in driving breast cancer progression.
Collapse
Affiliation(s)
- Miaomiao Hao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Pathology, School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Mingfang Sun
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Kexin Diao
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jian Wang
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Shiping Li
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qixue Cao
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Shundong Dai
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaoyi Mi
- Department of Pathology, College of Basic Medical Sciences, First Affiliated Hospital, China Medical University, Shenyang, China
- *Correspondence: Xiaoyi Mi,
| |
Collapse
|
9
|
Ruan X, Zhang R, Li R, Zhu H, Wang Z, Wang C, Cheng Z, Peng H. The Research Progress in Physiological and Pathological Functions of TRAF4. Front Oncol 2022; 12:842072. [PMID: 35242717 PMCID: PMC8885719 DOI: 10.3389/fonc.2022.842072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/26/2022] [Indexed: 11/22/2022] Open
Abstract
Tumour necrosis factor receptor-associated factor 4 (TRAF4) is a member of the TRAF protein family, a cytoplasmic bridging molecule closely associated with various immune functions. The physiological processes of TRAF4 are mainly involved in embryonic development, cell polarity, cell proliferation, apoptosis, regulation of reactive oxygen species production. TRAF4 is overexpressed in a variety of tumors and regulates the formation and development of a variety of tumors. In this review, we summarize the physiological and pathological regulatory functions of TRAF4 and focus on understanding the biological processes involved in this gene, to provide a reference for further studies on the role of this gene in tumorigenesis and development.
Collapse
Affiliation(s)
- Xueqin Ruan
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Rong Zhang
- Division of Cancer Immunotherapy, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Chiba, Japan
| | - Ruijuan Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Hongkai Zhu
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Canfei Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Molecular Hematology, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Long Non-coding RNA ZFPM2-AS1: A Novel Biomarker in the Pathogenesis of Human Cancers. Mol Biotechnol 2022; 64:725-742. [DOI: 10.1007/s12033-021-00443-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022]
|
11
|
Zhang F, Luo BH, Wu QH, Li QL, Yang KD. LncRNA HCG18 upregulates TRAF4/TRAF5 to facilitate proliferation, migration and EMT of epithelial ovarian cancer by targeting miR-29a/b. Mol Med 2022; 28:2. [PMID: 34983361 PMCID: PMC8725507 DOI: 10.1186/s10020-021-00415-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Although long noncoding RNA HLA complex group 18 (lncRNA HCG18) has been suggested to regulate cell growth in several tumours, the function of HCG18 in epithelial ovarian cancer (EOC) and its mechanism are still unclear. Methods shRNAs were applied to reduce HCG18 and related genes. For overexpression of miRNA, a miRNA mimic was transfected into cells. Quantitative real-time PCR (qRT–PCR) was used to detect levels of HCG18, miR-29a/b, and mRNAs. MTT, colony formation, wound healing and Transwell assays were used to evaluate cell proliferation, migration and invasion, respectively. A luciferase reporter assay was utilized to evaluate NF-κB activity and the binding of miRNAs with HCG18 or TRAF4/5. BALB nude mice injected with cells stably expressing shHCG18 or shNC were used for in vivo modelling. Subcutaneous tumour growth was monitored in nude mice, and immunohistochemistry (IHC) was used to determine expression of the proliferation marker Ki67. Results Abnormal expression of HCG18 and miR-29a/b was observed in EOC tissues. Knockdown of HCG18 using shRNA inhibited proliferation, migration, EMT and the proinflammatory pathway in EOC cells. miR-29a/b mimics and TRAF4/5 knockdown exhibited effects similar to HCG18 knockdown. Further experiments suggested that HCG18 directly targets miR-29a/b and upregulates TRAF4/5 expression, which are inhibited by targeting miR-29a/b. Moreover, overexpression of TRAF4/5 antagonized the inhibitory effect of HCG18 knockdown, suggesting that they are involved in HCG18-mediated oncogenic effects. Silencing HCG18 reduced tumour size and levels of Ki67 and TRAF4/5 while increasing miR-29a/b levels in vivo. Conclusions Taken together, our data revealed an oncogenic signalling pathway mediated by HCG18 in ovarian cell lines, which functions as a ceRNA of miR-29a/b and thus derepresses expression levels of TRAF4/5, facilitating NF-κB pathway-mediated promotion of EOC cell proliferation and migration. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00415-y.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China.,Department of Physiology, School of Basic Medical Science, Central South University, Changsha, 410008, Hunan Province, People's Republic of China
| | - Bai-Hua Luo
- Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan Province, People's Republic of China
| | - Qi-Hui Wu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan Province, People's Republic of China
| | - Qing-Ling Li
- Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan Province, People's Republic of China
| | - Ke-Da Yang
- Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Kaifu District, Changsha, 410008, Hunan Province, People's Republic of China.
| |
Collapse
|
12
|
ZFPM2-AS1 facilitates cell growth in esophageal squamous cell carcinoma via up-regulating TRAF4. Biosci Rep 2021; 40:222148. [PMID: 32065218 PMCID: PMC7133517 DOI: 10.1042/bsr20194352] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence has confirmed that long noncoding RNAs (lncRNAs) are strongly involved in tumor initiation and development. LncRNA ZFPM2 antisense RNA 1 (ZFPM2-AS1) has been identified as a tumor facilitator in some cancers; nevertheless, its functional significance and regulatory mechanism remain greatly unclear in esophageal squamous cell carcinoma (ESCC). Here, we detected ZFPM2-AS1 expression in ESCC cell lines using qRT-PCR. ZFPM2-AS1 knockdown models were established for investigating the biological function of ZFPM2-AS1 in ESCC cells. The association between miR-3612 and ZFPM2-AS1 or TRAF4 was assessed by RNA pull-down and luciferase reporter assays. The present study indicated that ZFPM2-AS1 was significantly up-regulated in ESCC cells. Functional assays manifested that ZFPM2-AS1 knockdown restrained cell proliferation, migration and invasion, and facilitated cell apoptosis in ESCC. Mechanistically, ZFPM2-AS1 promoted ESCC cell growth and up-regulated TRAF4 to trigger NF-κB pathway by sequestering miR-3612. Besides, miR-3612 was confirmed to be a tumor inhibitor in ESCC. Through restoration experiments, we observed that TRAF4 overexpression could recover the suppressive effect of ZFPM2-AS1 on ESCC cell growth. Collectively, all the results suggested that ZFPM2-AS1 was an oncogene in ESCC cell growth by up-regulating TRAF4 and activating NF-κB pathway.
Collapse
|
13
|
Xu J, Jia Q, Zhang Y, Yuan Y, Xu T, Yu K, Chai J, Wang K, Chen L, Xiao T, Li M. Prominent roles of ribosomal S6 kinase 4 (RSK4) in cancer. Pathol Res Pract 2021; 219:153374. [PMID: 33621918 DOI: 10.1016/j.prp.2021.153374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 11/19/2022]
Abstract
RSK4 refers to one Ser/Thr protein kinase functioning downstream pertaining to the signaling channel of protein kinase (MAPK) stimulated by Ras/mitogen. RSK4 can regulate numerous substrates impacting cells' surviving state, growing processes and proliferating process. Thus, dysregulated RSK4 active state display a relationship to several carcinoma categories, covering breast carcinoma, esophageal squamous cell carcinoma, glioma, colorectal carcinoma, lung carcinoma, ovarian carcinoma, leukemia, endometrial carcinoma, and kidney carcinoma. Whether RSK4 is a tumor suppressor gene or one oncogene remains controversial. No specific inhibiting elements for RSK4 have been found. This review briefs the existing information regarding RSK4 activating process, the function and mechanism of RSK4 in different tumors, and the research progress and limitations of existing RSK inhibitors. RSK4 may be a potential target of molecular therapy medicine in the future.
Collapse
Affiliation(s)
- Junpeng Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qingge Jia
- Xi'an International Medical Center, Northwest University, Xi'an, China
| | - Yan Zhang
- Children's Heart Disease Center, Sichuan Maternal and Child Health Hospital, Chengdu, China
| | - Yuan Yuan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kangjie Yu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jia Chai
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kaijing Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ligang Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Tian Xiao
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
14
|
Chen L, Xu T, Jia Q, Wang X, Li M, Liang G. RSK4: a new prognostic factor in glioma. Pathol Res Pract 2020; 216:153020. [PMID: 32703488 DOI: 10.1016/j.prp.2020.153020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022]
Abstract
Glioma is the most common and fatal brain tumour and has a poor prognosis. Ribosomal S6 protein kinase 4 (RSK4) has been found to be involved in multiple tumour types; however, the role of RSK4 in gliomas and its clinical relevance remain unclear. In the present study, RSK4 expression was found to be significantly increased in glioma tissues compared with matched adjunct non-noncancerous tissues. Moreover, the expression of RSK4 was significantly higher in high-grade (III and IV) glioma tissues than in low-grade (I and II) glioma tissues. The data showed that the expression of RSK4 was significantly correlated with WHO grade, three-year survival rate and five-year survival rate. Kaplan-Meier analyses showed that patients with high RSK4 expression had poor overall survival. In addition, multivariate Cox regression analysis showed that RSK4 might be an independent prognostic factor in glioma patients. Collectively, these results suggest that RSK4 may be a new prognostic factor in glioma patients, and RSK4 is expected to be a potential biomarker and a potential target for glioma therapy.
Collapse
Affiliation(s)
- Ligang Chen
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Qingge Jia
- Second Retired Cadres Sanitarium of Xi'an, Shaanxi Province Military Region, Xi'an 710032, China
| | - Xiaokai Wang
- Department of Pathology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Guobiao Liang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang 110016, China.
| |
Collapse
|
15
|
TRAF4 knockdown triggers synergistic lethality with simultaneous PARP1 inhibition in endometrial cancer. Hum Cell 2020; 33:801-809. [PMID: 32388810 DOI: 10.1007/s13577-020-00363-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/14/2020] [Indexed: 10/24/2022]
Abstract
Endometrial cancer (EC) is one of the most common cancers among females worldwide. Advanced stage patients of EC have poor prognosis. Inevitable side effects and treatment tolerance of chemotherapy for EC remain to be addressed. Our results in this study showed that EC cells with higher tumor necrosis factor receptor-associated factor 4 (TRAF4) expression have lower sensitivity to poly ADP-ribose polymerase 1 (PARP1) inhibitors. Upon TRAF4 knockdown, the colony numbers of EC cells were markedly down-regulated, and the markers of DNA double-strand breakage were significantly up-regulated after the treatment of olaparib, a PARP1 inhibitor. TRAF4 knockdown reduced the phosphorylation of protein kinase B (Akt), promoted DNA double-strand breakage, and decreased levels of DNA repair related proteins, including phosphorylated-DNA-dependent protein kinase (p-DNA-PK) and RAD51 recombinase (RAD51). In addition, TRAF4's effect on the sensitivity of EC cells to olaparib was further found to be mainly mediated by Akt phosphorylation. Moreover, in vivo results showed that TRAF4 knockdown enhanced the sensitivity of EC to PARP1 inhibitors using a mouse xenograft model. Collectively, our data suggest that combined application of TRAF4 knockdown and PARP1 inhibition can be used as a promising strategy for synthetic lethality in EC treatment.
Collapse
|
16
|
Ectopic Expression of miR-532-3p Suppresses Bone Metastasis of Prostate Cancer Cells via Inactivating NF-κB Signaling. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:267-277. [PMID: 32368615 PMCID: PMC7191128 DOI: 10.1016/j.omto.2020.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 11/23/2022]
Abstract
miR-532-3p is a widely documented microRNA (miRNA) involved in multifaceted processes of cancer tumorigenesis and metastasis. However, the clinical significance and biological functions of miR-532-3p in bone metastasis of prostate cancer (PCa) remain largely unknown. Herein, we report that miR-532-3p was downregulated in PCa tissues with bone metastasis, and downexpression of miR-532-3p was significantly associated with Gleason grade and serum prostate-specific antigen (PSA) levels and predicted poor bone metastasis-free survival in PCa patients. Upregulating miR-532-3p inhibited invasion and migration abilities of PCa cells in vitro, while silencing miR-532-3p yielded an opposite effect on invasion and migration abilities of PCa cells. Importantly, upregulating miR-532-3p repressed bone metastasis of PCa cells in vivo. Our results further demonstrated that overexpression of miR-532-3p inhibited activation of nuclear facto κB (NF-κB) signaling via simultaneously targeting tumor necrosis factor receptor-associated factor 1 (TRAF1), TRAF2, and TRAF4, which further promoted invasion, migration, and bone metastasis of PCa cells. Therefore, our findings reveal a novel mechanism contributing to the sustained activity of NF-κB signaling underlying the bone metastasis of PCa.
Collapse
|
17
|
Yan Z, Yang Q, Xue M, Wang S, Hong W, Gao X. YY1-induced lncRNA ZFPM2-AS1 facilitates cell proliferation and invasion in small cell lung cancer via upregulating of TRAF4. Cancer Cell Int 2020; 20:108. [PMID: 32280300 PMCID: PMC7126398 DOI: 10.1186/s12935-020-1157-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/29/2020] [Indexed: 12/26/2022] Open
Abstract
Background Newly identified lncRNA zinc finger protein, FOG family member 2 antisense RNA 1 (ZFPM2-AS1) is identified as an oncogenic gene. However, the role of ZFPM2-AS1 in small cell lung cancer (SCLC) is poorly comprehended. Methods The expression of genes in SCLC tissues and cells was measured by qRT-PCR. Colony formation, EdU, CCK-8, transwell and wound healing as well as in vivo assays revealed the function of ZFPM2-AS1 in SCLC. ChIP, luciferase reporter, RIP and RNA pull down assays demonstrated the binding relation among genes. Results ZFPM2-AS1 was significantly upregulated in SCLC tissues and cells. ZFPM2-AS1 deficiency attenuated SCLC cell proliferation, invasion and migration. In addition, ZFPM2-AS1 was transcriptionally activated by Yin Yang 1 (YY1) factor. Further, miR-3612 was confirmed as downstream miRNA of ZFPM2-AS1. Moreover, TNF receptor associated factor 4 (TRAF4) was the target gene of miR-3612 in SCLC. ZFPM2-AS1, miR-3612 and TRAF4 jointly constituted a competing endogenous RNA (ceRNA) network in SCLC. Finally, TRAF4 could countervail ZFPM2-AS1 downregulation-mediated function on SCLC cell proliferation and invasion in vitro and tumor growth in vivo. Conclusion Our study elucidated the oncogenic effect of ZFPM2-AS1 in SCLC progression, indicating it may be a therapeutic target for SCLC.
Collapse
Affiliation(s)
- Zhijun Yan
- 1Department of Respiratory Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199 China
| | - Qilian Yang
- 2Department of Pharmacy, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199 China
| | - Min Xue
- 1Department of Respiratory Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199 China
| | - Sheng Wang
- 3Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai, 200032 China
| | - Weijun Hong
- 1Department of Respiratory Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199 China
| | - Xiwen Gao
- 1Department of Respiratory Medicine, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai, 201199 China
| |
Collapse
|
18
|
Dai H, Zhang S, Ma R, Pan L. Celecoxib Inhibits Hepatocellular Carcinoma Cell Growth and Migration by Targeting PNO1. Med Sci Monit 2019; 25:7351-7360. [PMID: 31568401 PMCID: PMC6784684 DOI: 10.12659/msm.919218] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Celecoxib has shown anti-tumor activities against several types of cancer. Although the majority of research focuses on its mechanism via cyclooxygenase-2 (COX-2) enzyme inhibition, we identified a distinct mechanism behind celecoxib anti-cancer abilities. Material/Methods We treated hepatocellular carcinoma (HCC) Huh-7 cells and tumor xenograft mice models with celecoxib to test its effects on the tumor. Using gene chip method to identify the differential expressed genes after celecoxib treatment and using pathway enrichment analysis to predict the potential pathways for further study. We transfected cells with lentiviral shRNA to detect the effect of RNA binding gene partner of NOB1 (PNO1) on tumor growth in vitro and in vivo. Further we performed western blot to detect the effect of PNO1 on the protein kinase B (AKT) pathway. Results Celecoxib inhibited HCC cell growth in vitro and in vivo, and gene chip and pathway enrichment analysis revealed that PNO1 may be the potential target of celecoxib in HCC cells. Celecoxib significantly reduced levels of PNO1 in tumor tissue. Knockdown of PNO1 remarkably suppressed tumor growth and metastasis in vitro and in vivo. Disruption of PNO1 expression significantly reduced protein kinase B (AKT)/rapamycin (mTOR) signaling, indicating that this pathway may be involved in PNO1-mediated tumorigenic activity. Conclusions Celecoxib may exert its anti-tumor activity by inhibiting PNO1, and that AKT/mTOR signaling helps mediate the oncogenic effects of PNO1. This work offers the first evidence for a role of PNO1 as an HCC oncogene, which may open new avenues for prevention and treatment of HCC.
Collapse
Affiliation(s)
- Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland).,Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Suisui Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Riliang Ma
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland).,Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China (mainland)
| |
Collapse
|
19
|
Wang J, Quan X, Peng D, Hu G. Long non‑coding RNA DLEU1 promotes cell proliferation of glioblastoma multiforme. Mol Med Rep 2019; 20:1873-1882. [PMID: 31257517 DOI: 10.3892/mmr.2019.10428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/10/2019] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant tumor with high morbidity and mortality. This study investigated the role of long non‑coding RNAs (lncRNAs) in glioblastomagenesis progression. Using the GSE2223 and GSE59612 datasets, and RNA sequencing data of GBM from The Cancer Genome Atlas, differentially expressed (DE) genes including DE messenger RNAs (DEmRNAs) and DElncRNAs between GBM and normal controls were identified. Based on the competing endogenous RNA hypothesis, DElncRNA‑micro RNA (miRNA)‑DEmRNA interactions were obtained by target gene prediction. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes pathway analysis of DEmRNAs in the DElncRNA‑miRNA‑DEmRNA network was performed. Expression and function analyses of DElncRNAs were performed by reverse transcription‑polymerase chain reaction (RT‑PCR) and an established viability assay, respectively. In total, 712 DE genes were identified. Significant upregulation of lncRNA deleted in lymphocytic leukemia 1 (DLEU1) was revealed in GBM and a number of other types of cancer. DLEU1 interacted with 315 miRNAs and 105 DEmRNAs. The DEmRNAs were mainly enriched in tumorigenesis‑associated GO terms (angiogenesis, positive regulation of cell proliferation, positive regulation of fibroblast apoptotic processes and regulation of neutrophil migration) and pathways (Hippo signaling pathway, cancer pathways, and Wnt signaling pathway). Correlation analysis revealed that mRNA TNF receptor associated factor 4 (TRAF4) was associated with DLEU1 expression. RT‑PCR demonstrated that the expression levels of DLEU1 and TRAF4 were increased in GBM tissues. Small interfering RNA demonstrated that silencing DLEU1 downregulated TRAF4. The viability of GBM cells was significantly decreased following RNA interference with DLEU1 and TRAF4 production. The results demonstrate that DLEU1 and TRAF4 is highly expressed in GBM tissues and promotes proliferation of GBM cells. It may act as a competing endogenous RNA and influence tumorigenesis of GBM.
Collapse
Affiliation(s)
- Jiancun Wang
- Department of Neurosurgery, People's Hospital of Zhangjiajie, Zhangjiajie, Hunan 427000, P.R. China
| | - Xingyun Quan
- Department of Neurosurgery, People's Hospital of Zhangjiajie, Zhangjiajie, Hunan 427000, P.R. China
| | - Dingting Peng
- Department of Neurosurgery, People's Hospital of Zhangjiajie, Zhangjiajie, Hunan 427000, P.R. China
| | - Guancheng Hu
- Department of Neurosurgery, People's Hospital of Zhangjiajie, Zhangjiajie, Hunan 427000, P.R. China
| |
Collapse
|
20
|
Jiang W, Chen M, Xiao C, Yang W, Qin Q, Tan Q, Liang Z, Liao X, Mao A, Wei C. Triptolide Suppresses Growth of Breast Cancer by Targeting HMGB1 in Vitro and in Vivo. Biol Pharm Bull 2019; 42:892-899. [PMID: 30956264 DOI: 10.1248/bpb.b18-00818] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Triptolide has been indicated potent anti-cancer effect involving multiple molecular targets and signaling pathways. High-mobility group box 1 (HMGB1) is a highly conserved DNA-binding protein taking part in breast cancer development. The therapeutic effect of triptolide on HMGB1 has not been reported. Thus, our study aims to clarify the role of HMGB1 in triptolide-induced anti-growth effect on breast cancer in vitro and in vivo. We demonstrated that triptolide significantly suppressed growth of breast cancer cells by inhibition of cell viability, clonogenic ability. Further studies evidenced that triptolide treatment not only inhibited HMGB1 mRNA expression, but also decreased supernatant level of HMGB1 in vitro. In line with these observations, exogenous recombinant HMGB1 (rHMGB1) promoted cell proliferation of breast cancer, and triptolide reversed the rHMGB1-promoted proliferative effect. As well, triptolide enhanced the anti-proliferative activity of ethyl pyruvate (EP) (HMGB1 inhibitor). Furthermore, downstream correlation factors (Toll-like receptor 4 (TLR4) and phosphorylated-nuclear factor-kappaB (NF-κB) p65) of HMGB1 were significantly decreased in vitro after triptolide treatment. Consistantly, we confirmed that tumor growth was significantly inhibited after triptolide treatment in vivo. Meanwhile, immunohistochemical analyses showed that triptolide treatment significantly decreased the level of cytoplasmic HMGB1 and TLR4 expression, whereas the expression of NF-κB p65 was relatively higher in cytoplasm, and conversely lower in nucleus as compared to the control group. Collectively, these results demonstrate that triptolide suppresses the growth of breast cancer cells via reduction of HMGB1 expression in vitro and in vivo, which may provide new insights into the treament of breast cancer.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Medical Oncology, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Maojian Chen
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Chanchan Xiao
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Weiping Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Qinghong Qin
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Qixing Tan
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Zhijie Liang
- Department of Breast and Thyroid Surgery, The Fifth Affliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning
| | - Xiaoli Liao
- Department of Medical Oncology, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Anyun Mao
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Changyuan Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| |
Collapse
|
21
|
Jia YC, Wang JY, Liu YY, Li B, Guo H, Zang AM. LncRNA MAFG-AS1 facilitates the migration and invasion of NSCLC cell via sponging miR-339-5p from MMP15. Cell Biol Int 2019; 43:384-393. [PMID: 30599080 DOI: 10.1002/cbin.11092] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
Abstract
Non-small-cell carcinoma (NSCLC) is the most common cancer along with high mortality rate worldwide. In the present study, our data showed that lncRNA MAF BZIP Transcription Factor G Antisense RNA 1 (MAFG-AS1) was over-expressed in NSCLC tissues and cell lines. Overexpression of MAFG-AS1 promoted the migration, invasion and enhanced epithelial-mesenchymal transition (EMT) of NSCLC cell. In addition, miR-339-5p was predicted to be a target of MAFG-AS1 and the level of miR-339-5p was down-regulated in NSCLC. Over-expression of MAFG-AS1 significantly decreased the level of miR-339-5p in NSCLC cell. Moreover, the matrix metalloproteinase 15 (MMP15) was identified to be a target of miR-339-5p. The level of MMP15 was negatively regulated by miR-339-5p whereas positively controlled by MAFG-AS1. In addition, up-regulation of miR-339-5p neutralized the promoting impact of MAFG-AS1 on the migration, invasion and EMT of NSCLC cell. Finally, the xenograft model suggested that MAFG-AS1 promoted the metastasis of NSCLC cell in vivo. Altogether, we proved that MAFG-AS1-miR-339-5p-MMP15 axis might be a promising therapeutic target for the treatment of patients with NSCLC.
Collapse
Affiliation(s)
- You Chao Jia
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| | | | - Yu Ying Liu
- Department of Oncology, Dezhou People's Hospital, Dezhou, Shandong, 253014, China
| | - Bin Li
- Thoracic surgery, Dezhou People's Hospital, Dezhou, Shandong, 253014, China
| | - Hui Guo
- Pneumology Department, Dezhou People's Hospital, No. 1751 Xinhu Street, Dezhou, Shandong, 253014, China
| | - Ai Min Zang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, China
| |
Collapse
|
22
|
Zhu S, Jin J, Gokhale S, Lu AM, Shan H, Feng J, Xie P. Genetic Alterations of TRAF Proteins in Human Cancers. Front Immunol 2018; 9:2111. [PMID: 30294322 PMCID: PMC6158389 DOI: 10.3389/fimmu.2018.02111] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/28/2018] [Indexed: 12/25/2022] Open
Abstract
The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Angeli M. Lu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Haiyan Shan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianjun Feng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education of the People's Republic of China, Fisheries College of Jimei University, Xiamen, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Member, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
23
|
Zhu S, Jin J, Gokhale S, Lu AM, Shan H, Feng J, Xie P. Genetic Alterations of TRAF Proteins in Human Cancers. Front Immunol 2018. [PMID: 30294322 DOI: 10.3389/fimmu.2018.02111/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Angeli M Lu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Haiyan Shan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianjun Feng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education of the People's Republic of China, Fisheries College of Jimei University, Xiamen, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Member, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|