1
|
Zhang Y, Yang F. FAM84B promotes breast cancer tumorigenesis through activation of the NF-κB and death receptor signaling pathways. Pathol Res Pract 2023; 249:154785. [PMID: 37651838 DOI: 10.1016/j.prp.2023.154785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/31/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Breast cancer (BC) occurs predominantly in women and leads to numerous deaths every year. The identification of effective therapeutic targets will benefit BC patients and increase the likelihood of finding a cure. Family with similar sequence 84, member B (FAM84B) has been implicated in the progression of many kinds of cancers, but its function in BC remains to be explored. In this study, online database analysis revealed that FAM84B expression was higher in BC patient tissues, especially in luminal BC tissues, than in the corresponding normal tissues; furthermore, increased FAM84B expression was related to poor prognosis. Additionally, western blot (WB) analysis revealed that the FAM84B protein was highly expressed in luminal BC cell lines compared to normal and basal-like BC cell lines. Moreover, clinical BC patient tissues were collected and subjected to WB and immunohistochemical (IHC) analyses, and the results showed that FAM84B was expressed mainly in luminal BC samples. Therefore, to determine the function of FAM84B in luminal BC cells, luminal BC cell lines with FAM84B knockout and overexpression were generated. In addition, the functions of FAM84B were evaluated in vitro (via cell proliferation, wound healing, colony formation and invasion assays) and in vivo (via a subcutaneous xenograft experiment), and the results showed that FAM84B regulated cell proliferation but not cell invasion. Furthermore, the results of RNA sequencing analysis in ZR-75-1 FAM84B knockout and FAM84B-overexpressing cells showed that FAM84B could affect the TNF signaling pathway. Subsequently, WB analysis of death receptor signaling and immunofluorescence (IF) analysis of NF-κB p65 localization revealed that FAM84B affected death receptor signaling and promoted NF-κB p65 nuclear entry. In conclusion, we found that FAM84B promotes luminal BC tumorigenesis through the activation of the NF-κB and death receptor signaling pathways.
Collapse
Affiliation(s)
- Yanhua Zhang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fang Yang
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Ren D, Zhuang X, Lv Y, Zhang Y, Xu J, Gao F, Chen D, Wang Y. FAM84B promotes the proliferation of glioma cells through the cell cycle pathways. World J Surg Oncol 2022; 20:368. [PMID: 36419094 PMCID: PMC9686022 DOI: 10.1186/s12957-022-02831-8] [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: 01/11/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background This study aimed to investigate FAM84B expression in glioma tissues and explore the role of FAM84B in promoting the proliferation of glioma cells and the mechanism of regulating the cell cycle pathways. Methods The TCGA database was adopted to analyze FAM84B expression in glioma tissues. The FAM84B expression was detected by qRT-PCR in patients with glioma, especially that in glioma cells, U251, LN-229, U98, and U87. Two glioma cell lines U87 and T98 were selected for siRNA transfection, which were divided into si-NC si-FAM84B-1 and si-FAM84B-2 groups. The effect of FAM84B on the proliferation of glioma cells was detected with the MTT experiment and that on the glioma cell cycle was detected with the flow cytometry. The signaling pathways potentially regulated by FAM84B in glioma were analyzed through the bioinformatics analysis. The expression of proteins, Cyclin D1, CDK4, Cdk6, and p21, in the cell cycle-related pathways in cells of each group was detected by the Western blot. Results TCGA database results showed a significantly higher FAM84B expression in glioma tissues than that in paracancerous tissues. According to the detection of qRT-PCR, FAM84B expressed the highest in the glioma cell line U87 (P < 0.05). Compared with the serum of healthy controls, FAM84B mRNA expression significantly increased in patients with gliomas. And compared with the si-NC group, the proliferation ability of U87 and T98 cells decreased and the cell cycle was blocked in the G0/G1 phase in both si-FAM84B transfection groups (P < 0.05). According to the bioinformatics analysis, FAM84B regulated the cell cycle pathways in glioma. FAM84B siRNA inhibited the expression of key proteins, Cyclin D1, CDK2, CDK4, and Cdk6, of the cell cycle pathways in glioma cells and promoted the expression of P53 and P21 proteins. Conclusions In conclusion, FAM84B may inhibit the proliferation of glioma cells by regulating the cell cycle pathways. 1. FAM84B expressed highly in glioma tissues and cells. 2. Knockdown of FAM84B expression significantly inhibited the proliferation of glioma cells. 3. Knockdown of FAM84B inhibited the proliferation of glioma cells by regulating the cell cycle signaling pathways.
Collapse
|
3
|
Jiang J, Cheng Y, Dai S, Zou B, Guo X. Suppression of rhomboid domain-containing 1 produces anticancer effects in pancreatic adenocarcinoma through affection of the AKT/GSK-3β/β-catenin pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:1944-1956. [PMID: 35442567 DOI: 10.1002/tox.23541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/20/2022] [Accepted: 04/10/2022] [Indexed: 06/14/2023]
Abstract
The protumor role of rhomboid domain-containing 1 (RHBDD1) has been observed in multiple cancers. However, the relationship between RHBDD1 and pancreatic adenocarcinoma has not been addressed. This project focused on the potential relevance of RHBDD1 in pancreatic adenocarcinoma. Bioinformatic analysis by publicly available data revealed that RHBDD1 was abundantly expressed in pancreatic adenocarcinoma. We further verified that RHBDD1 was expressed highly in clinical specimens of pancreatic adenocarcinoma. The Kaplan-Meier curve demonstrated that high-RHBDD1 expression was associated with poor prognosis in pancreatic adenocarcinoma patients. The functional studies revealed that depletion of RHBDD1 produced in vitro anticancer effects in pancreatic adenocarcinoma cells, including retardation of proliferation, reduction of metastatic potential, and induction of cell-cycle arrest at the G0/G1 phase and apoptosis. Mechanistic studies indicated that loss of RHBDD1 affected the activation of β-catenin via regulation of AKT. Forced expression of β-catenin reversed the RHBDD1-loss-induced anticancer effects in pancreatic adenocarcinoma cells. Crucially, depletion of RHBDD1 retarded the growth of pancreatic adenocarcinoma xenografts in vivo, a phenomenon associated with the AKT/β-catenin pathway. Collectively, these findings delineated that restraint of RHBDD1 displayed remarkable anticancer effects in pancreatic adenocarcinoma by affecting the AKT/β-catenin pathway. Our work unveils a pivotal role of RHBDD1 in pancreatic adenocarcinoma and proposes it as a novel candidate target for anticancer therapy of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Jiong Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Cheng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shejiao Dai
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Baicang Zou
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
Canisius J, Wagner A, Bunk EC, Spille DC, Stögbauer L, Grauer O, Hess K, Thomas C, Paulus W, Stummer W, Senner V, Brokinkel B. Expression of decitabine-targeted oncogenes in meningiomas in vivo. Neurosurg Rev 2022; 45:2767-2775. [PMID: 35445910 PMCID: PMC9349086 DOI: 10.1007/s10143-022-01789-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/11/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
Treatment of meningiomas refractory to surgery and irradiation is challenging and effective chemotherapies are still lacking. Recently, in vitro analyses revealed decitabine (DCT, 5-aza-2’–deoxycytidine) to be effective in high-grade meningiomas and, moreover, to induce hypomethylation of distinct oncogenes only sparsely described in meningiomas in vivo yet. Expression of the corresponding onco- and tumor suppressor genes TRIM58, FAM84B, ELOVL2, MAL2, LMO3, and DIO3 were analyzed and scored by immunohistochemical staining and RT-PCR in samples of 111 meningioma patients. Correlations with clinical and histological variables and prognosis were analyzed in uni- and multivariate analyses. All analyzed oncogenes were highly expressed in meningiomas. Expression scores of TRIM58 tended to be higher in benign than in high-grade tumors 20 vs 16 (p = .002) and all 9 samples lacking TRIM58 expression displayed WHO grade II/III histology. In contrast, median expression scores for both FAM84B (6 vs 4, p ≤ .001) and ELOVL2 (9 vs 6, p < .001) were increased in high-grade as compared to benign meningiomas. DIO3 expression was distinctly higher in all analyzed samples as compared to the reference decitabine-resistant Ben-Men 1 cell line. Increased ELOVL2 expression (score ≥ 8) correlated with tumor relapse in both uni- (HR: 2.42, 95%CI 1.18–4.94; p = .015) and multivariate (HR: 2.09, 95%CI 1.01–4.44; p = .046) analyses. All oncogenes involved in DCT efficacy in vitro are also widely expressed in vivo, and expression is partially associated with histology and prognosis. These results strongly encourage further analyses of DCT efficiency in meningiomas in vitro and in situ.
Collapse
Affiliation(s)
- Julian Canisius
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
| | - Andrea Wagner
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
| | - Eva Christina Bunk
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Dorothee Cäcilia Spille
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Louise Stögbauer
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Oliver Grauer
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, North Rhine-Westphalia, Münster, Germany
| | - Katharina Hess
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
- Department of Pathology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christian Thomas
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
| | - Werner Paulus
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany
| | - Volker Senner
- Institute of Neuropathology, University Hospital Münster, Münster, North Rhine Westphalia, Germany
| | - Benjamin Brokinkel
- Department of Neurosurgery, University Hospital Münster, North Rhine Westphalia, Albert-Schweitzer-Campus 1, Building A1, 48149, Münster, Germany.
| |
Collapse
|
5
|
Elevated FAM84B promotes cell proliferation via interacting with NPM1 in esophageal squamous cell carcinoma. Cell Death Dis 2022; 8:182. [PMID: 35396552 PMCID: PMC8993864 DOI: 10.1038/s41420-022-00984-9] [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: 02/09/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022]
Abstract
Family with sequence similarity 84, member B (FAM84B) is a significant copy number amplification gene in the 8q24.21 locus identified by our previous WGS study in esophageal squamous cell carcinoma (ESCC). However, its clinical relevance and potential mechanisms have been elusive. Here, we performed the association analyses between FAM84BAmp and clinicopathological features using 507 ESCC samples. The results indicated that, compared with the FAM84Bnon-Amp patients, the FAM84BAmp patients showed a more aggressive and a worse prognosis. A significant correlation was discovered between the expression level of FAM84B and FAM84BAmp in the ESCC cohort. Furthermore, we found that the forced expression change of FAM84B can influence ESCC cell proliferation and cell-cycle status, which is probably mediated by NPM1. A direct interaction between FAM84B and the C-terminal (189–294aa) of NPM1 was identified, which increased the NPM1 nuclear expression. Over-expression of NPM1 could inhibit the CDKN2A protein expression, which might affect the ESCC cell cycle. Our results indicate FAM84B CNA may be a potential diagnostic and therapeutic biomarker in ESCC, meanwhile, reveal a novel mechanism of FAM84B that promotes tumorigenesis via interacting with NPM1 and suppressing CDKN2A.
Collapse
|
6
|
Zhang Z, Zhang HJ. Glycometabolic rearrangements-aerobic glycolysis in pancreatic ductal adenocarcinoma (PDAC): roles, regulatory networks, and therapeutic potential. Expert Opin Ther Targets 2021; 25:1077-1093. [PMID: 34874212 DOI: 10.1080/14728222.2021.2015321] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Glycometabolic rearrangements (aerobic glycolysis) is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and contributes to tumorigenesis and progression through numerous mechanisms. The targeting of aerobic glycolysis is recognized as a potential therapeutic strategy which offers the possibility of improving treatment outcomes for PDAC patients. AREAS COVERED In this review, the role of aerobic glycolysis and its regulatory networks in PDAC are discussed. The targeting of aerobic glycolysis in PDAC is examined, and its therapeutic potential is evaluated. The relevant literature published from 2001 to 2021 was searched in databases including PubMed, Scopus, and Embase. EXPERT OPINION Regulatory networks of aerobic glycolysis in PDAC are based on key factors such as c-Myc, hypoxia-inducible factor 1α, the mammalian target of rapamycin pathway, and non-coding RNAs. Experimental evidence suggests that modulators or inhibitors of aerobic glycolysis promote therapeutic effects in preclinical tumor models. Nevertheless, successful clinical translation of drugs that target aerobic glycolysis in PDAC is an obstacle. Moreover, it is necessary to identify the potential targets for future interventions from regulatory networks to design efficacious and safer agents.
Collapse
Affiliation(s)
- Zhong Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
7
|
Wang M, Li C, Shi W. FAM84B acts as a tumor promoter in human glioma via affecting the Akt/GSK-3β/β-catenin pathway. Biofactors 2021; 47:600-611. [PMID: 33759248 DOI: 10.1002/biof.1727] [Citation(s) in RCA: 4] [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: 01/29/2021] [Accepted: 03/03/2021] [Indexed: 01/23/2023]
Abstract
Family with sequence similarity 84, member B (FAM84B) has recently emerged as an oncoprotein in multiple types of cancer. However, whether FAM84B modulates the progression of glioma has not been determined. The goals of this work were to assess the possible relationship between FAM84B and glioma. Our data revealed high FAM84B level in glioma specimens and exhibited that the overexpression of FAM84B was correlated with a low survival rate in glioma patients. Cellular functional assays showed that silencing of FAM84B prohibited the proliferation and invasion, and induced the apoptosis of glioma cells. Further results determined that the knockdown of FAM84B remarkably decreased the levels of phosphorylated Akt and glycogen synthase kinase (GSK)-3β, and active β-catenin. Inhibition of Akt abolished the FAM84B-mediated promotion effects on Wnt/β-catenin pathway. The subcutaneous xenograft assay confirmed that the silencing of FAM84B significantly prohibited the tumorigenicity of glioma cells in vivo. Collectively, the findings from this work demonstrate that the downregulation of FAM84B exhibits a cancer-suppressive role in human glioma through the regulation of Akt/GSK-3β/β-catenin pathway.
Collapse
Affiliation(s)
- Minjuan Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Neurology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Chengliang Li
- Department of General Practice, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wei Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Zhou Y, Zhang Y, Wang J. Trefoil Factor 2 Regulates Proliferation and Apoptosis of Pancreatic Cancer Cells and LPS-Induced Normal Pancreatic Duct Cells by β-Catenin Pathway. Cancer Manag Res 2020; 12:10705-10713. [PMID: 33149677 PMCID: PMC7605628 DOI: 10.2147/cmar.s274578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 01/13/2023] Open
Abstract
Introduction Pancreatic cancer (PC) is a malignant tumor with poor prognosis. This study aimed to determine the role of trefoil factor 2 (TFF2) in the proliferation and apoptosis of LPS-induced normal pancreatic duct cells and pancreatic cancer cells through β-catenin pathway. Methods TFF2 expression in normal pancreatic duct cells, pancreatic cancer cells and LPS-induced normal pancreatic duct cells was detected by RT-qPCR analysis and Western blot analysis. The transfection effects in pancreatic cancer cells and LPS-induced normal pancreatic duct cells were analyzed by RT-qPCR analysis. After indicated transfection, proliferation, apoptosis and inflammation of these cells were respectively detected by CCK-8 assay, TUNEL assay and certain ELISA kits. Expression of β-catenin pathway-related proteins was analyzed by Western blot analysis. Co-immunoprecipitation assay determined the combination of TFF2 and β-catenin. Results TFF2 expression was increased in pancreatic cancer cells and LPS-induced HPDE cells compared with HPDE cells. According to TFF2 expression in these cells, PanC-1 cells and 5 μg/mL LPS were selected. In addition, TFF2 interference decreased the proliferation and promoted the apoptosis of PanC-1 cells and LPS-induced HPDE cells. However, TFF2 interference did not obviously change the levels of TNF-α, IL-1β and IL-6 in PanC-1 cells and LPS-induced HPDE cells. Furthermore, TFF2 interference suppressed the expression of β-catenin, c-Myc, Cyclin D1 and BIRC5 in PanC-1 cells and LPS-induced HPDE cells. TFF2 was demonstrated to combine with β-catenin. Discussion TFF2 interference inhibits proliferation and promotes apoptosis of PanC-1 cells and LPS-induced HPDE cells by suppressing β-catenin pathway.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| | - Yan Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| | - Jia Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| |
Collapse
|